1
|
Buthmann JL, Benmarhnia T, Huang JY, Huang P, Miller JG, Uy JP, Gluckman PD, Fortier MV, Chong YS, Tan AP, Meaney MJ, Gotlib IH. Exposure to Fine Particulate Matter During Pregnancy Is Associated With Hippocampal Development in Offspring. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100490. [PMID: 40290196 PMCID: PMC12032870 DOI: 10.1016/j.bpsgos.2025.100490] [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: 11/19/2024] [Revised: 02/14/2025] [Accepted: 03/12/2025] [Indexed: 04/30/2025] Open
Abstract
Background As the global climate crisis persists, it becomes increasingly important to understand how exposure to environmental toxins can affect the developing brain. Although researchers are beginning to document links between prenatal exposure to air pollution and brain structure, it is not clear when these associations emerge. Methods We leveraged data from the GUSTO (Growing Up Toward Healthy Outcomes in Singapore) longitudinal birth cohort study to examine prenatal exposure to air pollution and brain development during childhood. Spatiotemporally interpolated prenatal exposure to particulate matter <2.5 μm was averaged across each prenatal week. Structural magnetic resonance imaging data were obtained when children were ages 4.5, 6.0, 7.5, and 10.5 years (N = 325, 47.7% female) and segmented with FreeSurfer 7.1. A subset of parents completed the Child Behavior Checklist at the final assessment (n = 195, 46.7% female). We used latent growth modeling to estimate a slope of hippocampal volume growth in each hemisphere from ages 4.5 to 10.5 years, adjusted for intracranial volume. Results Distributed lag models indicated that late gestational exposure (during weeks 36-40) was associated with slower hippocampal growth in both hemispheres. Importantly, we also found that faster hippocampal volume growth in the right hemisphere was associated with more externalizing and attention problems at 10.5 years. Conclusions Future research should examine mechanisms that may underlie or contribute to these associations. These findings underscore the importance of efforts to reduce pollution, particularly for pregnant people and their children.
Collapse
Affiliation(s)
| | - Tarik Benmarhnia
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, California
- Irset Institut de Recherche en Santé, Environnement et Travail, Unité Mixte de Recherche, INSERM, University of Rennes, EHESP, Rennes, France
| | - Jonathan Y. Huang
- Thompson School of Social Work & Public Health, Office of Public Health Studies, University of Hawaii at Mānoa, Honolulu, Hawaii
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore
| | | | - Jonas G. Miller
- Department of Psychological Sciences, University of Connecticut, Storrs, Connecticut
| | - Jessica P. Uy
- Department of Psychology, Stanford University, Stanford, California
| | - Peter D. Gluckman
- SICS, A∗STAR, Singapore
- Centre for Human Evolution, Adaptation and Disease, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Marielle V. Fortier
- SICS, A∗STAR, Singapore
- Department of Diagnostic and Interventional Radiology, KK Women’s and Children’s Hospital, Singapore
| | - Yap-Seng Chong
- SICS, A∗STAR, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
| | - Ai Peng Tan
- SICS, A∗STAR, Singapore
- Department of Diagnostic Radiology, National University of Singapore, Singapore
- Department of Diagnostic Imaging, National University Hospital, Singapore
| | - Michael J. Meaney
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, Québec, Canada
| | - Ian H. Gotlib
- Department of Psychology, Stanford University, Stanford, California
| |
Collapse
|
2
|
Hoyniak CP, Donohue MR, Hennefield L, Whalen DJ. Preschool Mood Disorders: A Review of the Literature from 2017 to 2024. Child Adolesc Psychiatr Clin N Am 2025; 34:325-337. [PMID: 40044270 PMCID: PMC11885883 DOI: 10.1016/j.chc.2024.07.007] [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: 03/09/2025]
Abstract
Depression symptoms that onset during early childhood are increasingly recognized as a significant public health concern. A proliferation of research over the last 2 decades has identified preschool depression as a diagnostic reality that is associated with a severe and persistent course of depression throughout the lifespan. The current review summarizes the research on preschool depression from the last 7 years (2017-2024), with a particular focus on the assessment of, factors and outcomes associated with, neurobiological underpinnings of, and treatments for preschool depression. We also discuss potential avenues of inquiry that further elucidate the nature of preschool depression.
Collapse
Affiliation(s)
- Caroline P Hoyniak
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA.
| | - Meghan Rose Donohue
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Laura Hennefield
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Diana J Whalen
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
3
|
Shi L, Zhou X, Qu Y, Du Z, Zhou Q, Zhou Z, Jiang Y, Wang X, Zhu H. The impact of cortical and subcortical volumes on major depression risk: A genetic study. J Affect Disord 2025; 374:356-364. [PMID: 39824313 DOI: 10.1016/j.jad.2025.01.069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
OBJECTIVE This study aimed to explore the causal relationship between brain cortical and subcortical structures and major depressive disorder (MDD) using the Mendelian Randomization (MR) method. METHODS Single nucleotide polymorphisms (SNPs) were used as instrumental variables to analyze subcortical brain volume, cortical thickness, and surface area as exposure factors, with MDD as the outcome. Multiple sensitivity analyses were conducted to validate the robustness of the results. RESULTS MR analysis showed a significant negative correlation between subcortical brain volume and MDD. Specifically, the volumes of the caudate nucleus, hippocampus, intracranial region, and thalamus were inversely associated with the risk of MDD, indicating that larger subcortical brain volumes were linked to a lower risk of MDD. The thickness of the entorhinal cortex was also negatively correlated with MDD. At the same time, certain cortical regions showed significant positive correlations between surface area and MDD. Increased thickness of the entorhinal cortex and increased surface areas of the isthmus of the cingulate gyrus, middle temporal gyrus, and precuneus were associated with a higher risk of MDD. The sensitivity analyses revealed no significant heterogeneity or horizontal pleiotropy, and the results were consistent across different methods, confirming their robustness. CONCLUSION The study found that increased volumes of the caudate nucleus, hippocampus, internal capsule, and thalamus in subcortical brain regions and increased thickness of the entorhinal cortex in cortical regions were associated with a reduced risk of MDD, while increased surface areas of the isthmus of the cingulate gyrus, middle temporal gyrus, and precuneus in cortical regions were linked to an elevated risk of MDD.
Collapse
Affiliation(s)
- Lingyi Shi
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu 214151, China
| | - Xiangjun Zhou
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu 214151, China
| | - Yucai Qu
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu 214151, China
| | - Zhiqiang Du
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu 214151, China
| | - Qin Zhou
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu 214151, China
| | - Zhenhe Zhou
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu 214151, China
| | - Ying Jiang
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu 214151, China.
| | - Xinrong Wang
- Nanjing University of Chinese Medicine Wuxi Affiliated Hospital: Wuxi Hospital of Traditional Chinese Medicine, Wuxi, Jiangsu 214071, China.
| | - Haohao Zhu
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu 214151, China.
| |
Collapse
|
4
|
Hanson T, Spencer S, Harker SA, Barry F, Burton P, Beauchemin J, Mennenga SE, Braden BB, D'Sa V, Koinis-Mitchell D, Deoni SC, Lewis CR. Peripheral DNA Methylation of Cortisol- and Serotonin-Related Genes Predicts Hippocampal Volume in a Pediatric Population. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100421. [PMID: 39867566 PMCID: PMC11758844 DOI: 10.1016/j.bpsgos.2024.100421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 01/28/2025] Open
Abstract
Background Hippocampal volume increases throughout early development and is an important indicator of cognitive abilities and mental health. However, hippocampal development is highly vulnerable to exposures during development, as seen by smaller hippocampal volume and differential epigenetic programming in genes implicated in mental health. However, few studies have investigated hippocampal volume in relation to the peripheral epigenome across development, and even less is known about potential genetic moderators. Therefore, in this study, we explored relationships between hippocampal volume and peripheral DNA methylation of mental health-related genes, specifically NR3C1, FKBP5, and SLC6A4, throughout early development and whether these associations were moderated by age or genotype. Methods Bilateral hippocampal volume was computed from T2-weighted images through FreeSurfer, and DNA methylation was measured from saliva using the Illumina MethylationEPIC microarray in a pediatric population (N = 248, females = 112, meanage = 5.13 years, SDage = 3.60 years). Results Multiple linear regression and bootstrapping analyses revealed that DNA methylation of NR3C1, FKBP5, and SLC6A4 was associated with hippocampal volume and that these relationships were moderated by age and gene-specific variants. Conclusions These findings support the validity of peripheral DNA methylation profiles for indirectly assessing hippocampal volume and development and underscore the importance of genotype and age considerations in research. Therefore, peripheral epigenetic profiles may be a promising avenue for investigating the impacts of early-life stress on brain structure and subsequent mental health outcomes.
Collapse
Affiliation(s)
- Taena Hanson
- Department of Psychology, Arizona State University, Tempe, Arizona
| | - Sophia Spencer
- Department of Psychology, Arizona State University, Tempe, Arizona
| | | | - Fatoumata Barry
- Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Phoebe Burton
- Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | | | | | - B. Blair Braden
- College of Health Solutions, Arizona State University, Tempe, Arizona
| | - Viren D'Sa
- Maternal, Newborn, and Child Health Discovery & Tools, Bill & Melinda Gates Foundation; Seattle, Washington; Providence, Rhode Island
| | - Daphne Koinis-Mitchell
- Maternal, Newborn, and Child Health Discovery & Tools, Bill & Melinda Gates Foundation; Seattle, Washington; Providence, Rhode Island
| | - Sean C.L. Deoni
- Maternal, Newborn, and Child Health Discovery & Tools, Bill & Melinda Gates Foundation; Seattle, Washington; Providence, Rhode Island
- Advanced Baby Imaging Laboratory, Rhode Island Hospital, Providence, Rhode Island
| | - Candace R. Lewis
- Department of Psychology, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
- Neurogenomics, Translational Genomics Research Institute, Phoenix, Arizona
| |
Collapse
|
5
|
Khalil MH. Walking and Hippocampal Formation Volume Changes: A Systematic Review. Brain Sci 2025; 15:52. [PMID: 39851420 PMCID: PMC11763604 DOI: 10.3390/brainsci15010052] [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: 12/18/2024] [Revised: 01/01/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Sustaining the human brain's hippocampus from atrophy throughout ageing is critical. Exercise is proven to be effective in promoting adaptive hippocampal plasticity, and the hippocampus has a bidirectional relationship with the physical environment. Therefore, this systematic review explores the effects of walking, a simple physical activity in the environment, on hippocampal formation volume changes for lifelong brain and cognitive health. METHOD PubMed, Scopus, and Web of Science were searched for studies on humans published up to November 2022 examining hippocampal volume changes and walking. Twelve studies met the inclusion criteria. Study quality was assessed using the PEDro scale and ROBINS-I tool. A narrative synthesis explored walking factors associated with total, subregional, and hemisphere-specific hippocampal volume changes. RESULTS Overall, walking had positive effects on hippocampal volumes. Several studies found benefits of higher-intensity and greater amounts of walking for total hippocampal volume. The subiculum increased after low-intensity walking and nature exposure, while the parahippocampal gyrus benefited from vigorous intensity. The right hippocampus increased with spatial navigation during walking. No studies examined the effect of walking on the dentate gyrus. CONCLUSIONS This systematic review highlights walking as a multifaceted variable that can lead to manifold adaptive hippocampal volume changes. These findings support the promotion of walking as a simple, effective strategy to enhance brain health and prevent cognitive decline, suggesting the design of physical environments with natural and biophilic characteristics and layouts with greater walkability and cognitive stimulation. Future research is encouraged to explore the hippocampal subregional changes instead of focusing on total hippocampal volume, since the hippocampal formation is multicompartmental and subfields respond differently to different walking-related variables.
Collapse
|
6
|
Doretto VF, Salto ABR, Schivoletto S, Zugman A, Oliveira MC, Brañas M, Croci M, Ito LT, Santoro M, Jackowski AP, Bressan RA, Rohde LA, Salum G, Miguel EC, Pan PM. Childhood maltreatment and the structural development of hippocampus across childhood and adolescence. Psychol Med 2025; 54:1-9. [PMID: 39773537 PMCID: PMC11769901 DOI: 10.1017/s0033291724001636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 03/30/2024] [Accepted: 07/08/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Prior studies suggest that childhood maltreatment is associated with altered hippocampal volume. However, longitudinal studies are currently scarce, making it difficult to determine how alterations in hippocampal volume evolve over time. The current study examined the relationship between childhood maltreatment and hippocampal volumetric development across childhood and adolescence in a community sample. METHODS In this longitudinal study, a community sample of 795 participants underwent brain magnetic resonance imaging (MRI) in three waves spanning ages 6-21 years. Childhood maltreatment was assessed using parent-report and children´s self-report at baseline (6-12 years old). Mixed models were used to examine the relationship between childhood maltreatment and hippocampal volume across time. RESULTS The quadratic term of age was significantly associated with both right and left hippocampal volume development. High exposure to childhood maltreatment was associated with reduced offset of right hippocampal volume and persistent reduced volume throughout adolescence.Critically, the relationship between childhood maltreatment and reduced right hippocampal volume remained significant after adjusting for the presence of any depressive disorder during late childhood and adolescence and hippocampal volume polygenic risk scores. Time-by-CM and Sex-by-CM interactions were not statistically significant. CONCLUSIONS The present study showed that childhood maltreatment is associated with persistent reduction of hippocampal volume in children and adolescents, even after adjusting for the presence of major depressive disorder and genetic determinants of hippocampal structure.
Collapse
Affiliation(s)
- Victoria Fogaça Doretto
- Department of Psychiatry Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
| | - Ana Beatriz Ravagnani Salto
- Department of Psychiatry Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
| | - Sandra Schivoletto
- Department of Psychiatry Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Andre Zugman
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Department of Psychiatry, Laboratório Interdisciplinar Neurociências Clínicas (LiNC), Universidade Federal de São Paulo, São Paulo, Brazil
| | - Melaine Cristina Oliveira
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Department of Psychiatry, Laboratório Interdisciplinar Neurociências Clínicas (LiNC), Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marcelo Brañas
- Department of Psychiatry Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
| | - Marcos Croci
- Department of Psychiatry Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
| | - Lucas Toshio Ito
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Department of Psychiatry, Laboratório Interdisciplinar Neurociências Clínicas (LiNC), Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marcos Santoro
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Department of Psychiatry, Laboratório Interdisciplinar Neurociências Clínicas (LiNC), Universidade Federal de São Paulo, São Paulo, Brazil
| | - Andrea P. Jackowski
- Department of Psychiatry, Laboratório Interdisciplinar Neurociências Clínicas (LiNC), Universidade Federal de São Paulo, São Paulo, Brazil
- Department of Education, Information and Communications Technology (ICT) and Learning, Østfold University College, Halden, Norway
| | - Rodrigo A. Bressan
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Department of Psychiatry, Laboratório Interdisciplinar Neurociências Clínicas (LiNC), Universidade Federal de São Paulo, São Paulo, Brazil
| | - Luis Augusto Rohde
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Attention-Deficit/Hyperactivity Disorder and Developmental Psychiatry Programs, Hospital de Clínicas de Porto Alegre, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Giovanni Salum
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Department of Psychiatry, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Eurípedes Constantino Miguel
- Department of Psychiatry Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
| | - Pedro Mario Pan
- Department of Psychiatry Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo, Brazil
- Department of Psychiatry, Laboratório Interdisciplinar Neurociências Clínicas (LiNC), Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Kronman H, Singh A, Azam S, Guzman AS, Zelli D, Lau T, Dobbin J, Bigio B, Nasca C. Multidimensional Effects of Stress on Neuronal Exosome Levels and Simultaneous Transcriptomic Profiles. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100401. [PMID: 39720402 PMCID: PMC11667124 DOI: 10.1016/j.bpsgos.2024.100401] [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: 02/13/2024] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 12/26/2024] Open
Abstract
Background An excess of exosomes, nanovesicles released from all cells and key regulators of brain plasticity, is an emerging therapeutic target for stress-related mental illnesses. The effects of chronic stress on exosome levels are unknown; even less is known about molecular drivers of exosome levels in the stress response. Methods We used our state-of-the-art protocol with 2 complementary strategies to isolate neuronal exosomes from plasma, ventral dentate gyrus, basolateral amygdala, and olfactory bulbs of male mice to determine the effects of chronic restraint stress (CRS) on exosome levels. Next, we used RNA sequencing and bioinformatic analyses to identify molecular drivers of exosome levels. Results We found that CRS leads to an increase in the levels of neuronal exosomes but not total (i.e., not neuronally enriched) exosome levels assayed in plasma and the ventral dentate gyrus, whereas CRS leads to a decrease in neuronal exosome levels but not total exosome levels in the basolateral amygdala. There was a further specificity of effects as shown by a lack of changes in the levels of neuronal exosomes assayed in the olfactory bulbs. In pursuit of advancing translational applications, we showed that acetyl-L-carnitine administration restores the CRS-induced increase in neuronal exosome levels assayed in plasma (the most accessible specimen). Furthermore, the CRS-induced changes in neuronal exosome levels in the ventral dentate gyrus and basolateral amygdala mirrored the opposite pattern of CRS-induced transcriptional changes in these key brain areas, with β-estradiol signaling as a potential upstream driver of neuronal exosome levels. Conclusions This study provides a foundation for future studies of new forms of local and distant communication in stress neurobiology by demonstrating specific relationships between neuronal exosome levels assayed in plasma and the brain and providing new candidate targets for the normalization of exosome levels.
Collapse
Affiliation(s)
- Hope Kronman
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York
| | - Amarjyot Singh
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York
| | - Shofiul Azam
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York
| | - Andrea S. Guzman
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York
| | - Danielle Zelli
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, the Rockefeller University, New York, New York
| | - Timothy Lau
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, the Rockefeller University, New York, New York
| | - Josh Dobbin
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, the Rockefeller University, New York, New York
| | - Benedetta Bigio
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York
| | - Carla Nasca
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, the Rockefeller University, New York, New York
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, New York
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York
| |
Collapse
|
8
|
Fowler CH, Reuben A, Stapleton HM, Hoffman K, Herkert N, Barakat L, Gaffrey MS. Children's exposure to chemical contaminants: Demographic disparities and associations with the developing basal ganglia. ENVIRONMENTAL RESEARCH 2024; 263:119990. [PMID: 39304016 DOI: 10.1016/j.envres.2024.119990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 08/17/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Children are regularly exposed to chemical contaminants that may influence brain development. However, relatively little is known about how these contaminants impact the developing human brain. Here, we combined silicone wristband exposure assessments with neuroimaging for the first time to examine how chemical contaminant mixtures are associated with the developing basal ganglia-a brain region key for the healthy development of emotion, reward, and motor processing, and which may be particularly susceptible to contaminant harm. Further, we examined demographic disparities in exposures to clarify which children were at highest risk for any contaminant-associated neurobiological changes. Participants included 62 community children (average age 7.00 years, 53% female, 66% White) who underwent structural neuroimaging to provide data on their basal ganglia structure and wore a silicone wristband for seven days to track their chemical contaminant exposure. 45 chemical contaminants-including phthalates and their alternatives, brominated flame retardants, organophosphate esters, pesticides, polycyclic aromatic hydrocarbons, and polychlorinated biphenyls-were detected in over 75% of wristbands. Notable demographic disparities in exposure were present, such that Non-White and lower-income children were more exposed to several contaminants. Exposure to chemical contaminant mixtures was not associated with overall basal ganglia volume; however, two organophosphate esters (2IPPDPP and 4IPPDPP) were both associated with a larger globus pallidus, a basal ganglia sub-region. Results highlight demographic disparities in exposure and suggest possible risks to a brain region key for healthy emotional development.
Collapse
Affiliation(s)
| | | | | | | | | | - Lubna Barakat
- University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA
| | - Michael S Gaffrey
- Duke University, Durham, NC, 27708, USA; Children's Wisconsin, Milwaukee, WI, 53226, USA
| |
Collapse
|
9
|
Bertollo AG, Mingoti MED, de Medeiros J, da Silva GB, Capoani GT, Lindemann H, Cassol J, Manica D, de Oliveira T, Garcez ML, Bagatini MD, Bohnen LC, Junior WAR, Ignácio ZM. Hydroalcoholic Extract of Centella asiatica and Madecassic Acid Reverse Depressive-Like Behaviors, Inflammation and Oxidative Stress in Adult Rats Submitted to Stress in Early Life. Mol Neurobiol 2024; 61:10182-10197. [PMID: 38703344 DOI: 10.1007/s12035-024-04198-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 04/22/2024] [Indexed: 05/06/2024]
Abstract
Major depressive disorder (MDD) is a severe disorder that causes enormous loss of quality of life, and among the factors underlying MDD is stress in maternal deprivation (MD). In addition, classic pharmacotherapy has presented severe adverse effects. Centella asiatica (C. asiatica) demonstrates a potential neuroprotective effect but has not yet been evaluated in MD models. This study aimed to evaluate the effect of C. asiatica extract and the active compound madecassic acid on possible depressive-like behavior, inflammation, and oxidative stress in the hippocampus and serum of young rats submitted to MD in the first days of life. Rats (after the first day of birth) were separated from the mother for 3 h a day for 10 days. When adults, these animals were divided into groups and submitted to treatment for 14 days. After subjecting the animals to protocols of locomotor activity in the open field and behavioral despair in the forced swimming test, researchers then euthanized the animals. The hippocampus and serum were collected and analyzed for the inflammatory cytokines and oxidative markers. The C. asiatica extract and active compound reversed or reduced depressive-like behaviors, inflammation in the hippocampus, and oxidative stress in serum and hippocampus. These results suggest that C. asiatica and madecassic acid have potential antidepressant action, at least partially, through anti-inflammatory and antioxidant profiles.
Collapse
Affiliation(s)
- Amanda Gollo Bertollo
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Maiqueli Eduarda Dama Mingoti
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Jesiel de Medeiros
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Gilnei Bruno da Silva
- Multicentric Postgraduate Program in Biochemistry and Molecular Biology, State University of Santa Catarina, Lages, SC, Brazil
| | - Giovana Tamara Capoani
- Laboratory of Pharmacognosy, Community University of Chapecó Region, Unochapecó, Chapecó, SC, Brazil
| | - Heloisa Lindemann
- Laboratory of Pharmacognosy, Community University of Chapecó Region, Unochapecó, Chapecó, SC, Brazil
| | - Joana Cassol
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Daiane Manica
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Tacio de Oliveira
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Michelle Lima Garcez
- Department of Clinical Analysis, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Margarete Dulce Bagatini
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil
| | - Lilian Caroline Bohnen
- Postgraduate Program in Health Sciences, Community University of Chapecó Region, Unochapecó, Chapecó, SC, Brazil
| | - Walter Antônio Roman Junior
- Postgraduate Program in Health Sciences, Community University of Chapecó Region, Unochapecó, Chapecó, SC, Brazil
| | - Zuleide Maria Ignácio
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, 89815-899, Brazil.
| |
Collapse
|
10
|
Calabro FJ, Parr AC, Sydnor VJ, Hetherington H, Prasad KM, Ibrahim TS, Sarpal DK, Famalette A, Verma P, Luna B. Leveraging ultra-high field (7T) MRI in psychiatric research. Neuropsychopharmacology 2024; 50:85-102. [PMID: 39251774 PMCID: PMC11525672 DOI: 10.1038/s41386-024-01980-6] [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: 04/01/2024] [Revised: 06/21/2024] [Accepted: 07/23/2024] [Indexed: 09/11/2024]
Abstract
Non-invasive brain imaging has played a critical role in establishing our understanding of the neural properties that contribute to the emergence of psychiatric disorders. However, characterizing core neurobiological mechanisms of psychiatric symptomatology requires greater structural, functional, and neurochemical specificity than is typically obtainable with standard field strength MRI acquisitions (e.g., 3T). Ultra-high field (UHF) imaging at 7 Tesla (7T) provides the opportunity to identify neurobiological systems that confer risk, determine etiology, and characterize disease progression and treatment outcomes of major mental illnesses. Increases in scanner availability, regulatory approval, and sequence availability have made the application of UHF to clinical cohorts more feasible than ever before, yet the application of UHF approaches to the study of mental health remains nascent. In this technical review, we describe core neuroimaging methodologies which benefit from UHF acquisition, including high resolution structural and functional imaging, single (1H) and multi-nuclear (e.g., 31P) MR spectroscopy, and quantitative MR techniques for assessing brain tissue iron and myelin. We discuss advantages provided by 7T MRI, including higher signal- and contrast-to-noise ratio, enhanced spatial resolution, increased test-retest reliability, and molecular and neurochemical specificity, and how these have begun to uncover mechanisms of psychiatric disorders. Finally, we consider current limitations of UHF in its application to clinical cohorts, and point to ongoing work that aims to overcome technical hurdles through the continued development of UHF hardware, software, and protocols.
Collapse
Affiliation(s)
- Finnegan J Calabro
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Ashley C Parr
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Valerie J Sydnor
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Konasale M Prasad
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Tamer S Ibrahim
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Deepak K Sarpal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alyssa Famalette
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Piya Verma
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Beatriz Luna
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
11
|
Martinez M, Cai T, Yang B, Zhou Z, Shankman SA, Mittal VA, Haase CM, Qu Y. Depressive symptoms during the transition to adolescence: Left hippocampal volume as a marker of social context sensitivity. Proc Natl Acad Sci U S A 2024; 121:e2321965121. [PMID: 39226358 PMCID: PMC11406239 DOI: 10.1073/pnas.2321965121] [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: 12/19/2023] [Accepted: 06/17/2024] [Indexed: 09/05/2024] Open
Abstract
The transition to adolescence is a critical period for mental health development. Socio-experiential environments play an important role in the emergence of depressive symptoms with some adolescents showing more sensitivity to social contexts than others. Drawing on recent developmental neuroscience advances, we examined whether hippocampal volume amplifies social context effects in the transition to adolescence. We analyzed 2-y longitudinal data from the Adolescent Brain Cognitive Development (ABCD®) study in a diverse sample of 11,832 youth (mean age: 9.914 y; range: 8.917 to 11.083 y; 47.8% girls) from 21 sites across the United States. Socio-experiential environments (i.e., family conflict, primary caregiver's depressive symptoms, parental warmth, peer victimization, and prosocial school environment), hippocampal volume, and a wide range of demographic characteristics were measured at baseline. Youth's symptoms of major depressive disorder were assessed at both baseline and 2 y later. Multilevel mixed-effects linear regression analyses showed that negative social environments (i.e., family conflict, primary caregiver's depressive symptoms, and peer victimization) and the absence of positive social environments (i.e., parental warmth and prosocial school environment) predicted greater increases in youth's depressive symptoms over 2 y. Importantly, left hippocampal volume amplified social context effects such that youth with larger left hippocampal volume experienced greater increases in depressive symptoms in more negative and less positive social environments. Consistent with brain-environment interaction models of mental health, these findings underscore the importance of families, peers, and schools in the development of depression during the transition to adolescence and show how neural structure amplifies social context sensitivity.
Collapse
Affiliation(s)
- Matias Martinez
- School of Education and Social Policy, Northwestern University, Evanston, IL60208
- Institute for Innovations in Developmental Sciences, Northwestern University, Chicago, IL60611
- Institute for Policy Research, Northwestern University, Evanston, IL60208
| | - Tianying Cai
- School of Education and Social Policy, Northwestern University, Evanston, IL60208
- Institute of Child Development, University of Minnesota, Twin Cities, Minneapolis, MN55455
| | - Beiming Yang
- School of Education and Social Policy, Northwestern University, Evanston, IL60208
| | - Zexi Zhou
- Department of Human Development and Family Sciences, University of Texas, Austin, TX78712
| | - Stewart A. Shankman
- Institute for Innovations in Developmental Sciences, Northwestern University, Chicago, IL60611
- Department of Psychology, Northwestern University, Evanston, IL60208
- Department of Psychiatry, Northwestern University, Chicago, IL60611
| | - Vijay A. Mittal
- Institute for Innovations in Developmental Sciences, Northwestern University, Chicago, IL60611
- Institute for Policy Research, Northwestern University, Evanston, IL60208
- Department of Psychology, Northwestern University, Evanston, IL60208
- Department of Psychiatry, Northwestern University, Chicago, IL60611
| | - Claudia M. Haase
- School of Education and Social Policy, Northwestern University, Evanston, IL60208
- Institute for Innovations in Developmental Sciences, Northwestern University, Chicago, IL60611
- Institute for Policy Research, Northwestern University, Evanston, IL60208
- Department of Psychology, Northwestern University, Evanston, IL60208
- Department of Psychiatry, Northwestern University, Chicago, IL60611
- Interdepartmental Neuroscience, Northwestern University, Evanston, IL60611
- Buffett Institute for Global Studies, Northwestern University, Evanston, IL60201
| | - Yang Qu
- School of Education and Social Policy, Northwestern University, Evanston, IL60208
- Institute for Innovations in Developmental Sciences, Northwestern University, Chicago, IL60611
- Institute for Policy Research, Northwestern University, Evanston, IL60208
- Department of Psychology, Northwestern University, Evanston, IL60208
| |
Collapse
|
12
|
Maggioni E, Pigoni A, Fontana E, Delvecchio G, Bonivento C, Bianchi V, Mauri M, Bellina M, Girometti R, Agarwal N, Nobile M, Brambilla P. Right frontal cingulate cortex mediates the effect of prenatal complications on youth internalizing behaviors. Mol Psychiatry 2024; 29:2074-2083. [PMID: 38378927 PMCID: PMC11408263 DOI: 10.1038/s41380-024-02475-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/22/2024]
Abstract
Prenatal and perinatal complications represent well-known risk factors for the future development of psychiatric disorders. Such influence might become manifested during childhood and adolescence, as key periods for brain and behavioral changes. Internalizing and externalizing behaviors in adolescence have been associated with the risk of psychiatric onset later in life. Both brain morphology and behavior seem to be affected by obstetric complications, but a clear link among these three aspects is missing. Here, we aimed at analyzing the association between prenatal and perinatal complications, behavioral issues, and brain volumes in a group of children and adolescents. Eighty-two children and adolescents with emotional-behavioral problems underwent clinical and 3 T brain magnetic resonance imaging (MRI) assessments. The former included information on behavior, through the Child Behavior Checklist/6-18 (CBCL/6-18), and on the occurrence of obstetric complications. The relationships between clinical and gray matter volume (GMV) measures were investigated through multiple generalized linear models and mediation models. We found a mutual link between prenatal complications, GMV alterations in the frontal gyrus, and withdrawn problems. Specifically, complications during pregnancy were associated with higher CBCL/6-18 withdrawn scores and GMV reductions in the right superior frontal gyrus and anterior cingulate cortex. Finally, a mediation effect of these GMV measures on the association between prenatal complications and the withdrawn dimension was identified. Our findings suggest a key role of obstetric complications in affecting brain structure and behavior. For the first time, a mediator role of frontal GMV in the relationship between prenatal complications and internalizing symptoms was suggested. Once replicated on independent cohorts, this evidence will have relevant implications for planning preventive interventions.
Collapse
Affiliation(s)
- Eleonora Maggioni
- Department of Electronics Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Alessandro Pigoni
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Social and Affective Neuroscience Group, MoMiLab, IMT School for Advanced Studies Lucca, Lucca, Italy
| | - Elisa Fontana
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Giuseppe Delvecchio
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Valentina Bianchi
- Child and Adolescent Psychiatry Unit, Scientific Institute IRCCS "Eugenio Medea", Bosisio Parini (Lc), Italy
| | - Maddalena Mauri
- Child and Adolescent Psychiatry Unit, Scientific Institute IRCCS "Eugenio Medea", Bosisio Parini (Lc), Italy
| | - Monica Bellina
- Child and Adolescent Psychiatry Unit, Scientific Institute IRCCS "Eugenio Medea", Bosisio Parini (Lc), Italy
| | - Rossano Girometti
- Institute of Radiology, Department of Medicine (DMED), University of Udine, Udine, Italy
- University Hospital S. Maria Della Misericordia, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Nivedita Agarwal
- Neuroimaging Unit, Scientific Institute IRCCS "Eugenio Medea", Bosisio Parini (Lc), Italy
| | - Maria Nobile
- Child and Adolescent Psychiatry Unit, Scientific Institute IRCCS "Eugenio Medea", Bosisio Parini (Lc), Italy
| | - Paolo Brambilla
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.
| |
Collapse
|
13
|
Yang J, Guo H, Cai A, Zheng J, Liu J, Xiao Y, Ren S, Sun D, Duan J, Zhao T, Tang J, Zhang X, Zhu R, Wang J, Wang F. Aberrant Hippocampal Development in Early-onset Mental Disorders and Promising Interventions: Evidence from a Translational Study. Neurosci Bull 2024; 40:683-694. [PMID: 38141109 PMCID: PMC11178726 DOI: 10.1007/s12264-023-01162-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/01/2023] [Indexed: 12/24/2023] Open
Abstract
Early-onset mental disorders are associated with disrupted neurodevelopmental processes during adolescence. The methylazoxymethanol acetate (MAM) animal model, in which disruption in neurodevelopmental processes is induced, mimics the abnormal neurodevelopment associated with early-onset mental disorders from an etiological perspective. We conducted longitudinal structural magnetic resonance imaging (MRI) scans during childhood, adolescence, and adulthood in MAM rats to identify specific brain regions and critical windows for intervention. Then, the effect of repetitive transcranial magnetic stimulation (rTMS) intervention on the target brain region during the critical window was investigated. In addition, the efficacy of this intervention paradigm was tested in a group of adolescent patients with early-onset mental disorders (diagnosed with major depressive disorder or bipolar disorder) to evaluate its clinical translational potential. The results demonstrated that, compared to the control group, the MAM rats exhibited significantly lower striatal volume from childhood to adulthood (all P <0.001). In contrast, the volume of the hippocampus did not show significant differences during childhood (P >0.05) but was significantly lower than the control group from adolescence to adulthood (both P <0.001). Subsequently, rTMS was applied to the occipital cortex, which is anatomically connected to the hippocampus, in the MAM models during adolescence. The MAM-rTMS group showed a significant increase in hippocampal volume compared to the MAM-sham group (P <0.01), while the volume of the striatum remained unchanged (P >0.05). In the clinical trial, adolescents with early-onset mental disorders showed a significant increase in hippocampal volume after rTMS treatment compared to baseline (P <0.01), and these volumetric changes were associated with improvement in depressive symptoms (r = - 0.524, P = 0.018). These findings highlight the potential of targeting aberrant hippocampal development during adolescence as a viable intervention for early-onset mental disorders with neurodevelopmental etiology as well as the promise of rTMS as a therapeutic approach for mitigating aberrant neurodevelopmental processes and alleviating clinical symptoms.
Collapse
Affiliation(s)
- Jingyu Yang
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Huiling Guo
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
- School of Biomedical Engineering and Informatics, Nanjing, Medical University, Nanjing, 211166, China
| | - Aoling Cai
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
- School of Biomedical Engineering and Informatics, Nanjing, Medical University, Nanjing, 211166, China
- The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213004, China
| | - Junjie Zheng
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Juan Liu
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Yao Xiao
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Sihua Ren
- Department of Radiology, First Hospital of China Medical University, Shenyang, 110002, China
| | - Dandan Sun
- Department of Cardiac Function, The People's Hospital of China Medical University and the People's Hospital of Liaoning Province, Shenyang, 110067, China
| | - Jia Duan
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Tongtong Zhao
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Jingwei Tang
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Xizhe Zhang
- School of Biomedical Engineering and Informatics, Nanjing, Medical University, Nanjing, 211166, China
| | - Rongxin Zhu
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China.
| | - Jie Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, 430064, China.
- Institute of Neuroscience and Brain Diseases; Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021, China.
| | - Fei Wang
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China.
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
14
|
Li S, Yang D, Zhou X, Chen L, Liu L, Lin R, Li X, Liu Y, Qiu H, Cao H, Liu J, Cheng Q. Neurological and metabolic related pathophysiologies and treatment of comorbid diabetes with depression. CNS Neurosci Ther 2024; 30:e14497. [PMID: 37927197 PMCID: PMC11017426 DOI: 10.1111/cns.14497] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/26/2023] [Accepted: 10/03/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND The comorbidity between diabetes mellitus and depression was revealed, and diabetes mellitus increased the prevalence of depressive disorder, which ranked 13th in the leading causes of disability-adjusted life-years. Insulin resistance, which is common in diabetes mellitus, has increased the risk of depressive symptoms in both humans and animals. However, the mechanisms behind the comorbidity are multi-factorial and complicated. There is still no causal chain to explain the comorbidity exactly. Moreover, Selective serotonin reuptake inhibitors, insulin and metformin, which are recommended for treating diabetes mellitus-induced depression, were found to be a risk factor in some complications of diabetes. AIMS Given these problems, many researchers made remarkable efforts to analyze diabetes complicating depression from different aspects, including insulin resistance, stress and Hypothalamic-Pituitary-Adrenal axis, neurological system, oxidative stress, and inflammation. Drug therapy, such as Hydrogen Sulfide, Cannabidiol, Ascorbic Acid and Hesperidin, are conducive to alleviating diabetes mellitus and depression. Here, we reviewed the exact pathophysiology underlying the comorbidity between depressive disorder and diabetes mellitus and drug therapy. METHODS The review refers to the available literature in PubMed and Web of Science, searching critical terms related to diabetes mellitus, depression and drug therapy. RESULTS In this review, we found that brain structure and function, neurogenesis, brain-derived neurotrophic factor and glucose and lipid metabolism were involved in the pathophysiology of the comorbidity. Obesity might lead to diabetes mellitus and depression through reduced adiponectin and increased leptin and resistin. In addition, drug therapy displayed in this review could expand the region of potential therapy. CONCLUSIONS The review summarizes the mechanisms underlying the comorbidity. It also overviews drug therapy with anti-diabetic and anti-depressant effects.
Collapse
Affiliation(s)
- Sixin Li
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Dong Yang
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Xuhui Zhou
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Lu Chen
- Department of Gastroenterology, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of GastroenterologyBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Lini Liu
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Ruoheng Lin
- Department of Psychiatry, National Clinical Research Center for Mental DisordersThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Xinyu Li
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Ying Liu
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Huiwen Qiu
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Hui Cao
- Department of Psychiatry, The School of Clinical MedicineHunan University of Chinese MedicineChangshaHunanChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People's Hospital of Hunan Province)ChangshaHunanChina
| | - Jian Liu
- Center for Medical Research and Innovation, The First Hospital, Hunan University of Chinese MedicineChangshaHunanChina
| | - Quan Cheng
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| |
Collapse
|
15
|
Singh MK, Gorelik AJ, Stave C, Gotlib IH. Genetics, epigenetics, and neurobiology of childhood-onset depression: an umbrella review. Mol Psychiatry 2024; 29:553-565. [PMID: 38102485 DOI: 10.1038/s41380-023-02347-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 12/17/2023]
Abstract
Depression is a serious and persistent psychiatric disorder that commonly first manifests during childhood. Depression that starts in childhood is increasing in frequency, likely due both to evolutionary trends and to increased recognition of the disorder. In this umbrella review, we systematically searched the extant literature for genetic, epigenetic, and neurobiological factors that contribute to a childhood onset of depression. We searched PubMed, EMBASE, OVID/PsychInfo, and Google Scholar with the following inclusion criteria: (1) systematic review or meta-analysis from a peer-reviewed journal; (2) inclusion of a measure assessing early age of onset of depression; and (3) assessment of neurobiological, genetic, environmental, and epigenetic predictors of early onset depression. Findings from 89 systematic reviews of moderate to high quality suggest that childhood-onset depressive disorders have neurobiological, genetic, environmental, and epigenetic roots consistent with a diathesis-stress theory of depression. This review identified key putative markers that may be targeted for personalized clinical decision-making and provide important insights concerning candidate mechanisms that might underpin the early onset of depression.
Collapse
|
16
|
Sheng W, Cui Q, Guo Y, Tang Q, Fan YS, Wang C, Guo J, Lu F, He Z, Chen H. Cortical thickness reductions associate with brain network architecture in major depressive disorder. J Affect Disord 2024; 347:175-182. [PMID: 38000466 DOI: 10.1016/j.jad.2023.11.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 10/25/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023]
Abstract
BACKGROUND Cortical thickness reductions in major depressive disorder are distributed across multiple regions. Research has indicated that cortical atrophy is influenced by connectome architecture on a range of neurological and psychiatric diseases. However, whether connectome architecture contributes to changes in cortical thickness in the same manner as it does in depression is unclear. This study aims to explain the distribution of cortical thickness reductions across the cortex in depression by brain connectome architecture. METHODS Here, we calculated a differential map of cortical thickness between 110 depression patients and 88 age-, gender-, and education level-matched healthy controls by using T1-weighted images and a structural network reconstructed through the diffusion tensor imaging of control group. We then used a neighborhood deformation model to explore how cortical thickness change in an area is influenced by areas structurally connected to it. RESULTS We found that cortical thickness in the frontoparietal and default networks decreased in depression, regional cortical thickness changes were related to reductions in their neighbors and were mainly limited by the frontoparietal and default networks, and the epicenter was in the prefrontal lobe. CONCLUSION Current findings suggest that connectome architecture contributes to the irregular topographic distribution of cortical thickness reductions in depression and cortical atrophy is restricted by and dependent on structural foundation.
Collapse
Affiliation(s)
- Wei Sheng
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Qian Cui
- School of Public Affairs and Administration, University of Electronic Science and Technology of China, Chengdu, China.
| | - YuanHong Guo
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Qin Tang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Yun-Shuang Fan
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Chong Wang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Jing Guo
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Fengmei Lu
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Zongling He
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Huafu Chen
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China; MOE Key Lab for Neuroinformation, HighField Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
17
|
Khin YP, Yamaoka Y, Abe A, Fujiwara T. Association of child-specific and household material deprivation with depression among elementary and middle school students in Japan. Soc Psychiatry Psychiatr Epidemiol 2024; 59:329-339. [PMID: 37270468 PMCID: PMC10239275 DOI: 10.1007/s00127-023-02502-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/24/2023] [Indexed: 06/05/2023]
Abstract
PURPOSE This study aimed to investigate the association between child-specific and household material deprivation with depression among elementary and middle school students in Japan. METHODS We used cross-sectional data from 10,505 and 10,008 students for fifth-grade elementary school students (G5) and second-grade middle school students (G8), respectively, and their caregivers. The data were collected from August to September 2016 in 4 municipalities of Tokyo and from July to November 2017 in 23 municipalities of Hiroshima prefecture. Caregivers completed questionnaires including household income and material deprivation, and children completed child-specific material deprivation and depression status using the Japanese version of the Birleson depression self-rating scale for children (DSRS-C). To explore the associations, logistic regression was used after conducting multiple imputation for the missing data. RESULTS 14.2% of G5 students and 23.6% of G8 students had DSRS-C scores of more than or equal to 16, denoting the risk of depression. We found that household equivalent income was not associated with childhood depression in both G5 and G8 students when adjusted for material deprivations. While at least one item of household material deprivation was significantly associated with depression in G8 students (OR = 1.19, CI = 1.00, 1.41), but not in G5 children. Child-specific material deprivation of more than 5 items was significantly associated with depression in both age groups (G5: OR = 1.53, CI = 1.25, 1.88; G8: OR = 1.45, CI = 1.22, 1.73). CONCLUSION Future research on child mental health needs to consider children's perspectives, especially material deprivation in young children.
Collapse
Affiliation(s)
- Yu Par Khin
- Department of Global Health Promotion, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Yui Yamaoka
- Department of Global Health Promotion, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Aya Abe
- School of Humanities and Social Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Takeo Fujiwara
- Department of Global Health Promotion, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.
| |
Collapse
|
18
|
Filetti C, Kane-Grade F, Gunnar M. The Development of Stress Reactivity and Regulation in Children and Adolescents. Curr Neuropharmacol 2024; 22:395-419. [PMID: 37559538 PMCID: PMC10845082 DOI: 10.2174/1570159x21666230808120504] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/03/2023] [Accepted: 02/10/2023] [Indexed: 08/11/2023] Open
Abstract
Adversity experienced in early life can have detrimental effects on physical and mental health. One pathway in which these effects occur is through the hypothalamic-pituitary-adrenal (HPA) axis, a key physiological stress-mediating system. In this review, we discuss the theoretical perspectives that guide stress reactivity and regulation research, the anatomy and physiology of the axis, developmental changes in the axis and its regulation, brain systems regulating stress, the role of genetic and epigenetics variation in axis development, sensitive periods in stress system calibration, the social regulation of stress (i.e., social buffering), and emerging research areas in the study of stress physiology and development. Understanding the development of stress reactivity and regulation is crucial for uncovering how early adverse experiences influence mental and physical health.
Collapse
Affiliation(s)
- Clarissa Filetti
- Institute of Child Development, University of Minnesota, Minneapolis, USA
| | - Finola Kane-Grade
- Institute of Child Development, University of Minnesota, Minneapolis, USA
| | - Megan Gunnar
- Institute of Child Development, University of Minnesota, Minneapolis, USA
| |
Collapse
|
19
|
Wen Y, Li H, Huang Y, Qiao D, Ren T, Lei L, Li G, Yang C, Xu Y, Han M, Liu Z. Dynamic network characteristics of adolescents with major depressive disorder: Attention network mediates the association between anhedonia and attentional deficit. Hum Brain Mapp 2023; 44:5749-5769. [PMID: 37683097 PMCID: PMC10619388 DOI: 10.1002/hbm.26474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 08/10/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
Attention deficit is a critical symptom that impairs social functioning in adolescents with major depressive disorder (MDD). In this study, we aimed to explore the dynamic neural network activity associated with attention deficits and its relationship with clinical outcomes in adolescents with MDD. We included 188 adolescents with MDD and 94 healthy controls. By combining psychophysics, resting-state electroencephalography (EEG), and functional magnetic resonance imaging (fMRI) techniques, we aimed to identify dynamic network features through the investigation of EEG microstate characteristics and related temporal network features in adolescents with MDD. At baseline, microstate analysis revealed that the occurrence of Microstate C in the patient group was lower than that in healthy controls, whereas the duration and coverage of Microstate D increased in the MDD group. Mediation analysis revealed that the probability of transition from Microstate C to D mediated anhedonia and attention deficits in the MDD group. fMRI results showed that the temporal variability of the dorsal attention network (DAN) was significantly weaker in patients with MDD than in healthy controls. Importantly, the temporal variability of DAN mediated the relationship between anhedonia and attention deficits in the patient group. After acute-stage treatment, the response prediction group (RP) showed improvement in Microstates C and D compared to the nonresponse prediction group (NRP). For resting-state fMRI data, the temporal variability of DAN was significantly higher in the RP group than in the NRP group. Overall, this study enriches our understanding of the neural mechanisms underlying attention deficits in patients with MDD and provides novel clinical biomarkers.
Collapse
Affiliation(s)
- Yujiao Wen
- Department of PsychiatryThe First Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Hong Li
- Department of PsychiatryThe First Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Yangxi Huang
- Department of PsychiatryThe First Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Dan Qiao
- Department of PsychiatryThe First Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Tian Ren
- Department of PsychiatryThe First Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Lei Lei
- Department of PsychiatryThe First Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Gaizhi Li
- Department of PsychiatryThe First Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Chunxia Yang
- Department of PsychiatryThe First Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Yifan Xu
- Department of PsychiatryThe First Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Min Han
- Department of PsychiatryThe First Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Zhifen Liu
- Department of PsychiatryThe First Hospital of Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
20
|
Zeng Z, Dong Y, Zou L, Xu D, Luo X, Chu T, Wang J, Ren Q, Liu Q, Li X. GluCEST Imaging and Structural Alterations of the Bilateral Hippocampus in First-Episode and Early-Onset Major Depression Disorder. J Magn Reson Imaging 2023; 58:1431-1440. [PMID: 36808678 DOI: 10.1002/jmri.28651] [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: 12/02/2022] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Glutamate dysregulation is one of the key pathogenic mechanisms of major depressive disorder (MDD), and glutamate chemical exchange saturation transfer (GluCEST) has been used for glutamate measurement in some brain diseases but rarely in depression. PURPOSE To investigate the GluCEST changes in hippocampus in MDD and the relationship between glutamate and hippocampal subregional volumes. STUDY TYPE Cross-sectional. SUBJECTS Thirty-two MDD patients (34% males; 22.03 ± 7.21 years) and 47 healthy controls (HCs) (43% males; 22.00 ± 3.28 years). FIELD STRENGTH/SEQUENCE 3.0 T; magnetization prepared rapid gradient echo (MPRAGE) for three-dimensional T1-weighted images, two-dimensional turbo spin echo GluCEST, and multivoxel chemical shift imaging (CSI) for proton magnetic resonance spectroscopy (1 H MRS). ASSESSMENT GluCEST data were quantified by magnetization transfer ratio asymmetry (MTRasym ) analysis and assessed by the relative concentration of 1 H MRS-measured glutamate. FreeSurfer was used for hippocampus segmentation. STATISTICAL TESTS The independent sample t test, Mann-Whitney U test, Spearman's correlation, and partial correlation analysis were used. P < 0.05 was considered statistically significant. RESULTS In the left hippocampus, GluCEST values were significantly decreased in MDD (2.00 ± 1.08 [MDD] vs. 2.62 ± 1.41 [HCs]) and showed a significantly positive correlation with Glx/Cr (r = 0.37). GluCEST values were significantly positively correlated with the volumes of CA1 (r = 0.40), subiculum (r = 0.40) in the left hippocampus and CA1 (r = 0.51), molecular_layer_HP (r = 0.50), GC-ML-DG (r = 0.42), CA3 (r = 0.44), CA4 (r = 0.44), hippocampus-amygdala-transition-area (r = 0.46), and the whole hippocampus (r = 0.47) in the right hippocampus. Hamilton Depression Rating Scale scores showed significantly negative correlations with the volumes of the left presubiculum (r = -0.40), left parasubiculum (r = -0.47), and right presubiculum (r = -0.41). DATA CONCLUSION GluCEST can be used to measure glutamate changes and help to understand the mechanism of hippocampal volume loss in MDD. Hippocampal volume changes are associated with disease severity. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Zhen Zeng
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Yingying Dong
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, China
| | - Linxuan Zou
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Donghao Xu
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Xunrong Luo
- Department of Radiology, Cancer Hospital of Chongqing University, Chongqing, China
| | - Tongpeng Chu
- Department of Radiology, Yantai Yuhuangding Hospital, Affiliated Hospital of Qingdao University, Yantai, China
| | - Jing Wang
- Department of Radiology, Binzhou Medical University Hospital, Binzhou, China
| | - Qingfa Ren
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Quanyuan Liu
- Department of Radiology, Binzhou Medical University Hospital, Binzhou, China
| | - Xianglin Li
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| |
Collapse
|
21
|
Mázala-de-Oliveira T, Silva BT, Campello-Costa P, Carvalho VF. The Role of the Adrenal-Gut-Brain Axis on Comorbid Depressive Disorder Development in Diabetes. Biomolecules 2023; 13:1504. [PMID: 37892186 PMCID: PMC10604999 DOI: 10.3390/biom13101504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/15/2023] [Accepted: 08/26/2023] [Indexed: 10/29/2023] Open
Abstract
Diabetic patients are more affected by depression than non-diabetics, and this is related to greater treatment resistance and associated with poorer outcomes. This increase in the prevalence of depression in diabetics is also related to hyperglycemia and hypercortisolism. In diabetics, the hyperactivity of the HPA axis occurs in parallel to gut dysbiosis, weakness of the intestinal permeability barrier, and high bacterial-product translocation into the bloodstream. Diabetes also induces an increase in the permeability of the blood-brain barrier (BBB) and Toll-like receptor 4 (TLR4) expression in the hippocampus. Furthermore, lipopolysaccharide (LPS)-induced depression behaviors and neuroinflammation are exacerbated in diabetic mice. In this context, we propose here that hypercortisolism, in association with gut dysbiosis, leads to an exacerbation of hippocampal neuroinflammation, glutamatergic transmission, and neuronal apoptosis, leading to the development and aggravation of depression and to resistance to treatment of this mood disorder in diabetic patients.
Collapse
Affiliation(s)
- Thalita Mázala-de-Oliveira
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (T.M.-d.-O.); (B.T.S.)
| | - Bruna Teixeira Silva
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (T.M.-d.-O.); (B.T.S.)
- Programa de Pós-Graduação em Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói 24210-201, Brazil;
| | - Paula Campello-Costa
- Programa de Pós-Graduação em Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói 24210-201, Brazil;
| | - Vinicius Frias Carvalho
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (T.M.-d.-O.); (B.T.S.)
- Programa de Pós-Graduação em Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói 24210-201, Brazil;
- Laboratório de Inflamação, Instituto Nacional de Ciência e Tecnologia em Neuroimunomodulação—INCT-NIM, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
| |
Collapse
|
22
|
Wang K, Li X, Wang X, Hommel B, Xia X, Qiu J, Fu Y, Zhou Z. In vivo analyses reveal hippocampal subfield volume reductions in adolescents with schizophrenia, but not with major depressive disorder. J Psychiatr Res 2023; 165:56-63. [PMID: 37459779 DOI: 10.1016/j.jpsychires.2023.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/25/2023] [Accepted: 07/10/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND Adult studies have reported atypicalities in the hippocampus and subfields in patients with schizophrenia (SCZ) and major depressive disorder (MDD). Both affective and psychotic disorders typically onset in adolescence, when human brain develops rapidly and shows increased susceptibility to adverse environments. However, few in vivo studies have investigated whether hippocampus subfield abnormalities occur in adolescence and whether they differ between SCZ and MDD cases. METHODS We recruited 150 adolescents (49 SCZ patients, 67 MDD patients, and 34 healthy controls) and obtained their structural images. We used FreeSurfer to automatically segment hippocampus into 12 subfields and analyzed subfield volumetric differences between groups by analysis of covariance, covarying for age, sex, and intracranial volume. Composite measures by summing subfield volumes were further compared across groups and analyzed in relation to clinical characteristic. RESULTS SCZ adolescents showed significant volume reductions in subfields of CA1, molecular layer, subiculum, parasubiculum, dentate gyrus and CA4 than healthy controls, and almost significant reductions, as compared to the MDD group, in left molecular layer, dentate gyrus, CA2/3 and CA4. Composite analyses showed smaller volumes in SCZ group than in healthy controls in all bilateral composite measures, and reduced volumes in comparison to MDD group in all left composite measures only. CONCLUSIONS SCZ adolescents exhibited both hippocampal subfield and composite volumes reduction, and also showed greater magnitude of deviance than those diagnosed with MDD, particularly in core CA regions. These results indicate a hippocampal disease process, suggesting a potential intervention marker of early psychotic patients and risk youths.
Collapse
Affiliation(s)
- Kangcheng Wang
- School of Psychology, Shandong Normal University, Jinan, 250358, China; Shandong Mental Health Center, Shandong University, Jinan, 250014, China
| | - Xingyan Li
- School of Psychology, Shandong Normal University, Jinan, 250358, China
| | - Xiaotong Wang
- School of Psychology, Shandong Normal University, Jinan, 250358, China
| | - Bernhard Hommel
- School of Psychology, Shandong Normal University, Jinan, 250358, China
| | - Xiaodi Xia
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiang Qiu
- Faculty of Psychology, Southwest University, Chongqing, 400715, China
| | - Yixiao Fu
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Zheyi Zhou
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China.
| |
Collapse
|
23
|
Ji F, Feng C, Qin J, Wang C, Zhang D, Su L, Wang W, Zhang M, Li H, Ma L, Lu W, Liu C, Teng Z, Hu B, Jian F, Xie J, Jiao J. Brain-specific Pd1 deficiency leads to cortical neurogenesis defects and depressive-like behaviors in mice. Cell Death Differ 2023; 30:2053-2065. [PMID: 37553426 PMCID: PMC10482844 DOI: 10.1038/s41418-023-01203-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 07/18/2023] [Accepted: 08/02/2023] [Indexed: 08/10/2023] Open
Abstract
Embryonic neurogenesis is tightly regulated by multiple factors to ensure the precise development of the cortex. Deficiency in neurogenesis may result in behavioral abnormalities. Pd1 is a well-known inhibitory immune molecule, but its function in brain development remains unknown. Here, we find brain specific deletion of Pd1 results in abnormal cortical neurogenesis, including enhanced proliferation of neural progenitors and reduced neuronal differentiation. In addition, neurons in Pd1 knockout mice exhibit abnormal morphology, both the total length and the number of primary dendrites were reduced. Moreover, Pd1cKO mice exhibit depressive-like behaviors, including immobility, despair, and anhedonia. Mechanistically, Pd1 regulates embryonic neurogenesis by targeting Pax3 through the β-catenin signaling pathway. The constitutive expression of Pax3 partly rescues the deficiency of neurogenesis in the Pd1 deleted embryonic brain. Besides, the administration of β-catenin inhibitor, XAV939, not only rescues abnormal brain development but also ameliorates depressive-like behaviors in Pd1cKO mice. Simultaneously, Pd1 plays a similar role in human neural progenitor cells (hNPCs) proliferation and differentiation. Taken together, our findings reveal the critical role and regulatory mechanism of Pd1 in embryonic neurogenesis and behavioral modulation, which could contribute to understanding immune molecules in brain development.
Collapse
Affiliation(s)
- Fen Ji
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| | - Chao Feng
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Sino-Danish College at University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Jie Qin
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Sino-Danish College at University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Chong Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Dongming Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Libo Su
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Wenwen Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Mengtian Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Hong Li
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Longbing Ma
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China
| | - Weicheng Lu
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Changmei Liu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Zhaoqian Teng
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Baoyang Hu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Fengzeng Jian
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China.
| | - Jingdun Xie
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China.
| | - Jianwei Jiao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|
24
|
Bigio B, Sagi Y, Barnhill O, Dobbin J, El Shahawy O, de Angelis P, Nasca C. Epigenetic embedding of childhood adversity: mitochondrial metabolism and neurobiology of stress-related CNS diseases. Front Mol Neurosci 2023; 16:1183184. [PMID: 37564785 PMCID: PMC10411541 DOI: 10.3389/fnmol.2023.1183184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/21/2023] [Indexed: 08/12/2023] Open
Abstract
This invited article ad memoriam of Bruce McEwen discusses emerging epigenetic mechanisms underlying the long and winding road from adverse childhood experiences to adult physiology and brain functions. The conceptual framework that we pursue suggest multidimensional biological pathways for the rapid regulation of neuroplasticity that utilize rapid non-genomic mechanisms of epigenetic programming of gene expression and modulation of metabolic function via mitochondrial metabolism. The current article also highlights how applying computational tools can foster the translation of basic neuroscience discoveries for the development of novel treatment models for mental illnesses, such as depression to slow the clinical manifestation of Alzheimer's disease. Citing an expression that many of us heard from Bruce, while "It is not possible to roll back the clock," deeper understanding of the biological pathways and mechanisms through which stress produces a lifelong vulnerability to altered mitochondrial metabolism can provide a path for compensatory neuroplasticity. The newest findings emerging from this mechanistic framework are among the latest topics we had the good fortune to discuss with Bruce the day before his sudden illness when walking to a restaurant in a surprisingly warm evening that preluded the snowstorm on December 18th, 2019. With this article, we wish to celebrate Bruce's untouched love for Neuroscience.
Collapse
Affiliation(s)
- Benedetta Bigio
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
| | - Yotam Sagi
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - Olivia Barnhill
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, NY, United States
| | - Josh Dobbin
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, NY, United States
| | - Omar El Shahawy
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States
| | - Paolo de Angelis
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, NY, United States
| | - Carla Nasca
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, NY, United States
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
25
|
Ahmad Hariza AM, Mohd Yunus MH, Murthy JK, Wahab S. Clinical Improvement in Depression and Cognitive Deficit Following Electroconvulsive Therapy. Diagnostics (Basel) 2023; 13:diagnostics13091585. [PMID: 37174977 PMCID: PMC10178332 DOI: 10.3390/diagnostics13091585] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Electroconvulsive therapy (ECT) is a long-standing treatment choice for disorders such as depression when pharmacological treatments have failed. However, a major drawback of ECT is its cognitive side effects. While numerous studies have investigated the therapeutic effects of ECT and its mechanism, much less research has been conducted regarding the mechanism behind the cognitive side effects of ECT. As both clinical remission and cognitive deficits occur after ECT, it is possible that both may share a common mechanism. This review highlights studies related to ECT as well as those investigating the mechanism of its outcomes. The process underlying these effects may lie within BDNF and NMDA signaling. Edema in the astrocytes may also be responsible for the adverse cognitive effects and is mediated by metabotropic glutamate receptor 5 and the protein Homer1a.
Collapse
Affiliation(s)
- Ahmad Mus'ab Ahmad Hariza
- Department of Physiology, Faculty of Medicine, UKM Medical Centre, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - Mohd Heikal Mohd Yunus
- Department of Physiology, Faculty of Medicine, UKM Medical Centre, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - Jaya Kumar Murthy
- Department of Physiology, Faculty of Medicine, UKM Medical Centre, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - Suzaily Wahab
- Department of Psychiatry, Faculty of Medicine, UKM Medical Centre, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
26
|
Quaioto BR, Borçoi AR, Mendes SO, Doblas PC, Dos Santos Vieira T, Arantes Moreno IA, Dos Santos JG, Hollais AW, Olinda AS, de Souza MLM, Freitas FV, Pinheiro JA, Cunha ER, Sorroche BP, Arantes LMRB, Álvares-da-Silva AM. Tobacco use modify exon IV BDNF gene methylation levels in depression. J Psychiatr Res 2023; 159:240-248. [PMID: 36753898 DOI: 10.1016/j.jpsychires.2023.01.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023]
Abstract
This study aimed to investigate BDNF gene methylation in individuals with depression based on tobacco use. Therefore, 384 adults from southeastern Brazil were recruited to assess depression, socioeconomic status, lifestyle, and methylation by pyrosequencing exon IV promoter region of the BDNF gene. The Generalized Linear Model (GzLM) was used to check the effect of depression, tobacco, and the interaction between depression and tobacco use in methylation levels. In addition, the Kruskal-Wallis test, followed by Dunn's post hoc test, was used to compare methylation levels. Interaction between depression and tobacco use was significant at levels of BDNF methylation in the CpG 5 (p = 0.045), 8 (p = 0.016), 9 (p = 0.042), 10 (p = 0.026) and mean 5-11 (p < 0.001). Dunn's post hoc test showed that individuals with depression and tobacco use compared to those with or without depression who did not use tobacco had lower levels of BDNF methylation in CpG 5, 6, 7, 8, 9, 11, and mean 5-11. Therefore, we suggest that tobacco use appears to interfere with BDNF gene methylation in depressed individuals.
Collapse
Affiliation(s)
- Bárbara Risse Quaioto
- Biotechnology Postgraduate Program/RENORBIO, Universidade Federal Do Espírito Santo, Vitória, Espírito Santo, Brazil.
| | - Aline Ribeiro Borçoi
- Biotechnology Postgraduate Program/RENORBIO, Universidade Federal Do Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Suzanny Oliveira Mendes
- Biotechnology Postgraduate Program/RENORBIO, Universidade Federal Do Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Paola Cerbino Doblas
- Biotechnology Postgraduate Program/RENORBIO, Universidade Federal Do Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Tamires Dos Santos Vieira
- Biotechnology Postgraduate Program/RENORBIO, Universidade Federal Do Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Ivana Alece Arantes Moreno
- Biotechnology Postgraduate Program/RENORBIO, Universidade Federal Do Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Joaquim Gasparini Dos Santos
- ICESP, Center for Translational Research in Oncology, Instituto Do Câncer Do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Multiprofessional Residency Program in Adult Oncology Care, Comissão de Residência Multiprofissional/Hospital Das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - André Willian Hollais
- Department of Physiological Sciences, Universidade Federal Do Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Amanda Sgrancio Olinda
- Biotechnology Postgraduate Program/RENORBIO, Universidade Federal Do Espírito Santo, Vitória, Espírito Santo, Brazil
| | | | - Flávia Vitorino Freitas
- Department of Pharmacy and Nutrition, Universidade Federal Do Espírito Santo, Alegre, Espírito Santo, Brazil
| | - Júlia Assis Pinheiro
- Biotechnology Postgraduate Program/RENORBIO, Universidade Federal Do Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Ester Ribeiro Cunha
- Department of Morphology, Universidade Federal Do Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Bruna Pereira Sorroche
- Molecular Oncology Research Center, Hospital Do Câncer de Barretos, Barretos, São Paulo, Brazil
| | | | - Adriana Madeira Álvares-da-Silva
- Biotechnology Postgraduate Program/RENORBIO, Universidade Federal Do Espírito Santo, Vitória, Espírito Santo, Brazil; Department of Morphology, Universidade Federal Do Espírito Santo, Vitória, Espírito Santo, Brazil
| |
Collapse
|
27
|
Han S, Zheng R, Li S, Zhou B, Jiang Y, Fang K, Wei Y, Wen B, Pang J, Li H, Zhang Y, Chen Y, Cheng J. Altered structural covariance network of nucleus accumbens is modulated by illness duration and severity of symptom in depression. J Affect Disord 2023; 324:334-340. [PMID: 36608848 DOI: 10.1016/j.jad.2022.12.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/19/2022] [Accepted: 12/31/2022] [Indexed: 01/06/2023]
Abstract
The differential structural covariance of nucleus accumbens (NAcc), playing a vital role in etiology and treatment, remains unclear in depression. We aimed to investigate whether structural covariance of NAcc was altered and how it was modulated by illness duration and severity of symptom measured with Hamilton Depression scale (HAMD). T1-weighted anatomical images of never-treated first-episode patients with depression (n = 195) and matched healthy controls (HCs, n = 78) were acquired. Gray matter volumes were calculated using voxel-based morphometry analysis for each subject. Then, we explored abnormal structural covariance of NAcc and how the abnormality was modulated by illness duration and severity of symptom. Patients with depression exhibited altered structural covariance of NAcc connected to key brain regions in reward system including the medial orbitofrontal cortex, amygdala, insula, parahippocampa gyrus, precuneus, thalamus, hippocampus and cerebellum. In addition, the structural covariance of the NAcc was distinctly modulated by illness duration and the severity of symptom in patients with depression. What is more, the structural covariance of the NAcc connected to hippocampus was modulated by these two factors at the same time. These results elucidate altered structural covariance of the NAcc and its distinct modulation of illness duration and severity of symptom.
Collapse
Affiliation(s)
- Shaoqiang Han
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Key Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, China; Engineering Technology Research Center for Detection and Application of Brain Function of Henan Province, China; Engineering Research Center of medical imaging intelligent diagnosis and treatment of Henan Province, China; Key Laboratory of Magnetic Resonance and Brain function of Henan Province, China; Key Laboratory of Brain Function and Cognitive Magnetic Resonance Imaging of Zhengzhou, China; Key Laboratory of Imaging Intelligence Research Medicine of Henan Province, China; Henan Engineering Research Center of Brain Function Development and Application, China.
| | - Ruiping Zheng
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Key Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, China; Engineering Technology Research Center for Detection and Application of Brain Function of Henan Province, China; Engineering Research Center of medical imaging intelligent diagnosis and treatment of Henan Province, China; Key Laboratory of Magnetic Resonance and Brain function of Henan Province, China; Key Laboratory of Brain Function and Cognitive Magnetic Resonance Imaging of Zhengzhou, China; Key Laboratory of Imaging Intelligence Research Medicine of Henan Province, China; Henan Engineering Research Center of Brain Function Development and Application, China
| | - Shuying Li
- Department of Psychiatry, the First Affiliated Hospital of Zhengzhou University, China
| | - Bingqian Zhou
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Key Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, China; Engineering Technology Research Center for Detection and Application of Brain Function of Henan Province, China; Engineering Research Center of medical imaging intelligent diagnosis and treatment of Henan Province, China; Key Laboratory of Magnetic Resonance and Brain function of Henan Province, China; Key Laboratory of Brain Function and Cognitive Magnetic Resonance Imaging of Zhengzhou, China; Key Laboratory of Imaging Intelligence Research Medicine of Henan Province, China; Henan Engineering Research Center of Brain Function Development and Application, China
| | - Yu Jiang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Key Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, China; Engineering Technology Research Center for Detection and Application of Brain Function of Henan Province, China; Engineering Research Center of medical imaging intelligent diagnosis and treatment of Henan Province, China; Key Laboratory of Magnetic Resonance and Brain function of Henan Province, China; Key Laboratory of Brain Function and Cognitive Magnetic Resonance Imaging of Zhengzhou, China; Key Laboratory of Imaging Intelligence Research Medicine of Henan Province, China; Henan Engineering Research Center of Brain Function Development and Application, China
| | - Keke Fang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, China
| | - Yarui Wei
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Key Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, China; Engineering Technology Research Center for Detection and Application of Brain Function of Henan Province, China; Engineering Research Center of medical imaging intelligent diagnosis and treatment of Henan Province, China; Key Laboratory of Magnetic Resonance and Brain function of Henan Province, China; Key Laboratory of Brain Function and Cognitive Magnetic Resonance Imaging of Zhengzhou, China; Key Laboratory of Imaging Intelligence Research Medicine of Henan Province, China; Henan Engineering Research Center of Brain Function Development and Application, China
| | - Baohong Wen
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Key Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, China; Engineering Technology Research Center for Detection and Application of Brain Function of Henan Province, China; Engineering Research Center of medical imaging intelligent diagnosis and treatment of Henan Province, China; Key Laboratory of Magnetic Resonance and Brain function of Henan Province, China; Key Laboratory of Brain Function and Cognitive Magnetic Resonance Imaging of Zhengzhou, China; Key Laboratory of Imaging Intelligence Research Medicine of Henan Province, China; Henan Engineering Research Center of Brain Function Development and Application, China
| | - Jianyue Pang
- Department of Psychiatry, the First Affiliated Hospital of Zhengzhou University, China
| | - Hengfen Li
- Department of Psychiatry, the First Affiliated Hospital of Zhengzhou University, China
| | - Yong Zhang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Key Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, China; Engineering Technology Research Center for Detection and Application of Brain Function of Henan Province, China; Engineering Research Center of medical imaging intelligent diagnosis and treatment of Henan Province, China; Key Laboratory of Magnetic Resonance and Brain function of Henan Province, China; Key Laboratory of Brain Function and Cognitive Magnetic Resonance Imaging of Zhengzhou, China; Key Laboratory of Imaging Intelligence Research Medicine of Henan Province, China; Henan Engineering Research Center of Brain Function Development and Application, China.
| | - Yuan Chen
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Key Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, China; Engineering Technology Research Center for Detection and Application of Brain Function of Henan Province, China; Engineering Research Center of medical imaging intelligent diagnosis and treatment of Henan Province, China; Key Laboratory of Magnetic Resonance and Brain function of Henan Province, China; Key Laboratory of Brain Function and Cognitive Magnetic Resonance Imaging of Zhengzhou, China; Key Laboratory of Imaging Intelligence Research Medicine of Henan Province, China; Henan Engineering Research Center of Brain Function Development and Application, China.
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Key Laboratory for Functional Magnetic Resonance Imaging and Molecular Imaging of Henan Province, China; Engineering Technology Research Center for Detection and Application of Brain Function of Henan Province, China; Engineering Research Center of medical imaging intelligent diagnosis and treatment of Henan Province, China; Key Laboratory of Magnetic Resonance and Brain function of Henan Province, China; Key Laboratory of Brain Function and Cognitive Magnetic Resonance Imaging of Zhengzhou, China; Key Laboratory of Imaging Intelligence Research Medicine of Henan Province, China; Henan Engineering Research Center of Brain Function Development and Application, China.
| |
Collapse
|
28
|
Li Y, Yang L, Li J, Gao W, Zhao Z, Dong K, Duan W, Dai B, Guo R. Antidepression of Xingpijieyu formula targets gut microbiota derived from depressive disorder. CNS Neurosci Ther 2022; 29:669-681. [PMID: 36550591 PMCID: PMC9873506 DOI: 10.1111/cns.14049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE This investigation aims to determine the antidepressant role of Xingpijieyu formula (XPJYF) mediated via gut microbiota (GM)-brain axis. METHODS We collected fecal microbiota from patients with depressive disorder (DD) and cultured microbiota in vitro. Some of microbiota were transplanted into germ-free rats with the intragastric administration of XPJYF grain at the dose of 1.533 g/kg/day. The behaviors were studied by forced swimming test, open field test, sucrose preference test, and body weight. Products of hypothalamus-pituitary-adrenocortical (HPA) axis, neurotransmitter, and serum cytokines were investigated by enzyme linked immunosorbent assay. Glial fibrillary acidic protein (GFAP), a biomarker of astrocyte, was quantified using immunofluorescence. Microbiota culturing in vitro after XPJYF treatment was analyze by 16 s RNA sequencing technology. We used lipopolysaccharide (LPS) to mimic activated rat primary astrocyte in vitro. Brain-derived neurotrophic factor (BDNF), cytokines, and oxidative stress factors were determined by western blotting, and glycometabolism in astrocyte was investigated by 2-deoxy-D-glucose (2-DG) uptake, adenosine triphosphate (ATP), and glucose-1-phosphate (G1P) kits. RESULTS Microbiota composition during 8 mg/ml of XPJYF (H12-8) for 12 h showed the more consistency. Lactococcus is enriched in DD-derived microbiota composition, and Biffdobacterium and Lactobacillus in H12-8 group. GLUCOSE1PMETAB-PWY and PWY-7328 of which biofunctions were dominantly encoded by Biffdobacterium were the top two of altered pathways. XPJYF improved behaviors and repressed astrocyte activation in depression rats. XPJYF elevated 2-DG uptake, ATP, glucose-1-phosphate, and brain-derived neurotrophic factor (BDNF), and inhibited cytokines and oxidative stress in LPS-induced astrocyte. CONCLUSION XPJYF treatment targets inflammation, activation, and glycometabolim in astrocyte via gut microbiota modulation, thereby improve animal behaviors, HPA axis dysfunction, and neurotransmitter synthesis in depression rats.
Collapse
Affiliation(s)
- Yannan Li
- Second Clinical Medical CollegeBeijing University of Chinese MedicineBeijingChina,Department of NeurologyDongfang Hospital Beijing University of Chinese MedicineBeijingChina
| | - Lixuan Yang
- Second Clinical Medical CollegeBeijing University of Chinese MedicineBeijingChina,Department of NeurologyDongfang Hospital Beijing University of Chinese MedicineBeijingChina
| | - Junnan Li
- Second Clinical Medical CollegeBeijing University of Chinese MedicineBeijingChina,Department of NeurologyDongfang Hospital Beijing University of Chinese MedicineBeijingChina
| | - Wei Gao
- Department of Mental HealthTsinghua University Yuquan HospitalBeijingChina
| | - Zhonghui Zhao
- Second Clinical Medical CollegeBeijing University of Chinese MedicineBeijingChina,Department of NeurologyDongfang Hospital Beijing University of Chinese MedicineBeijingChina
| | - Kaiqiang Dong
- Second Clinical Medical CollegeBeijing University of Chinese MedicineBeijingChina,Department of NeurologyDongfang Hospital Beijing University of Chinese MedicineBeijingChina
| | - Wenzhe Duan
- Second Clinical Medical CollegeBeijing University of Chinese MedicineBeijingChina,Department of NeurologyDongfang Hospital Beijing University of Chinese MedicineBeijingChina
| | - Baoan Dai
- Second Clinical Medical CollegeBeijing University of Chinese MedicineBeijingChina,Department of NeurologyDongfang Hospital Beijing University of Chinese MedicineBeijingChina
| | - Rongjuan Guo
- Department of NeurologyDongfang Hospital Beijing University of Chinese MedicineBeijingChina
| |
Collapse
|
29
|
Demers CH, Hankin BL, Hennessey EMP, Haase MH, Bagonis MM, Kim SH, Gilmore JH, Hoffman MC, Styner MA, Davis EP. Maternal adverse childhood experiences and infant subcortical brain volume. Neurobiol Stress 2022; 21:100487. [PMID: 36532374 PMCID: PMC9755027 DOI: 10.1016/j.ynstr.2022.100487] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/19/2022] [Accepted: 09/13/2022] [Indexed: 10/31/2022] Open
Abstract
Background A large body of research supports the deleterious effects of adverse childhood experiences (ACEs) on disease susceptibility and health for both the exposed individual and the next generation. It is likely that there is an intergenerational transmission of risk from mother to child; however, the mechanisms through which such risk is conferred remain unknown. The current study evaluated the association between maternal ACEs, neonatal brain development of the amygdala and hippocampus, and later infant negative emotionality at six months of age. Methods The sample included 85 mother-infant dyads (44 female infants) from a longitudinal study. Maternal ACEs were assessed with the Adverse Childhood Experiences Questionnaire (ACE-Q) and neonatal hippocampal and amygdala volume was assessed using structural magnetic resonance imaging (MRI). Infant negative emotionality was assessed at 6 months using the Infant Behavior Questionnaire (IBQ). Results Multivariate analyses demonstrated that maternal ACEs were associated with bilateral amygdala volume (F(2,78) = 3.697,p = .029). Specifically, higher maternal ACEs were associated with smaller left (β = -0.220, t(79) = -2.661, p = .009, R2 = 0.494, and right (β = -0.167, t(79) = -2.043, p = .044, R2 = 0.501) amygdala volume. No significant association between maternal ACEs and bilateral hippocampal volume (F(2,78) = 0.215,p = .0807) was found. Follow-up regression analyses demonstrated that both high maternal ACEs and smaller left amygdala volume were associated with higher infant negative emotionality at six months of age (β = .232, p = .040, R2 = 0.094, and β = -0.337, p = .022, R2 = 0.16, respectively) although statistically significant mediation of this effect was not observed (Indirect effect = 0.0187, 95% CI [-0.0016-0.0557]). Conclusions Maternal ACEs are associated with both newborn amygdala volume and subsequent infant negative emotionality. These findings linking maternal adverse childhood experiences and infant brain development and temperament provide evidence to support the intergenerational transmission of adversity from mother to child.
Collapse
Affiliation(s)
- Catherine H. Demers
- Department of Psychology, University of Denver, Denver, CO, USA
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Corresponding author. University of Denver, Department of Psychology, 2155 South Race Street, Denver, CO, 80208-3500, USA.
| | - Benjamin L. Hankin
- Department of Psychology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | | | | | - Maria M. Bagonis
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- PrimeNeuro, Durham, NC, USA
| | - Sun Hyung Kim
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John H. Gilmore
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - M. Camille Hoffman
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Obstetrics and Gynecology, Division of Maternal and Fetal Medicine, University of Colorado Denver School of Medicine, Aurora, CO, USA
| | - Martin A. Styner
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elysia Poggi Davis
- Department of Psychology, University of Denver, Denver, CO, USA
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
30
|
Brain structure and synaptic protein expression alterations after antidepressant treatment in a Wistar-Kyoto rat model of depression. J Affect Disord 2022; 314:293-302. [PMID: 35878834 DOI: 10.1016/j.jad.2022.07.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/20/2022] [Accepted: 07/17/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Structural MRI has demonstrated brain alterations in depression pathology and antidepressants treatment. While synaptic plasticity has been previously proposed as the potential underlying mechanism of MRI findings at a cellular and molecular scale, there is still insufficient evidence to link the MRI findings and synaptic plasticity mechanisms in depression pathology. METHODS In this study, a Wistar-Kyoto (WKY) depression rat model was treated with antidepressants (citalopram or Jie-Yu Pills) and tested in a series of behavioral tests and a 7.0 MRI scanner. We then measured dendritic spine density within altered brain regions. We also examined expression of synaptic marker proteins (PSD-95 and SYP). RESULTS WKY rats exhibited depression-like behaviors in the sucrose preference test (SPT) and forced swim test (FST), and anxiety-like behaviors in the open field test (OFT). Both antidepressants reversed behavioral changes in SPT and OFT but not in FST. We found a correlation between SPT performance and brain volumes as detected by MRI. All structural changes were consistent with alterations of the corpus callosum (white matter), dendritic spine density, as well as PSD95 and SYP expression at different levels. Two antidepressants similarly reversed these macro- and micro-changes. LIMITATIONS The single dose of antidepressants was the major limitation of this study. Further studies should focus on the white matter microstructure changes and myelin-related protein alterations, in addition to comparing the effects of ketamine. CONCLUSION Translational evidence links structural MRI changes and synaptic plasticity alterations, which promote our understanding of SPT mechanisms and antidepressant response in WKY rats.
Collapse
|
31
|
Lidauer K, Pulli EP, Copeland A, Silver E, Kumpulainen V, Hashempour N, Merisaari H, Saunavaara J, Parkkola R, Lähdesmäki T, Saukko E, Nolvi S, Kataja EL, Karlsson L, Karlsson H, Tuulari JJ. Subcortical and hippocampal brain segmentation in 5-year-old children: validation of FSL-FIRST and FreeSurfer against manual segmentation. Eur J Neurosci 2022; 56:4619-4641. [PMID: 35799402 PMCID: PMC9543285 DOI: 10.1111/ejn.15761] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 06/06/2022] [Accepted: 06/06/2022] [Indexed: 11/28/2022]
Abstract
Developing accurate subcortical volumetric quantification tools is crucial for neurodevelopmental studies, as they could reduce the need for challenging and time‐consuming manual segmentation. In this study, the accuracy of two automated segmentation tools, FSL‐FIRST (with three different boundary correction settings) and FreeSurfer, were compared against manual segmentation of the hippocampus and subcortical nuclei, including the amygdala, thalamus, putamen, globus pallidus, caudate and nucleus accumbens, using volumetric and correlation analyses in 80 5‐year‐olds. Both FSL‐FIRST and FreeSurfer overestimated the volume on all structures except the caudate, and the accuracy varied depending on the structure. Small structures such as the amygdala and nucleus accumbens, which are visually difficult to distinguish, produced significant overestimations and weaker correlations with all automated methods. Larger and more readily distinguishable structures such as the caudate and putamen produced notably lower overestimations and stronger correlations. Overall, the segmentations performed by FSL‐FIRST's default pipeline were the most accurate, whereas FreeSurfer's results were weaker across the structures. In line with prior studies, the accuracy of automated segmentation tools was imperfect with respect to manually defined structures. However, apart from amygdala and nucleus accumbens, FSL‐FIRST's agreement could be considered satisfactory (Pearson correlation > 0.74, intraclass correlation coefficient (ICC) > 0.68 and Dice score coefficient (DSC) > 0.87) with highest values for the striatal structures (putamen, globus pallidus, caudate) (Pearson correlation > 0.77, ICC > 0.87 and DSC > 0.88, respectively). Overall, automated segmentation tools do not always provide satisfactory results, and careful visual inspection of the automated segmentations is strongly advised.
Collapse
Affiliation(s)
- Kristian Lidauer
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland
| | - Elmo P Pulli
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland.,Department of Psychiatry, Turku University Hospital, University of Turku, Turku, Finland
| | - Anni Copeland
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland.,Department of Psychiatry, Turku University Hospital, University of Turku, Turku, Finland
| | - Eero Silver
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland.,Department of Psychiatry, Turku University Hospital, University of Turku, Turku, Finland
| | - Venla Kumpulainen
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland
| | - Niloofar Hashempour
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland
| | - Harri Merisaari
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland.,Department of Radiology, University of Turku, Turku, Finland
| | - Jani Saunavaara
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | - Riitta Parkkola
- Department of Radiology, University of Turku, Turku, Finland.,Department of Radiology, Turku University Hospital, Turku, Finland
| | - Tuire Lähdesmäki
- Department of Paediatric Neurology, Turku University Hospital and University of Turku, Turku, Finland
| | - Ekaterina Saukko
- Department of Radiology, Turku University Hospital, Turku, Finland
| | - Saara Nolvi
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland.,Turku Institute for Advanced Studies, University of Turku, Turku, Finland.,Department of Psychology and Speech-Language Pathology, University of Turku, Turku, Finland
| | - Eeva-Leena Kataja
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland
| | - Linnea Karlsson
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland.,Department of Psychiatry, Turku University Hospital, University of Turku, Turku, Finland.,Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Hasse Karlsson
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland.,Department of Psychiatry, Turku University Hospital, University of Turku, Turku, Finland.,Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Jetro J Tuulari
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland.,Department of Psychiatry, Turku University Hospital, University of Turku, Turku, Finland.,Turku Collegium for Science, Medicine and Technology, University of Turku, Turku, Finland.,Department of Psychiatry, University of Oxford, UK (Sigrid Juselius Fellowship), United Kingdom
| |
Collapse
|
32
|
Li N, Song Q, Su W, Guo X, Wang H, Liang Q, Liang M, Qu G, Ding X, Zhou X, Sun Y. Exposure to indoor air pollution from solid fuel and its effect on depression: a systematic review and meta-analysis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:49553-49567. [PMID: 35593981 DOI: 10.1007/s11356-022-20841-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/11/2022] [Indexed: 06/15/2023]
Abstract
A growing body of research has investigated the relationship between indoor air pollution from solid fuel and depression risk. Our study aimed to elucidate the relationship between indoor air pollution from solid fuel and depression in observational studies. The effect of indoor air pollution on depression was estimated using pooled odds ratios (ORs) with 95% confidence intervals (CIs). Heterogeneity was evaluated by the I-squared value (I2), and the random-effects model was adopted as the summary method. We finalized nine articles with 70,214 subjects. The results showed a statistically positive relationship between the use of household solid fuel and depression (OR = 1.22, 95% CI = 1.09-1.36). Subgroup analysis based on fuel type groups demonstrated that indoor air pollution from solid fuel was a higher risk to depression (OR = 1.24, 95% CI = 1. 10-1.39; I2 = 67.0%) than that from biomass (OR = 1.18, 95% CI = 0.96-1.45; I2 = 66.5%). In terms of fuel use, the use of solid fuel for cooking and heating increased depression risk, and the pooled ORs were 1.21 (95% CI = 1.08-1.36) and 1.23 (95% CI = 1.13-1.34). Exposure to indoor air pollution from solid fuel might increase depression risk.
Collapse
Affiliation(s)
- Ning Li
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Qiuxia Song
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Wanying Su
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Xianwei Guo
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Hao Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Qiwei Liang
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Anhui Provincial Children's Hospital/Children's Hospital of Anhui Medical University, Hefei, 230051, People's Republic of China
| | - Mingming Liang
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Guangbo Qu
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Xiuxiu Ding
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Xiaoqin Zhou
- Chaohu Hospital, Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China
| | - Yehuan Sun
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China.
- Chaohu Hospital, Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China.
- Center for Evidence-Based Practice, Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China.
| |
Collapse
|
33
|
Blank TS, Meyer BM, Wieser M, Rabl U, Schögl P, Pezawas L. Brain morphometry and connectivity differs between adolescent- and adult-onset major depressive disorder. Depress Anxiety 2022; 39:387-396. [PMID: 35421280 PMCID: PMC9323432 DOI: 10.1002/da.23254] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 03/05/2022] [Accepted: 03/13/2022] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Early-onset (EO) major depressive disorder (MDD) patients experience more depressive episodes and an increased risk of relapse. Thus, on a neurobiological level, adult EO patients might display brain structure and function different from adult-onset (AO) patients. METHODS A total of 103 patients (66 females) underwent magnetic resonance imaging. Structural measures of gray matter volume (GMV) and functional connectivity networks during resting state were compared between EO (≤19 years) and AO groups. Four residual major depression symptoms, mood, anxiety, insomnia, and somatic symptoms, were correlated with GMV between groups. RESULTS We found comparatively increased GMV in the EO group, namely the medial prefrontal and insular cortex, as well as the anterior hippocampus. Functional networks in EO patients showed a comparatively weaker synchronization of the left hippocampus with the adjacent amygdala, and a stronger integration with nodes in the contralateral prefrontal cortex and supramarginal gyrus. Volumetric analysis of depression symptoms associated the caudate nuclei with symptoms of insomnia, and persisting mood symptoms with the right amygdala, while finding no significant clusters for somatic and anxiety symptoms. CONCLUSIONS The study highlights the important role of the hippocampus and the prefrontal cortex in EO patients as part of emotion-regulation networks. Results in EO patients demonstrated subcortical volume changes irrespective of sleep and mood symptom recovery, which substantiates adolescence as a pivotal developmental phase for MDD. Longitudinal studies are needed to differentiate neural recovery trajectories while accounting for age of onset.
Collapse
Affiliation(s)
- Thomas S. Blank
- Department of Psychiatry and PsychotherapyMedical University of ViennaWienAustria
| | - Bernhard M. Meyer
- Department of Psychiatry and PsychotherapyMedical University of ViennaWienAustria
| | - Marie‐Kathrin Wieser
- Department of Psychiatry and PsychotherapyMedical University of ViennaWienAustria
| | - Ulrich Rabl
- Department of Psychiatry and PsychotherapyMedical University of ViennaWienAustria
| | - Paul Schögl
- Department of Psychiatry and PsychotherapyMedical University of ViennaWienAustria
| | - Lukas Pezawas
- Department of Psychiatry and PsychotherapyMedical University of ViennaWienAustria
| |
Collapse
|
34
|
Lee SM, Milillo MM, Krause-Sorio B, Siddarth P, Kilpatrick L, Narr KL, Jacobs JP, Lavretsky H. Gut Microbiome Diversity and Abundance Correlate with Gray Matter Volume (GMV) in Older Adults with Depression. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19042405. [PMID: 35206594 PMCID: PMC8872347 DOI: 10.3390/ijerph19042405] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/08/2022] [Accepted: 02/15/2022] [Indexed: 01/27/2023]
Abstract
Growing evidence supports the concept that bidirectional brain–gut microbiome interactions play an important mechanistic role in aging, as well as in various neuropsychiatric conditions including depression. Gray matter volume (GMV) deficits in limbic regions are widely observed in geriatric depression (GD). We therefore aimed to explore correlations between gut microbial measures and GMV within these regions in GD. Sixteen older adults (>60 years) with GD (37.5% female; mean age, 70.6 (SD = 5.7) years) were included in the study and underwent high-resolution T1-weighted structural MRI scanning and stool sample collection. GMV was extracted from bilateral regions of interest (ROI: hippocampus, amygdala, nucleus accumbens) and a control region (pericalcarine). Fecal microbiota composition and diversity were assessed by 16S ribosomal RNA gene sequencing. There were significant positive associations between alpha diversity measures and GMV in both hippocampus and nucleus accumbens. Additionally, significant positive associations were present between hippocampal GMV and the abundance of genera Family_XIII_AD3011_group, unclassified Ruminococcaceae, and Oscillibacter, as well as between amygdala GMV and the genera Lachnospiraceae_NK4A136_group and Oscillibacter. Gut microbiome may reflect brain health in geriatric depression. Future studies with larger samples and the experimental manipulation of gut microbiome may clarify the relationship between microbiome measures and neuroplasticity.
Collapse
Affiliation(s)
- Sungeun Melanie Lee
- Department of Psychiatry, Semel Institute for Neuroscience, University of California Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90024, USA; (S.M.L.); (M.M.M.); (B.K.-S.); (P.S.); (L.K.)
| | - Michaela M. Milillo
- Department of Psychiatry, Semel Institute for Neuroscience, University of California Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90024, USA; (S.M.L.); (M.M.M.); (B.K.-S.); (P.S.); (L.K.)
| | - Beatrix Krause-Sorio
- Department of Psychiatry, Semel Institute for Neuroscience, University of California Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90024, USA; (S.M.L.); (M.M.M.); (B.K.-S.); (P.S.); (L.K.)
| | - Prabha Siddarth
- Department of Psychiatry, Semel Institute for Neuroscience, University of California Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90024, USA; (S.M.L.); (M.M.M.); (B.K.-S.); (P.S.); (L.K.)
| | - Lisa Kilpatrick
- Department of Psychiatry, Semel Institute for Neuroscience, University of California Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90024, USA; (S.M.L.); (M.M.M.); (B.K.-S.); (P.S.); (L.K.)
| | - Katherine L. Narr
- Brain Research Institute, 635 Charles E Young Drive South, Los Angeles, CA 90095, USA;
| | - Jonathan P. Jacobs
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave., Los Angeles, CA 90095, USA;
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave., Los Angeles, CA 90095, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System and Department of Medicine and Human Genetics, 11301 Wilshire Blvd., Los Angeles, CA 90073, USA
| | - Helen Lavretsky
- Department of Psychiatry, Semel Institute for Neuroscience, University of California Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90024, USA; (S.M.L.); (M.M.M.); (B.K.-S.); (P.S.); (L.K.)
- Correspondence:
| |
Collapse
|
35
|
Xie T, Li R, Long X, Chen J, Ye L, Wang J, Jiang G, Lv J. Magnetic resonance imaging features of hippocampus and mechanism of neurocognitive dysfunction for antiepileptic drugs in treatment of depression rats. Bioengineered 2022; 13:4646-4657. [PMID: 35148670 PMCID: PMC8973768 DOI: 10.1080/21655979.2021.2018537] [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] [Indexed: 11/02/2022] Open
Abstract
To explore the effects of antiepileptic drug sodium valproate on magnetic resonance imaging (MRI) images, neurological cognition, and JAK1/STAT3 pathway in hippocampus of rats with depression, 30 Sprague Dawley (SD) rats were included. The depression model (DM) was prepared through the chronic stress restraint test. Some model rats were injected with 10 mg/kg sodium valproate into abdominal cavity before modeling (RT group)), and healthy rats were selected as controls (healthy control (HC) group). Depth of split brain was greatly increased in DM group, and nitrogen-acetyl aspartic acid (NAA)/creatine (Cr), glutamic acid (Glu)/Cr, and choline (Cho)/Cr ratios were greatly reduced (P < 0.05). Behavioral test results showed that sugar water preference rate, escape latency, and divergence index in DM group were greatly reduced (P < 0.05), and cumulative immobility time, target quadrant stay time, and number of crossings in forced swimming and tail suspension were prolonged dramatically (P < 0.05), with no difference between the two groups (P > 0.05). Expression levels of interleukin 1β (IL-1β) and interleukin 6 (IL-6) in hippocampus of DM group were obviously increased (P < 0.05), and expression levels of JAK1 and STAT3 were decreased visibly (P < 0.05), with no difference between the two (P > 0.05). In summary, anti-epileptic drug sodium valproate effectively improves hippocampal volume characteristics and memory and neurocognitive dysfunction of depression models.
Collapse
Affiliation(s)
- Tuxiu Xie
- Department of General Practice, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Ran Li
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Xiaobing Long
- Department of Emergency, the Center of Emergency and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Jun Chen
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Lu Ye
- Department of Emergency, the Center of Emergency and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Jing Wang
- Department of Emergency, the Center of Emergency and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Guijun Jiang
- Department of Emergency, the Center of Emergency and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Jingjun Lv
- Department of Emergency, the Center of Emergency and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
36
|
Hackman DA, Duan L, McConnell EE, Lee WJ, Beak AS, Kraemer DJM. School Climate, Cortical Structure, and Socioemotional Functioning: Associations across Family Income Levels. J Cogn Neurosci 2022; 34:1842-1865. [PMID: 35171285 DOI: 10.1162/jocn_a_01833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
School climates are important for children's socioemotional development and may also serve as protective factors in the context of adversity. Nevertheless, little is known about the potential neural mechanisms of such associations, as there has been limited research concerning the relation between school climate and brain structure, particularly for brain regions relevant for mental health and socioemotional functioning. Moreover, it remains unclear whether the role of school climate differs depending on children's socioeconomic status. We addressed these questions in baseline data for 9- to 10-year-olds from the Adolescent Brain and Cognitive Development study (analytic sample for socioemotional outcomes, n = 8,887), conducted at 21 sites across the United States. Cortical thickness, cortical surface area, and subcortical volume were derived from T1-weighted brain magnetic resonance imaging. School climate was measured by youth report, and socioemotional functioning was measured by both youth and parent report. A positive school climate and higher family income were associated with lower internalizing and externalizing symptoms, with no evidence of moderation. There were no associations between school climate and cortical thickness or subcortical volume, although family income was positively associated with hippocampal volume. For cortical surface area, however, there was both a positive association with family income and moderation: There was an interaction between school climate and income for total cortical surface area and locally in the lateral orbitofrontal cortex. In all cases, there was an unexpected negative association between school climate and cortical surface area in the lower-income group. Consequently, although the school climate appears to be related to better socioemotional function for all youth, findings suggest that the association between a positive school environment and brain structure only emerges in the context of socioeconomic stress and adversity. Longitudinal data are needed to understand the role of these neural differences in socioemotional functioning over time.
Collapse
Affiliation(s)
| | - Lei Duan
- University of Southern California, Los Angeles
| | | | | | | | | |
Collapse
|
37
|
Tsotsi S, Rifkin-Graboi A, Borelli JL, Chong YS, Rajadurai VS, Chua MC, Broekman B, Meaney M, Qiu A. Neonatal brain and physiological reactivity in preschoolers: An initial investigation in an Asian sample. J Psychiatr Res 2022; 146:219-227. [PMID: 34809993 DOI: 10.1016/j.jpsychires.2021.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 10/04/2021] [Accepted: 11/04/2021] [Indexed: 11/18/2022]
Abstract
Parasympathetic nervous system (PNS) activity is important to physiological regulation. Limbic structures are important in determining what information the PNS receives, potentially influencing concurrent physiological responsivity and, ultimately, shaping PNS development. Yet, whether individual differences in these structures are linked to PNS activity in early childhood remains unclear. Here, in an exploratory capacity, we examined the association between neonatal limbic structures (i.e., the left and right amygdala and hippocampus) and preschoolers' resting-state respiratory sinus arrhythmia (RSA). RSA is a measure of heart-rate variability, a physiological marker that reflects fluctuation in the PNS and is often found predictive of emotion regulation and psychological wellbeing. Data were extracted from the "Growing Up in Singapore towards Healthy Outcomes" (GUSTO) cohort (n = 73, 39 girls). Neonatal limbic volume was collected within two weeks after birth while infants were asleep. Resting-state RSA was collected during a coloring session at 42 months of age. After controlling for potential confounders, a Bonferroni-corrected significant association between neonatal left hippocampal volume and resting-state RSA emerged wherein larger hippocampal volume was associated with higher resting-state RSA. No significant associations were present between resting-state RSA and right or left amygdala, or right hippocampal volume. These findings contribute to an increasing body of evidence aiming at enhancing our understanding of neurobiological underpinnings of parasympathetic activity and modulation. Results are also discussed with reference to ideas concerning biological sensitivity to context, as both left hippocampal volume and resting-state RSA were previously found to moderate associations between adversity and psychological function.
Collapse
Affiliation(s)
- Stella Tsotsi
- PROMENTA Research Centre, Department of Psychology, University of Oslo, Oslo, Norway.
| | - Anne Rifkin-Graboi
- Centre for Research in Child Development, National Institute of Education, Nanyang Technological University, Singapore
| | - Jessica L Borelli
- Department of Psychological Science, School of Social Ecology, University of California, Irvine, USA
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore; Department of Obstetrics & Gynecology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore
| | - Victor Samuel Rajadurai
- Department of Neonatology, Kandang Kerbau Women and Children's Hospital, Singapore; Duke-NUS Medical School, Singapore
| | - Mei Chien Chua
- Department of Neonatology, Kandang Kerbau Women and Children's Hospital, Singapore; Duke-NUS Medical School, Singapore
| | - Birit Broekman
- Department of Psychiatry, OLVG and Amsterdam UMC, VU University, Amsterdam, the Netherlands
| | - Michael Meaney
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore; McGill University, Montreal, Canada
| | - Anqi Qiu
- Department of Biomedical Engineering, National University of Singapore, Singapore.
| |
Collapse
|
38
|
Wang X, Tong B, Hui R, Hou C, Zhang Z, Zhang L, Xie B, Ni Z, Cong B, Ma C, Wen D. The Role of Hyperthermia in Methamphetamine-Induced Depression-Like Behaviors: Protective Effects of Coral Calcium Hydride. Front Mol Neurosci 2022; 14:808807. [PMID: 35058751 PMCID: PMC8764150 DOI: 10.3389/fnmol.2021.808807] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022] Open
Abstract
Methamphetamine (METH) abuse causes irreversible damage to the central nervous system and leads to psychiatric symptoms including depression. Notably, METH-induced hyperthermia is a crucial factor in the development of these symptoms, as it aggravates METH-induced neurotoxicity. However, the role of hyperthermia in METH-induced depression-like behaviors needs to be clarified. In the present study, we treated mice with different doses of METH under normal (NAT) or high ambient temperatures (HAT). We found that HAT promoted hyperthermia after METH treatment and played a key role in METH-induced depression-like behaviors in mice. Intriguingly, chronic METH exposure (10 mg/kg, 7 or 14 days) or administration of an escalating-dose (2 ∼ 15 mg/kg, 3 days) of METH under NAT failed to induce depression-like behaviors. However, HAT aggravated METH-induced damage of hippocampal synaptic plasticity, reaction to oxidative stress, and neuroinflammation. Molecular hydrogen acts as an antioxidant and anti-inflammatory agent and has been shown to have preventive and therapeutic applicability in a wide range of diseases. Coral calcium hydride (CCH) is a newly identified hydrogen-rich powder which produces hydrogen gas gradually when exposed to water. Herein, we found that CCH pretreatment significantly attenuated METH-induced hyperthermia, and administration of CCH after METH exposure also inhibited METH-induced depression-like behaviors and reduced the hippocampal synaptic plasticity damage. Moreover, CCH effectively reduced the activity of lactate dehydrogenase and decreased malondialdehyde, TNF-α and IL-6 generation in hippocampus. These results suggest that CCH is an efficient hydrogen-rich agent, which has a potential therapeutic applicability in the treatment of METH abusers.
Collapse
Affiliation(s)
- Xintao Wang
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Bonan Tong
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Rongji Hui
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Congcong Hou
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Zilu Zhang
- The First Clinical Medical College of Peking University Health Science Center, Peking University, Beijing, China
| | - Ludi Zhang
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Bing Xie
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Zhiyu Ni
- School of Basic Medical Sciences, Hebei University, Baoding, China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
- *Correspondence: Chunling Ma,
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
- Di Wen,
| |
Collapse
|
39
|
Murphy F, Nasa A, Cullinane D, Raajakesary K, Gazzaz A, Sooknarine V, Haines M, Roman E, Kelly L, O'Neill A, Cannon M, Roddy DW. Childhood Trauma, the HPA Axis and Psychiatric Illnesses: A Targeted Literature Synthesis. Front Psychiatry 2022; 13:748372. [PMID: 35599780 PMCID: PMC9120425 DOI: 10.3389/fpsyt.2022.748372] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Studies of early life stress (ELS) demonstrate the long-lasting effects of acute and chronic stress on developmental trajectories. Such experiences can become biologically consolidated, creating individual vulnerability to psychological and psychiatric issues later in life. The hippocampus, amygdala, and the medial prefrontal cortex are all important limbic structures involved in the processes that undermine mental health. Hyperarousal of the sympathetic nervous system with sustained allostatic load along the Hypothalamic Pituitary Adrenal (HPA) axis and its connections has been theorized as the basis for adult psychopathology following early childhood trauma. In this review we synthesize current understandings and hypotheses concerning the neurobiological link between childhood trauma, the HPA axis, and adult psychiatric illness. We examine the mechanisms at play in the brain of the developing child and discuss how adverse environmental stimuli may become biologically incorporated into the structure and function of the adult brain via a discussion of the neurosequential model of development, sensitive periods and plasticity. The HPA connections and brain areas implicated in ELS and psychopathology are also explored. In a targeted review of HPA activation in mood and psychotic disorders, cortisol is generally elevated across mood and psychotic disorders. However, in bipolar disorder and psychosis patients with previous early life stress, blunted cortisol responses are found to awakening, psychological stressors and physiological manipulation compared to patients without previous early life stress. These attenuated responses occur in bipolar and psychosis patients on a background of increased cortisol turnover. Although cortisol measures are generally raised in depression, the evidence for a different HPA activation profile in those with early life stress is inconclusive. Further research is needed to explore the stress responses commonalities between bipolar disorder and psychosis in those patients with early life stress.
Collapse
Affiliation(s)
- Felim Murphy
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Anurag Nasa
- Department of Psychiatry, Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | | | - Kesidha Raajakesary
- Department of Psychiatry, Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Areej Gazzaz
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Vitallia Sooknarine
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Madeline Haines
- Department of Psychiatry, Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Elena Roman
- Department of Psychiatry, Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Linda Kelly
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Aisling O'Neill
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mary Cannon
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Darren William Roddy
- Department of Psychiatry, Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
40
|
Preschool sleep and depression interact to predict gray matter volume trajectories across late childhood to adolescence. Dev Cogn Neurosci 2021; 53:101053. [PMID: 34933170 PMCID: PMC8693016 DOI: 10.1016/j.dcn.2021.101053] [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: 07/21/2021] [Revised: 11/29/2021] [Accepted: 12/15/2021] [Indexed: 11/21/2022] Open
Abstract
There is a close relationship between sleep and depression, and certain maladaptive outcomes of sleep problems may only be apparent in individuals with heightened levels of depression. In a sample enriched for preschool depression, we examined how sleep and depression in early childhood interact to predict later trajectories of gray matter volume. Participants (N = 161) were recruited and assessed during preschool (ages 3–6 years) and were later assessed with five waves of structural brain imaging, spanning from late childhood to adolescence. Sleep and depression were assessed using a semi-structured parent interview when the children were preschool-aged, and total gray matter volume was calculated at each scan wave. Although sleep disturbances alone did not predict gray matter volume/trajectories, preschool sleep and depression symptoms interacted to predict later total gray matter volume and the trajectory of decline in total gray matter volume. Sleep disturbances in the form of longer sleep onset latencies, increased irregularity in the child’s sleep schedule, and higher levels of daytime sleepiness in early childhood were all found to interact with early childhood depression severity to predict later trajectories of cortical gray matter volume. Findings provide evidence of the interactive effects of preschool sleep and depression symptoms on later neurodevelopment.
Collapse
|
41
|
Tsyglakova M, Huskey AM, Hurst EH, Telep NM, Wilding MC, Babington ME, Rainville JR, Hodes GE. Sex and region-specific effects of variable stress on microglia morphology. Brain Behav Immun Health 2021; 18:100378. [PMID: 34820640 PMCID: PMC8600001 DOI: 10.1016/j.bbih.2021.100378] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/25/2022] Open
Abstract
Major Depressive Disorder (MDD) is a common and debilitating mood disorder that is more prevalent in women than men. In humans, PET imaging of microglia activation is currently being explored as a potential biomarker of MDD and suicidal ideation. Stress is a trigger for many mood disorders, including MDD. Microglial changes in morphology and activation state in response to stress has been reported in various brain regions, but most studies only examined male subjects. Here we report changes in microglia morphology in the nucleus accumbens (NAc) and subregions of the hippocampus (HPC) in both male and female mice following variable stress of 6 or 28 days in duration. Our data demonstrate that after 6 days of stress, microglia in the female NAc and dentate gyrus have a reduction in homeostatic associated morphology and an increase in primed microglia. After 28 days some of these sex specific stress effects were still present in microglia within the NAc but not the dentate gyrus. There were no effects of stress in either sex at either timepoint in CA1. In female mice, anti-inflammatory activation of microglia using rosiglitazone promoted sociability behavior after 6 days of stress. Furthermore, both drug and stress have impact on microglia morphology and activation state in the NAc. These data suggest that microglia morphology and activation state are altered by 6 days of variable stress in a region-specific manner and may contribute to, or potentially compensate for, the onset of stress susceptibility rather than impacting long term exposure to stress.
Collapse
Affiliation(s)
- Mariya Tsyglakova
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Alisa M. Huskey
- Department of Psychology, University of Arizona, Tucson, AZ, USA
| | - Emily H. Hurst
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Natalie M. Telep
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Mary C. Wilding
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Meghan E. Babington
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Jennifer R. Rainville
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Georgia E. Hodes
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| |
Collapse
|
42
|
Chen Y, Li L, Zhang J, Cui H, Wang J, Wang C, Shi M, Fan H. Dexmedetomidine Alleviates Lipopolysaccharide-Induced Hippocampal Neuronal Apoptosis via Inhibiting the p38 MAPK/c-Myc/CLIC4 Signaling Pathway in Rats. Mol Neurobiol 2021; 58:5533-5547. [PMID: 34363182 DOI: 10.1007/s12035-021-02512-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/02/2021] [Indexed: 12/11/2022]
Abstract
Dexmedetomidine (DEX) has multiple biological effects. Here, we investigated the neuroprotective role and molecular mechanism of DEX against lipopolysaccharide (LPS)-induced hippocampal neuronal apoptosis. Sprague Dawley rats were intraperitoneally injected with LPS (10 mg/kg) and/or DEX (30 µg/kg). We found that DEX improved LPS-induced alterations of hippocampal microstructure (necrosis and neuronal loss in the CA1 and CA3 regions) and ultrastructure (mitochondrial damage). DEX also attenuated LPS-induced inflammation and hippocampal apoptosis by inhibiting the increase of interleukin-1β, interleukin-6, interleukin-18, and tumor necrosis factor-α levels and downregulating the expression of mitochondrial apoptosis pathway-related proteins. Moreover, DEX prevented the LPS-induced activation of the c-Myc/chloride intracellular channel 4 (CLIC4) pathway. DEX inhibited the p38 MAPK pathway, but not JNK and ERK. To further clarify whether DEX alleviated LPS-induced neuronal apoptosis through the p38 MAPK/c-Myc/CLIC4 pathway, we treated PC12 cells with p38 MAPK inhibitor SB203582 (10 µM). DEX had the same effect as SB203582 in reducing the protein and mRNA expression of c-Myc and CLIC4. Furthermore, DEX and SB203582 diminished LPS-induced apoptosis, indicated by decreased Bax and Tom20 fluorescent double-stained cells, reduced annexin V-FITC/PI apoptosis rate, and reduced protein expression levels of Bax, cytochrome C, cleaved caspase-9, and cleaved caspase-3. Taken together, the findings indicate that DEX attenuates LPS-induced hippocampal neuronal apoptosis by regulating the p38 MAPK/c-Myc/CLIC4 signaling pathway. These findings provide new insights into the mechanism of Alzheimer's disease and depression and may help aid in drug development for these diseases.
Collapse
Affiliation(s)
- Yongping Chen
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China
| | - Lin Li
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China
| | - Jiuyan Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China
| | - Hailin Cui
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China
| | - Jiucheng Wang
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China
| | - Chuqiao Wang
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China
| | - Mingxian Shi
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China
| | - Honggang Fan
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
43
|
Qiu W, Cai X, Zheng C, Qiu S, Ke H, Huang Y. Update on the Relationship Between Depression and Neuroendocrine Metabolism. Front Neurosci 2021; 15:728810. [PMID: 34531719 PMCID: PMC8438205 DOI: 10.3389/fnins.2021.728810] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/11/2021] [Indexed: 12/27/2022] Open
Abstract
Through the past decade of research, the correlation between depression and metabolic diseases has been noticed. More and more studies have confirmed that depression is comorbid with a variety of metabolic diseases, such as obesity, diabetes, metabolic syndrome and so on. Studies showed that the underlying mechanisms of both depression and metabolic diseases include chronic inflammatory state, which is significantly related to the severity. In addition, they also involve endocrine, immune systems. At present, the effects of clinical treatments of depression is limited. Therefore, exploring the co-disease mechanism of depression and metabolic diseases is helpful to find a new clinical therapeutic intervention strategy. Herein, focusing on the relationship between depression and metabolic diseases, this manuscript aims to provide an overview of the comorbidity of depression and metabolic.
Collapse
Affiliation(s)
- Wenxin Qiu
- Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaodan Cai
- Fujian Medical University, Fuzhou, Fujian, China
| | | | - Shumin Qiu
- Fujian Medical University, Fuzhou, Fujian, China
| | - Hanyang Ke
- Fujian Medical University, Fuzhou, Fujian, China
| | - Yinqiong Huang
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
44
|
A Role of BDNF in the Depression Pathogenesis and a Potential Target as Antidepressant: The Modulator of Stress Sensitivity "Shati/Nat8l-BDNF System" in the Dorsal Striatum. Pharmaceuticals (Basel) 2021; 14:ph14090889. [PMID: 34577589 PMCID: PMC8469819 DOI: 10.3390/ph14090889] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Abstract
Depression is one of the most common mental diseases, with increasing numbers of patients globally each year. In addition, approximately 30% of patients with depression are resistant to any treatment and do not show an expected response to first-line antidepressant drugs. Therefore, novel antidepressant agents and strategies are required. Although depression is triggered by post-birth stress, while some individuals show the pathology of depression, others remain resilient. The molecular mechanisms underlying stress sensitivity remain unknown. Brain-derived neurotrophic factor (BDNF) has both pro- and anti-depressant effects, dependent on brain region. Considering the strong region-specific contribution of BDNF to depression pathogenesis, the regulation of BDNF in the whole brain is not a beneficial strategy for the treatment of depression. We reviewed a novel finding of BDNF function in the dorsal striatum, which induces vulnerability to social stress, in addition to recent research progress regarding the brain regional functions of BDNF, including the prefrontal cortex, hippocampus, and nucleus accumbens. Striatal BDNF is regulated by Shati/Nat8l, an N-acetyltransferase through epigenetic regulation. Targeting of Shati/Nat8l would allow BDNF to be striatum-specifically regulated, and the striatal Shati/Nat8l-BDNF pathway could be a promising novel therapeutic agent for the treatment of depression by modulating sensitivity to stress.
Collapse
|
45
|
Fowler CH, Bogdan R, Gaffrey MS. Stress-induced cortisol response is associated with right amygdala volume in early childhood. Neurobiol Stress 2021; 14:100329. [PMID: 33997154 PMCID: PMC8102621 DOI: 10.1016/j.ynstr.2021.100329] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 03/18/2021] [Accepted: 04/12/2021] [Indexed: 01/26/2023] Open
Abstract
Rodent research suggests that dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis and the resulting cortisol stress response can alter the structure of the hippocampus and amygdala. Because early-life changes in brain structure can produce later functional impairment and potentially increase risk for psychiatric disorder, it is critical to understand the relationship between the cortisol stress response and brain structure in early childhood. However, no study to date has characterized the concurrent association between cortisol stress response and hippocampal and amygdala volume in young children. In the present study, 42 young children (M age = 5.97, SD = 0.76), completed a frustration task and cortisol response to stress was measured. Children also underwent magnetic resonance imaging (MRI), providing structural scans from which their hippocampal and amygdala volumes were extracted. Greater cortisol stress response was associated with reduced right amygdala volume, controlling for whole brain volume, age, sex, and number of cortisol samples. There were no significant associations between cortisol stress response and bilateral hippocampus or left amygdala volumes. The association between right amygdala volume and cortisol stress response raises the non-mutually exclusive possibilities that the function of the HPA axis may shape amygdala structure and/or that amygdala structure may shape HPA axis function. As both cortisol stress response and amygdala volume have been associated with risk for psychopathology, it is possible that the relationship between cortisol stress response and amygdala volume is part of a broader pathway contributing to psychiatric risk.
Collapse
Affiliation(s)
- Carina H. Fowler
- Department of Psychology & Neuroscience, Duke University, Reuben-Cooke Building, 417 Chapel Drive, Durham, NC, 27708, USA
| | - Ryan Bogdan
- Department of Psychological and Brain Sciences, Washington University in St. Louis, Somers Family Hall, Forsyth Blvd, St. Louis, Missouri, 63105, USA
| | - Michael S. Gaffrey
- Department of Psychology & Neuroscience, Duke University, Reuben-Cooke Building, 417 Chapel Drive, Durham, NC, 27708, USA
| |
Collapse
|
46
|
Varela P, Carvalho G, Martin RP, Pesquero JB. Fabry disease: GLA deletion alters a canonical splice site in a family with neuropsychiatric manifestations. Metab Brain Dis 2021; 36:265-272. [PMID: 33156427 DOI: 10.1007/s11011-020-00640-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/30/2020] [Indexed: 11/26/2022]
Abstract
Fabry disease (FD) is a rare X-linked glycosphingolipidosis caused by mutations in GLA, a gene responsible for encoding α-galactosidase A, an enzyme required for degradation of glycosphingolipids, mainly globotriaosylceramide (Gb3) in all cells of the body. FD patients present a broad spectrum of clinical phenotype and many symptoms are shared with other diseases, making diagnosis challenging. Here we describe a novel GLA variant located in the 5' splice site of the intron 3, in four members of a family with neuropsychiatric symptoms. Analysis of the RNA showed the variant promotes alteration of the wild type donor site, affecting splicing and producing two aberrant transcripts. The functional characterization showed absence of enzymatic activity in cells expressing both transcripts, confirming their pathogenicity. The family presents mild signs of FD, as angiokeratoma, cornea verticillata, acroparesthesia, tinnitus, vertigo, as well as accumulation of plasma lyso-Gb3 and urinary Gb3. Interestingly, the man and two women present psychiatric symptoms, as depression or schizophrenia. Although psychiatric illnesses, especially depression, are frequently reported in patients with FD and studies have shown that the hippocampus is an affected brain structure in these patients, it is not clear whether the Gb3 accumulation in the brain is responsible for these symptoms or they are secondary. Therefore, new studies are needed to understand whether the accumulation of Gb3 could produce neuronal alterations leading to psychiatric symptoms.
Collapse
Affiliation(s)
- Patrícia Varela
- Center for Research and Molecular Diagnostic of Genetic Diseases - Department of Biophysics, Universidade Federal de São Paulo, Rua Pedro de Toledo, 669 - 9o andar, São Paulo, 04039-032, Brazil
| | - Gerson Carvalho
- Medical Genetics Unit, Hospital de Apoio de Brasília, Brasília, DF, Brazil
| | - Renan Paulo Martin
- Center for Research and Molecular Diagnostic of Genetic Diseases - Department of Biophysics, Universidade Federal de São Paulo, Rua Pedro de Toledo, 669 - 9o andar, São Paulo, 04039-032, Brazil
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - João Bosco Pesquero
- Center for Research and Molecular Diagnostic of Genetic Diseases - Department of Biophysics, Universidade Federal de São Paulo, Rua Pedro de Toledo, 669 - 9o andar, São Paulo, 04039-032, Brazil.
| |
Collapse
|
47
|
He Y, Yu Q, Yang T, Zhang Y, Zhang K, Jin X, Wu S, Gao X, Huang C, Cui X, Luo X. Abnormalities in Electroencephalographic Microstates Among Adolescents With First Episode Major Depressive Disorder. Front Psychiatry 2021; 12:775156. [PMID: 34975577 PMCID: PMC8718790 DOI: 10.3389/fpsyt.2021.775156] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/24/2021] [Indexed: 12/28/2022] Open
Abstract
Background: Recent studies have reported changes in the electroencephalograms (EEG) of patients with major depressive disorder (MDD). However, little research has explored EEG differences between adolescents with MDD and healthy controls, particularly EEG microstates differences. The aim of the current study was to characterize EEG microstate activity in adolescents with MDD and healthy controls (HCs). Methods: A total of 35 adolescents with MDD and 35 HCs were recruited in this study. The depressive symptoms were assessed by Hamilton Depression Scale (HAMD) and Children's Depression Inventory (CDI), and the anxiety symptoms were assessed by Chinese version of DSM-5 Level 2-Anxiety-Child scale. A 64-channel EEG was recorded for 5 min (eye closed, resting-state) and analyzed using microstate analysis. Microstate properties were compared between groups and correlated with patients' depression scores. Results: We found increased occurrence and contribution of microstate B in MDD patients compared to HCs, and decreased occurrence and contribution of microstate D in MDD patients compared to HCs. While no significant correlation between depression severity (HAMD score) and the microstate metrics (occurrence and contribution of microstate B and D) differing between MDD adolescents and HCs was found. Conclusions: Adolescents with MDD showed microstate B and microstate D changes. The obtained results may deepen our understanding of dynamic EEG changes among adolescents with MDD and provide some evidence of changes in brain development in adolescents with MDD.
Collapse
Affiliation(s)
- Yuqiong He
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qianting Yu
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Tingyu Yang
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yaru Zhang
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Kun Zhang
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xingyue Jin
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuxian Wu
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xueping Gao
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China.,Autism Center of the Second Xiangya Hospital, Central South University, Changsha, China
| | - Chunxiang Huang
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China.,Autism Center of the Second Xiangya Hospital, Central South University, Changsha, China
| | - Xilong Cui
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China.,Autism Center of the Second Xiangya Hospital, Central South University, Changsha, China
| | - Xuerong Luo
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China.,Autism Center of the Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
48
|
Validation of Chronic Restraint Stress Model in Young Adult Rats for the Study of Depression Using Longitudinal Multimodal MR Imaging. eNeuro 2020; 7:ENEURO.0113-20.2020. [PMID: 32669346 PMCID: PMC7396811 DOI: 10.1523/eneuro.0113-20.2020] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/15/2020] [Accepted: 07/03/2020] [Indexed: 12/25/2022] Open
Abstract
Prior research suggests that the neurobiological underpinnings of depression include aberrant brain functional connectivity, neurometabolite levels, and hippocampal volume. Chronic restraint stress (CRS) depression model in rats has been shown to elicit behavioral, gene expression, protein, functional connectivity, and hippocampal volume changes similar to those in human depression. However, no study to date has examined the association between behavioral changes and brain changes within the same animals. This study specifically addressed the correlation between the outcomes of behavioral tests and multiple 9.4 T magnetic resonance imaging (MRI) modalities in the CRS model using data collected longitudinally in the same animals. CRS involved placing young adult male Sprague Dawley rats in individual transparent tubes for 2.5 h daily over 13 d. Elevated plus maze (EPM) and forced swim tests (FSTs) confirmed the presence of anxiety-like and depression-like behaviors, respectively, postrestraint. Resting-state functional MRI (rs-fMRI) data revealed hypoconnectivity within the salience and interoceptive networks and hyperconnectivity of several brain regions to the cingulate cortex. Proton magnetic resonance spectroscopy revealed decreased sensorimotor cortical glutamate (Glu), glutamine (Gln), and combined Glu-Gln (Glx) levels. Volumetric analysis of T2-weighted images revealed decreased hippocampal volume. Importantly, these changes parallel those found in human depression, suggesting that the CRS rodent model has utility for translational studies and novel intervention development for depression.
Collapse
|
49
|
Lind A, Salomäki S, Parkkola R, Haataja L, Rautava P, Junttila N, Koikkalainen J, Lötjönen J, Saunavaara V, Korja R. Brain volumes in relation to loneliness and social competence in preadolescents born very preterm. Brain Behav 2020; 10:e01640. [PMID: 32333525 PMCID: PMC7303371 DOI: 10.1002/brb3.1640] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/02/2020] [Accepted: 03/28/2020] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION The aim of the present study was to assess how regional brain volumes associate with self-experienced social and emotional loneliness and social competence in very preterm and term-born preadolescents. MATERIALS AND METHODS Thirty-four very preterm subjects (birthweight ≤1,500 g and/or gestational age <32 weeks) without neurodevelopmental impairments and/or major brain pathologies and 31 term-born subjects underwent magnetic resonance imaging at 12 years of age. Regional brain volumes were measured using an automated image quantification tool. At 11 years of age, social and emotional loneliness were assessed with the Peer Network and Dyadic Loneliness Scale-self-report questionnaire and cooperating skills, empathy, impulsivity, and disruptiveness with the Multisource Assessment of Children's Social Competence Scale-self-report questionnaire. RESULTS In the very preterm group, a number of significant associations were found between smaller regional brain volumes and self-experienced emotional loneliness, more impulsivity and more disruptiveness. In the control group, brain volumes and loneliness were not associated, and brain volumes and social competence were associated with a lesser degree than in the very preterm group. CONCLUSION Experiences of emotional loneliness and poorer social competence appear to be more related to brain volumes in very preterm preadolescents than in those born full-term. It also appears that in very preterm preadolescents, emotional loneliness may be more reflected in brain development than social loneliness.
Collapse
Affiliation(s)
- Annika Lind
- Department of Psychology, University of Turku, Turku, Finland.,Turku Institute for Advanced Studies (TIAS), University of Turku, Turku, Finland
| | | | - Riitta Parkkola
- Department of Radiology, University of Turku and Turku University Hospital, Turku, Finland
| | - Leena Haataja
- Children's Hospital and Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Päivi Rautava
- Public Health, University of Turku and Clinical Research Centre of Turku University Hospital, Turku, Finland
| | - Niina Junttila
- Department of Teacher Education, University of Turku, Turku, Finland.,Finnish National Agency for Education, Helsinki, Finland
| | | | | | - Virva Saunavaara
- Division of Medical Imaging, Department of Medical Physics, Turku University Hospital, Turku, Finland.,Turku PET Center, University of Turku and Turku University Hospital, Turku, Finland
| | - Riikka Korja
- Department of Psychology, University of Turku, Turku, Finland
| | | |
Collapse
|
50
|
Schmaal L, Pozzi E, C Ho T, van Velzen LS, Veer IM, Opel N, Van Someren EJW, Han LKM, Aftanas L, Aleman A, Baune BT, Berger K, Blanken TF, Capitão L, Couvy-Duchesne B, R Cullen K, Dannlowski U, Davey C, Erwin-Grabner T, Evans J, Frodl T, Fu CHY, Godlewska B, Gotlib IH, Goya-Maldonado R, Grabe HJ, Groenewold NA, Grotegerd D, Gruber O, Gutman BA, Hall GB, Harrison BJ, Hatton SN, Hermesdorf M, Hickie IB, Hilland E, Irungu B, Jonassen R, Kelly S, Kircher T, Klimes-Dougan B, Krug A, Landrø NI, Lagopoulos J, Leerssen J, Li M, Linden DEJ, MacMaster FP, M McIntosh A, Mehler DMA, Nenadić I, Penninx BWJH, Portella MJ, Reneman L, Rentería ME, Sacchet MD, G Sämann P, Schrantee A, Sim K, Soares JC, Stein DJ, Tozzi L, van Der Wee NJA, van Tol MJ, Vermeiren R, Vives-Gilabert Y, Walter H, Walter M, Whalley HC, Wittfeld K, Whittle S, Wright MJ, Yang TT, Zarate C, Thomopoulos SI, Jahanshad N, Thompson PM, Veltman DJ. ENIGMA MDD: seven years of global neuroimaging studies of major depression through worldwide data sharing. Transl Psychiatry 2020; 10:172. [PMID: 32472038 PMCID: PMC7260219 DOI: 10.1038/s41398-020-0842-6] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 04/09/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023] Open
Abstract
A key objective in the field of translational psychiatry over the past few decades has been to identify the brain correlates of major depressive disorder (MDD). Identifying measurable indicators of brain processes associated with MDD could facilitate the detection of individuals at risk, and the development of novel treatments, the monitoring of treatment effects, and predicting who might benefit most from treatments that target specific brain mechanisms. However, despite intensive neuroimaging research towards this effort, underpowered studies and a lack of reproducible findings have hindered progress. Here, we discuss the work of the ENIGMA Major Depressive Disorder (MDD) Consortium, which was established to address issues of poor replication, unreliable results, and overestimation of effect sizes in previous studies. The ENIGMA MDD Consortium currently includes data from 45 MDD study cohorts from 14 countries across six continents. The primary aim of ENIGMA MDD is to identify structural and functional brain alterations associated with MDD that can be reliably detected and replicated across cohorts worldwide. A secondary goal is to investigate how demographic, genetic, clinical, psychological, and environmental factors affect these associations. In this review, we summarize findings of the ENIGMA MDD disease working group to date and discuss future directions. We also highlight the challenges and benefits of large-scale data sharing for mental health research.
Collapse
Affiliation(s)
- Lianne Schmaal
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia.
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia.
| | - Elena Pozzi
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Tiffany C Ho
- Department of Psychology, Stanford University, Stanford, CA, USA
- Department of Psychiatry & Behavioral Sciences, Stanford University, Stanford, CA, USA
- Department of Psychiatry & Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Laura S van Velzen
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Ilya M Veer
- Division of Mind and Brain Research, Department of Psychiatry and Psychotherapy CCM, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nils Opel
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Eus J W Van Someren
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience (NIN), an institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, VU University Amsterdam, Amsterdam, The Netherlands
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Laura K M Han
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Lybomir Aftanas
- FSSBI Scientific Research Institute of Physiology & Basic Medicine, Laboratory of Affective, Cognitive & Translational Neuroscience, Novosibirsk, Russia
- Department of Neuroscience, Novosibirsk State University, Novosibirsk, Russia
| | - André Aleman
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bernhard T Baune
- Department of Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, University of Melbourne, Melbourne, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Tessa F Blanken
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience (NIN), an institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, VU University Amsterdam, Amsterdam, The Netherlands
| | - Liliana Capitão
- Department of Psychiatry, Oxford University, Oxford, UK
- Oxford Health NHS Foundation Trust, Oxford, UK
| | | | - Kathryn R Cullen
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Udo Dannlowski
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Christopher Davey
- Department of Psychiatry, University of Melbourne, Melbourne, VIC, Australia
| | - Tracy Erwin-Grabner
- Laboratory of Systems Neuroscience and Imaging in Psychiatry (SNIP-Lab), University Medical Center Göttingen, Göttingen, Germany
| | - Jennifer Evans
- Experimental Therapeutics Branch, NIMH, NIH, Bethesda, MD, USA
| | - Thomas Frodl
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - Cynthia H Y Fu
- School of Psychology, University of East London, London, UK
- Centre for Affective Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | | | - Ian H Gotlib
- Department of Psychology, Stanford University, Stanford, CA, USA
| | - Roberto Goya-Maldonado
- Laboratory of Systems Neuroscience and Imaging in Psychiatry (SNIP-Lab), University Medical Center Göttingen, Göttingen, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Germany
| | - Nynke A Groenewold
- Department of Psychiatry & Mental Health, University of Cape Town, Cape Town, South Africa
| | | | - Oliver Gruber
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Geoffrey B Hall
- Department of Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, ON, Canada
| | - Ben J Harrison
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Melbourne, VIC, Australia
| | - Sean N Hatton
- Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - Marco Hermesdorf
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Ian B Hickie
- Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - Eva Hilland
- Clinical Neuroscience Research Group, Department of Psychology, University of Oslo, Oslo, Norway
- Department of Psychiatry, Diakonhjemmet Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Benson Irungu
- Department of Psychiatry & Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rune Jonassen
- Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
| | - Sinead Kelly
- Beth Israel Deaconess Medical Centre, Harvard Medical School, Boston, MA, USA
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | | | - Axel Krug
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Nils Inge Landrø
- Clinical Neuroscience Research Group, Department of Psychology, University of Oslo, Oslo, Norway
- Department of Psychiatry, Diakonhjemmet Hospital, Oslo, Norway
| | - Jim Lagopoulos
- Sunshine Coast Mind and Neuroscience Thompson Institute, University of the Sunshine Coast, Birtinya, QLD, Australia
| | - Jeanne Leerssen
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience (NIN), an institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, VU University Amsterdam, Amsterdam, The Netherlands
| | - Meng Li
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - David E J Linden
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
- MRC Center for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
- Cardiff University Brain Research Imaging Center, Cardiff University, Cardiff, UK
| | - Frank P MacMaster
- Psychiatry and Pediatrics, University of Calgary, Addictions and Mental Health Strategic Clinical Network, Calgary, AB, Canada
| | - Andrew M McIntosh
- Centre for Clinical Brain Science, University of Edinburgh, Edinburgh, UK
| | - David M A Mehler
- Department of Psychiatry, University of Münster, Münster, Germany
- MRC Center for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
- Cardiff University Brain Research Imaging Center, Cardiff University, Cardiff, UK
| | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Marburg University Hospital UKGM, Marburg, Germany
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Maria J Portella
- Institut d'Investigació Biomèdica-Sant Pau, Barcelona, Spain
- CIBERSAM, Madrid, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Liesbeth Reneman
- Department of Radiology and Nuclear Medicine, location AMC, Amsterdam UMC, Amsterdam, The Netherlands
| | - Miguel E Rentería
- Department of Genetics & Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Matthew D Sacchet
- Center for Depression, Anxiety, and Stress Research, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | | | - Anouk Schrantee
- Department of Radiology and Nuclear Medicine, location AMC, Amsterdam UMC, Amsterdam, The Netherlands
| | - Kang Sim
- West Region/Institute of Mental Health, Singapore, Singapore
- Yong Loo Lin School of Medicine/National University of Singapore, Singapore, Singapore
| | - Jair C Soares
- Department of Psychiatry & Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dan J Stein
- SA MRC Research Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry & Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Leonardo Tozzi
- Department of Psychiatry & Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Nic J A van Der Wee
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Institute for Brain and Cognition, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie-José van Tol
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Robert Vermeiren
- Curium-LUMC, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Henrik Walter
- Division of Mind and Brain Research, Department of Psychiatry and Psychotherapy CCM, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Jena, Germany
- Clinical Affective Neuroimaging Laboratory, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Heather C Whalley
- Centre for Clinical Brain Science, University of Edinburgh, Edinburgh, UK
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Germany
| | - Sarah Whittle
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Melbourne, VIC, Australia
| | - Margaret J Wright
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Tony T Yang
- Department of Psychiatry & Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Carlos Zarate
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, Bethesda, MD, USA
| | - Sophia I Thomopoulos
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Dick J Veltman
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| |
Collapse
|