1
|
Cui F, Liu Q, Lv X, Tian H, Wei J, Zhang K, Zhu G, Chen Q, Wang G, Wang X, Zhang N, Huang Y, Si T, Yu X. Effect of childhood trauma on cognitive function in individuals with major depressive disorder and healthy controls. J Affect Disord 2025; 371:196-204. [PMID: 39433132 DOI: 10.1016/j.jad.2024.10.059] [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: 02/06/2024] [Revised: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND Individuals with major depressive disorder (MDD) exhibit cognitive impairment, while childhood trauma (CT) is associated with an elevated risk of both MDD and cognitive dysfunction. The effect of CT on cognitive function in MDD patients and healthy controls (HCs) is unclear. METHODS MDD patients and HCs were enrolled between December 2013 and December 2016. The Childhood Trauma Questionnaire (CTQ) was used to assess CT. Depressive symptoms and cognitive function were assessed at baseline and after 8-week acute-phase treatment with selective serotonin reuptake inhibitors (SSRIs) in MDD patients. RESULTS A total of 909 people were included in the analysis. The interaction between MDD and CT had a main effect on Digit Symbol-Coding Test (DSCT), Stroop Color Test (SCT), and Stroop Color-Word Test (SCWT) scores. The effect of CT on cognitive function disappeared after adjusting for MDD diagnosis and years of education. Neglect could predict poor performance on SCT and SCWT in the HC group. After acute-phase treatment with SSRIs, CT did not significantly predict changes in cognitive function or depressive symptoms. LIMITATIONS The CTQ assessment might cause recall bias, and the cross-sectional design could not establish the causal link between CT and cognitive function. CONCLUSION The effect of CT on cognitive function was modulated by MDD diagnosis and years of education. CT did not predict changes in depressive symptoms or cognitive function after acute-phase treatment with SSRIs. The direct influence of CT on cognitive function in MDD patients may be over-estimated. TRIAL REGISTRATION ClinicalTrials.gov: NCT02023567; registration date: December 2013.
Collapse
Affiliation(s)
- Feihuan Cui
- Department of Psychology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Qi Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China; NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China.
| | - Xiaozhen Lv
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China; NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Hongjun Tian
- Nankai University Affiliated Anding Hospital, Tianjin Mental Health Center, Tianjin, China
| | - Jing Wei
- Department of Psychological Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Kerang Zhang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Gang Zhu
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Qiaoling Chen
- Department of Psychiatry, Dalian Seventh People's Hospital, Dalian, China
| | - Gang Wang
- Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Xueyi Wang
- Department of Psychiatry, The First Hospital of Hebei Medical University, Mental Health Institute of Hebei Medical University, Shijiazhuang, China
| | - Nan Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yu Huang
- National Engineering Research Center for Software Engineering, Peking University, Beijing, China
| | - Tianmei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China; NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xin Yu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China; NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| |
Collapse
|
2
|
Danielsen JT, Zachariae R, Schmidt H, Kallehauge JF, Thomadsen JK, Wefel JS, Wu LM, Amidi A. Cognitive impairment in patients with melanoma before adjuvant immune checkpoint inhibitor therapy and associations with brain gray matter, catechol-O-methyltransferase genotype, and psychological factors. Cancer 2025; 131:e35683. [PMID: 39663713 DOI: 10.1002/cncr.35683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/28/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Cancer-related cognitive impairment (CRCI) is a significant concern in patients with cancer but understanding its prevalence and risk factors in patients with malignant melanoma (MMPs) remains limited. This study explores CRCI via a multifaceted approach integrating neurobiological, genetic, and psychological assessments. METHODS Cognitive functioning across multiple domains was assessed via neuropsychological tests in 47 MMPs before adjuvant immune checkpoint inhibitor therapy, compared with 53 matched healthy controls (HCs). Self-reported cognitive complaints, brain gray matter (GM) properties, catechol-O-methyltransferase (COMT) genotype, and psychological and behavioral factors were evaluated. Between-group differences were analyzed with t-tests and χ2 tests, and associations were explored with correlation analyses. GM properties were assessed in a subset of 23 MMPs and 47 HCs. RESULTS MMPs exhibited significantly lower cognitive functioning across multiple tests (all p < .05), with a high CRCI prevalence (68.1% vs. 26.4% in HCs). MMPs reported higher fatigue, anxiety, and insomnia severity and poorer sleep quality and quality of life (all p < .01). Self-reported cognitive complaints in MMPs were associated with some cognitive test scores (all p < .05), fatigue (p < .001), and anxiety (p = .045). GM analyses revealed a smaller left cuneus volume in MMPs and significant associations between MMPs' processing speed and cortical thickness (right precentral and left inferior parietal regions) and between delayed verbal memory and right postcentral GM volume (all p < .01). CONCLUSIONS These findings underscore the need for comprehensive assessments in MMPs to better understand and address CRCI. A multifaceted approach would provide valuable insights that could inform future interventions and improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Josefine T Danielsen
- Unit for Psycho-Oncology and Health Psychology, Department of Psychology and Behavioral Sciences, Aarhus University, Aarhus, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Robert Zachariae
- Unit for Psycho-Oncology and Health Psychology, Department of Psychology and Behavioral Sciences, Aarhus University, Aarhus, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jesper F Kallehauge
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | - Julie K Thomadsen
- Unit for Psycho-Oncology and Health Psychology, Department of Psychology and Behavioral Sciences, Aarhus University, Aarhus, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jeffrey S Wefel
- Section of Neuropsychology, Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lisa M Wu
- Unit for Psycho-Oncology and Health Psychology, Department of Psychology and Behavioral Sciences, Aarhus University, Aarhus, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Department of Psychology, Reykjavik University, Reykjavik, Iceland
| | - Ali Amidi
- Unit for Psycho-Oncology and Health Psychology, Department of Psychology and Behavioral Sciences, Aarhus University, Aarhus, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
3
|
Leone G, Casanave H, Postel C, Fraisse F, Vallée T, de La Sayette V, Dayan J, Peschanski D, Eustache F, Gagnepain P. Plasticity of human resilience mechanisms. SCIENCE ADVANCES 2025; 11:eadq8336. [PMID: 39772669 PMCID: PMC11708882 DOI: 10.1126/sciadv.adq8336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025]
Abstract
The hippocampus's vulnerability to trauma-induced stress can lead to pathophysiological disturbances that precipitate the development of posttraumatic stress disorder (PTSD). The mechanisms of resilience that foster remission and mitigate the adverse effects of stress remain unknown. We analyzed the evolution of hippocampal morphology between 2016/2017 and 2018/2019, as well as the memory control mechanisms crucial for trauma resilience. Participants were individuals exposed to the 2015 Paris terrorist attacks (N = 100), including chronic (N = 34) and remitted (N = 19) PTSD, and nonexposed (N = 72). We found that normalization of inhibitory control processes, which regulate the resurgence of intrusive memories in the hippocampus, not only predicted PTSD remission but also preceded a reduction in traumatic memories. Improvement in control mechanisms was associated with the interruption of stress-induced atrophy in a hippocampal region that includes the dentate gyrus. Human resilience to trauma is characterized by the plasticity of memory control circuits, which interacts with hippocampal neuroplasticity.
Collapse
Affiliation(s)
- Giovanni Leone
- Normandie Univ, UNICAEN, PSL Research University, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, 14000 Caen, France
- Laboratory of Behavioural Neurology and Imaging of Cognition, Department of Neuroscience, Campus Biotech, University of Geneva, Geneva, Switzerland
- Swiss Center for Affective Sciences, University of Geneva, Geneva, Switzerland
| | - Hannah Casanave
- Normandie Univ, UNICAEN, PSL Research University, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, 14000 Caen, France
| | - Charlotte Postel
- Normandie Univ, UNICAEN, PSL Research University, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, 14000 Caen, France
| | - Florence Fraisse
- Normandie Univ, UNICAEN, PSL Research University, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, 14000 Caen, France
| | - Thomas Vallée
- Normandie Univ, UNICAEN, PSL Research University, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, 14000 Caen, France
| | - Vincent de La Sayette
- Normandie Univ, UNICAEN, PSL Research University, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, 14000 Caen, France
| | - Jacques Dayan
- Normandie Univ, UNICAEN, PSL Research University, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, 14000 Caen, France
- Pôle Hospitalo-Universitaire de Psychiatrie de l’Enfant et de l’Adolescent, Centre Hospitalier Guillaume Régnier, Université Rennes 1, 35700 Rennes, France
| | - Denis Peschanski
- Université Paris I Panthéon Sorbonne, HESAM Université, EHESS, CNRS, UMR8209, Paris, France
| | - Francis Eustache
- Normandie Univ, UNICAEN, PSL Research University, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, 14000 Caen, France
| | - Pierre Gagnepain
- Normandie Univ, UNICAEN, PSL Research University, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, 14000 Caen, France
| |
Collapse
|
4
|
Lee EJ, Lee MJ, Ryu YJ, Nam SH, Kim R, Song S, Park K, Park YJ, Kim JI, Koh SH, Chang MS. Neuroplasticity therapy using glia-like cells derived from human mesenchymal stem cells for the recovery of cerebral infarction sequelae. Mol Ther 2025; 33:356-374. [PMID: 39563032 DOI: 10.1016/j.ymthe.2024.11.022] [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: 04/23/2024] [Revised: 10/12/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024] Open
Abstract
Despite a dramatic increase in ischemic stroke incidence worldwide, effective therapies for attenuating sequelae of cerebral infarction are lacking. This study investigates the use of human mesenchymal stem cells (hMSCs) induced toward glia-like cells (ghMSCs) to ameliorate chronic sequelae resulting from cerebral infarction. Transcriptome analysis demonstrated that ghMSCs exhibited astrocytic characteristics, and assessments conducted ex vivo using organotypic brain slice cultures demonstrated that ghMSCs exhibited superior neuroregenerative and neuroprotective activity against ischemic damage compared to hMSCs. The observed beneficial effects of ghMSCs were diminished by pre-treatment with a CXCR2 antagonist, indicating a direct role for CXCR2 signaling. Studies conducted in rats subjected to cerebral infarction demonstrated that ghMSCs restored neurobehavioral functions and reduced chronic brain infarction in a dose-dependent manner when transplanted at the subacute-to-chronic phase. These beneficial impacts were also inhibited by a CXCR2 antagonist. Molecular analyses confirmed that increased neuroplasticity contributed to ghMSCs' neuroregenerative effects. These data indicate that ghMSCs hold promise for treating refractory sequelae resulting from cerebral infarction by enhancing neuroplasticity and identify CXCR2 signaling as an important mediator of ghMSCs' mechanism of action.
Collapse
Affiliation(s)
- Eun Ji Lee
- Department of Translational Medical Science, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul 04763, Republic of Korea
| | - Min-Ju Lee
- Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy and Dental Research Institute, Seoul National University School of Dentistry, Seoul 03080, Republic of Korea
| | - Ye Jin Ryu
- Department of Translational Medical Science, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul 04763, Republic of Korea
| | - Sang-Hyeon Nam
- Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy and Dental Research Institute, Seoul National University School of Dentistry, Seoul 03080, Republic of Korea
| | - Rokhyun Kim
- Genomic Medicine Institute (GMI), Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sehyeon Song
- Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy and Dental Research Institute, Seoul National University School of Dentistry, Seoul 03080, Republic of Korea; Interdisciplinary Program in Neuroscience, Seoul National University College of Natural Sciences, Seoul 08826, Republic of Korea
| | - Kyunghyuk Park
- Genomic Medicine Institute (GMI), Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea
| | - Young Jun Park
- Genomic Medicine Institute (GMI), Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea
| | - Jong-Il Kim
- Genomic Medicine Institute (GMI), Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Seong-Ho Koh
- Department of Translational Medical Science, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul 04763, Republic of Korea; Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Gyeonggi-do 11923, Republic of Korea.
| | - Mi-Sook Chang
- Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy and Dental Research Institute, Seoul National University School of Dentistry, Seoul 03080, Republic of Korea; Interdisciplinary Program in Neuroscience, Seoul National University College of Natural Sciences, Seoul 08826, Republic of Korea; Neuroscience Research Institute, Seoul National University, Seoul 03080, Republic of Korea.
| |
Collapse
|
5
|
Raise-Abdullahi P, Rahmani M, Tabaei NS, Rezamohammadi F, Vafaei AA, Ghanbari A, Rashidipour H, Meamar M, Rashidy-Pour A. Corticosteroid receptor antagonism in the medial prefrontal cortex reduces morphine-induced place preference and dopamine transporter expression decline in rats. Neuroscience 2025; 567:209-218. [PMID: 39793849 DOI: 10.1016/j.neuroscience.2025.01.012] [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: 08/17/2024] [Revised: 12/12/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
Corticosteroid signaling plays a critical role in modulating the neural systems underlying reward and addiction, but the specific contributions of glucocorticoid receptors (GRs) and mineralocorticoid receptors (MRs) in the medial prefrontal cortex (mPFC) to opioid reward and dopaminergic plasticity remain unclear. Here, we investigated the effects of intra-mPFC injection of corticosteroid receptor ligand (corticosterone; CORT), glucocorticoid receptor antagonist (RU38486; RU), and mineralocorticoid receptor antagonist (spironolactone; SP) on morphine-induced conditioned place preference (CPP) and dopamine transporter (DAT) expression in the mPFC. Adult male Wistar rats received intra-mPFC injections of CORT, RU, SP, or their respective vehicles prior to morphine CPP conditioning. Blockade of GRs with RU (10 or 100 ng) or MRs with a low dose of SP (10 ng) attenuated the expression of morphine CPP. Morphine reduced DAT expression in the mPFC, but RU and SP prevented this effect. These findings demonstrate that corticosteroid receptor signaling within the mPFC modulates the rewarding properties of morphine and morphine-induced dopaminergic plasticity. This preclinical study suggests that targeting GRs and MRs in the mPFC could be a possible therapeutic approach for treating opioid addiction. By targeting these receptors, it may be possible to reduce opioid reward and counteract the neuroadaptations in dopamine systems associated with addiction.
Collapse
Affiliation(s)
| | - Mehrnoush Rahmani
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Narges Sadat Tabaei
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Abbas Ali Vafaei
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Ghanbari
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Hamed Rashidipour
- College of International Education, Dalian Medical University, Dalian, China
| | - Morvarid Meamar
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Clinical Research Development Unit, Kowsar Educational Research and Therapeutic Hospital, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| |
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 2025:1-9. [PMID: 39773537 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] [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
|
Shang G, Zhou T, Yan X, He K, Liu B, Feng Z, Xu J, Yu X, Zhang Y. Multi-scale Analysis Reveals Hippocampal Subfield Vulnerabilities to Chronic Cortisol Overexposure: Evidence from Cushing's Disease. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2025:S2451-9022(25)00014-X. [PMID: 39793703 DOI: 10.1016/j.bpsc.2024.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/05/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025]
Abstract
BACKGROUND Chronic cortisol overexposure plays a significant role in the development of neuropathological changes associated with neuropsychiatric and neurodegenerative disorders. The hippocampus, the primary target of cortisol, may exhibit characteristic regional responses due to its internal heterogeneity. This study explores structural and functional alterations of hippocampal subfields in Cushing's disease (CD), an endogenous model of chronic cortisol overexposure. METHODS Utilizing structural and resting-state functional magnetic resonance imaging data from 169 participants (86 CD patients and 83 healthy controls) recruited from a single center, we investigated specific structural changes in hippocampal subfields and explored the functional connectivity alterations driven by these structural abnormalities. We also analyzed potential associative mechanisms between these changes and biological attributes, neuropsychiatric representations, cognitive function, and gene expression profiles. RESULTS Compared to healthy controls, CD patients exhibited significant bilateral volume reductions in multiple hippocampal subfields. Notably, volumetric decreases in the left hippocampal body and tail subfields were significantly correlated with cortisol levels, Montreal Cognitive Assessment scores, and quality of life measures. Disrupted connectivity between the structurally abnormal hippocampal subfields and ventromedial prefrontal cortex may impair reward-based decision making and emotional regulation, with this dysconnectivity linked to structural changes in right hippocampal subfields. Additionally, another region exhibiting dysconnectivity was located in the left pallidum and putamen. Gene expression patterns associated with synaptic components may underlie these macrostructural alterations. CONCLUSIONS Our findings elucidate the subfield-specific effects of chronic cortisol overexposure on the hippocampus, enhancing understanding of shared neuropathological traits linked to cortisol dysregulation in neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Guosong Shang
- Department of Neurosurgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, China; Chinese PLA Medical School, Beijing, China
| | - Tao Zhou
- Department of Neurosurgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, China; Neurosurgery Institute, Chinese PLA General Hospital, Beijing, PR China
| | - Xinyuan Yan
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Kunyu He
- Department of Neurosurgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, China; Chinese PLA Medical School, Beijing, China
| | - Bin Liu
- Department of Neurosurgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, China; Chinese PLA Medical School, Beijing, China
| | - Zhebin Feng
- Department of Neurosurgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, China; Chinese PLA Medical School, Beijing, China
| | - Junpeng Xu
- Department of Neurosurgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, China; Chinese PLA Medical School, Beijing, China
| | - Xinguang Yu
- Department of Neurosurgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, China; Chinese PLA Medical School, Beijing, China; Neurosurgery Institute, Chinese PLA General Hospital, Beijing, PR China.
| | - Yanyang Zhang
- Department of Neurosurgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, China; Neurosurgery Institute, Chinese PLA General Hospital, Beijing, PR China.
| |
Collapse
|
8
|
Hamilton HK, Roach BJ, Bachman PM, Belger A, Carrión RE, Duncan E, Johannesen JK, Light GA, Niznikiewicz MA, Addington J, Bearden CE, Cadenhead KS, Cornblatt BA, Perkins DO, Tsuang MT, Walker EF, Woods SW, Cannon TD, Mathalon DH. Mismatch Negativity as an Index of Auditory Short-Term Plasticity: Associations with Cortisol, Inflammation, and Gray Matter Volume in Youth at Clinical High Risk for Psychosis. Clin EEG Neurosci 2025; 56:46-59. [PMID: 39552576 DOI: 10.1177/15500594241294035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Mismatch negativity (MMN) event-related potential (ERP) component reduction, indexing N-methyl-D-aspartate receptor (NMDAR)-dependent auditory echoic memory and short-term plasticity, is a well-established biomarker of schizophrenia that is sensitive to psychosis risk among individuals at clinical high-risk (CHR-P). Based on the NMDAR-hypofunction model of schizophrenia, NMDAR-dependent plasticity is predicted to contribute to aberrant neurodevelopmental processes involved in the pathogenesis of schizophrenia during late adolescence or young adulthood, including gray matter loss. Moreover, stress and inflammation disrupt plasticity. Therefore, using data collected during the 8-center North American Prodrome Longitudinal Study (NAPLS-2), we explored relationships between MMN amplitudes and salivary cortisol, gray matter volumes, and inflammatory cytokines. Participants included 303 CHR-P individuals with baseline electroencephalography (EEG) data recorded during an MMN paradigm as well as structural magnetic resonance imaging (MRI) and salivary cortisol, of which a subsample (n = 57) also completed blood draws. More deficient MMN amplitudes were associated with greater salivary cortisol and pro-inflammatory cytokine levels in future CHR-Converters, but not among those who did not convert to psychosis within the next two years. More deficient MMN amplitude was also associated with smaller total gray matter volume across participants regardless of future clinical outcomes, and with subcortical gray matter volumes among future CHR-Converters only. These findings are consistent with the theory that deficient NMDAR-dependent plasticity results in an overabundance of weak synapses that are subject to over-pruning during psychosis onset, contributing to gray matter loss. Further, MMN plasticity mechanisms may interact with stress, cortisol, and neuroinflammatory processes, representing a proximal influence of psychosis.
Collapse
Affiliation(s)
- Holly K Hamilton
- Mental Health Service, Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
- Department of Psychiatry & Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Brian J Roach
- Mental Health Service, San Francisco Veterans Affairs Health Care System, San Francisco, CA, USA
| | - Peter M Bachman
- Department of Psychiatry, Boston Children's Hospital, Boston, MA, USA
| | - Aysenil Belger
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ricardo E Carrión
- Division of Psychiatry Research, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY, USA
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, North Shore-Long Island Jewish Health System, Manhasset, NY, USA
| | - Erica Duncan
- Mental Health Service, Atlanta Veterans Affairs Health Care System, Decatur, GA, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Jason K Johannesen
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, USA
| | - Gregory A Light
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Mental Health Service, Veterans Affairs San Diego Health Care System, La Jolla, CA, USA
| | - Margaret A Niznikiewicz
- Department of Psychiatry, Harvard Medical School at Beth Israel Deaconess Medical Center and Massachusetts General Hospital, Boston, MA, USA
- Mental Health Service, Veterans Affairs Boston Health Care System, Brockton, MA, USA
| | - Jean Addington
- Hotchkiss Brain Institute Department of Psychiatry, University of Calgary, Calgary, Alberta, Canada
| | - Carrie E Bearden
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
- Department of Psychology, University of California, Los Angeles, CA, USA
| | - Kristin S Cadenhead
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Barbara A Cornblatt
- Division of Psychiatry Research, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY, USA
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, North Shore-Long Island Jewish Health System, Manhasset, NY, USA
- Department of Molecular Medicine, Hofstra North Shore-LIJ School of Medicine, Hempstead, NY, USA
| | - Diana O Perkins
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ming T Tsuang
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Elaine F Walker
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Department of Psychology, Emory University, Atlanta, GA, USA
| | - Scott W Woods
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, USA
| | - Tyrone D Cannon
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, USA
- Department of Psychology, Yale University, School of Medicine, New Haven, CT, USA
| | - Daniel H Mathalon
- Department of Psychiatry & Behavioral Sciences, University of California, San Francisco, CA, USA
- Mental Health Service, San Francisco Veterans Affairs Health Care System, San Francisco, CA, USA
| |
Collapse
|
9
|
Begni V, Silipo DM, Bottanelli C, Papp M, Cattaneo A, Riva MA. Chronic treatment with the antipsychotic lurasidone modulates the neuroinflammatory changes associated with the vulnerability to chronic mild stress exposure in female rats. Brain Behav Immun 2025; 123:586-596. [PMID: 39384053 DOI: 10.1016/j.bbi.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/12/2024] [Accepted: 10/05/2024] [Indexed: 10/11/2024] Open
Abstract
Stress exposure is a key risk factor for the developmentof depressive-like conditions. However, despite the higher incidence of Major Depressive Disorder in the female population, classical stress-based experimental paradigms have primarily focused on males. In the present study, we used the well-established chronic mild stress (CMS) paradigm to investigate the development of anhedonia, a cardinal symptom of affective disorders, in the female animals and we also studied the potential effect of the antipsychotic drug lurasidone in normalizing the alterations brought about by stress exposure. We found that three weeks of CMS exposure produced a significant reduction of sucrose intake in 50% of the animals (vulnerable, CMS-V), whereas the others were resilient (CMS-R). The development of an anhedonic phenotype in CMS-V was associated with a significant elevation of different immune markers, such as Complement C3 and C4, and inflammatory cytokines, including INFß and Il1ß in dorsal and ventral hippocampus. Interestingly, sub-chronic treatment with the antipsychotic drug lurasidone was able to revert the anhedonic phenotype while normalizing most of the molecular alterations found in rats vulnerable to CMS exposure. This study extends the ability of lurasidone to normalize the anhedonic phenotype in CMS rats also to females. Moreover, we provide novel evidence on lurasidone's potential effectiveness in treating mental disorders characterized by immune-inflammatory dysfunction.
Collapse
Affiliation(s)
- Veronica Begni
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | - Diana Morena Silipo
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | - Chiara Bottanelli
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | - Mariusz Papp
- Maj Institute of Pharmacology Polish Academy of Sciences, Smetna Street 12, Krakow 31-343, Poland
| | - Annamaria Cattaneo
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Via Balzaretti 9, 20133 Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Via Pilastroni 4, 25125 Brescia, Italy
| | - Marco Andrea Riva
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Via Balzaretti 9, 20133 Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Via Pilastroni 4, 25125 Brescia, Italy.
| |
Collapse
|
10
|
Bishop CL, Lean RE, Smyser TA, Smyser CD, Rogers CE. Adverse Childhood Experiences and Socioemotional Outcomes of Children Born Very Preterm. J Pediatr 2025; 276:114377. [PMID: 39442792 DOI: 10.1016/j.jpeds.2024.114377] [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: 04/22/2024] [Revised: 09/30/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE To examine whether adverse childhood experiences (ACEs) confer risk for socioemotional problems in children born very preterm (VPT). STUDY DESIGN As part of a longitudinal study, 96 infants born VPT at 23-30 weeks of gestation were recruited from a level III neonatal intensive care unit and underwent follow-up at ages 2 and 5 years. Eighty-three full-term (FT) (37-41 weeks gestation) children were recruited from an adjoining obstetric service and the local community. ACEs were assessed with the Child Life Events Scale at age 2 and Preschool Age Psychiatric Assessment at age 5. At age 5, internalizing, externalizing, and attention deficit hyperactivity disorder (ADHD) symptoms were assessed with the Child Behavior Checklist and Conner's Rating Scale-Revised, respectively. Covariates including socioeconomic disadvantage, maternal distress, and parent ADHD symptoms were assessed at the 2- and/or 5-year follow-up. Mediation and moderation analysis, accounting for family clustering, examined associations between birth group, ACEs, and socioemotional outcomes. RESULTS After covariate adjustment, children born VPT experienced more ACEs (P < .001), particularly medical ACEs (P < .01), and had worse ADHD and internalizing outcomes (P < .05) than full-term children. ACEs mediated the association between birth group and ADHD outcomes (95% CI, 0.11-4.08). There was no evidence of mediation for internalizing outcomes. Higher parent ADHD symptoms (P < .001) and maternal distress (P < .05) were associated with poorer internalizing outcomes. CONCLUSIONS Screening for childhood ACEs should be embedded in the follow-up care of children born VPT and their families. Strategies to screen for and address parent psychosocial functioning may be important to support children's socioemotional development.
Collapse
Affiliation(s)
- Callie L Bishop
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO
| | - Rachel E Lean
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO.
| | - Tara A Smyser
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO
| | - Christopher D Smyser
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO; Department of Radiology, Washington University School of Medicine, St. Louis, MO; Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Cynthia E Rogers
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
11
|
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
|
12
|
Finlay S, Adegboye O, McDermott B, Rudd D, Sarnyai Z. Linking childhood allostatic load, early adversity and the emergence of mental health symptoms in early adulthood: Analysis of the ALSPAC longitudinal birth cohort. Psychoneuroendocrinology 2024; 172:107276. [PMID: 39787866 DOI: 10.1016/j.psyneuen.2024.107276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/28/2024] [Accepted: 12/29/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND It has been well-established that the allostatic load (AL) index, a cumulative score of multi-system dysregulation in response to chronic stress, is significantly increased at the time of a psychiatric diagnosis. However, no studies have investigated if there is an association between the AL index in childhood and the later development of mental health symptoms in young adults. METHODS Using data from the Avon Longitudinal Study of Parents and Children (ALSPAC), a population cohort from Bristol, United Kingdom, we investigated the AL index at age 9 years and the risks for mental health symptoms at age 24 years. We used multinomial logistic regression analysis to investigate the association between AL threshold (categorised into bottom third: AL index ≤ 7, middle third: AL index = 7.1-9.9, and top third: AL index ≥ 10) and mental health symptoms while adjusting for sex, the age of mother at delivery, and social class. We used a relative risk ratio (RRR) and 95 % confidence interval(CI) for each variable. We further investigated the association between adverse childhood experiences (ACEs) and mental health symptoms. RESULTS We identified a significant association between sex and mental health symptoms, with more females (59 % vs 41 %) showing mental health symptoms than males. We found no direct association between the AL index at age 9 and the later development of mental health symptoms. However, an RRR analysis showed that individuals in the middle and the top third of the AL index had an RRR of 1.99 and 2.20, respectively, to develop mental health symptoms if they were females. We found that individuals who experienced ACE had a much higher risk of developing mental health symptoms as young adults, with the adjusted RRR of 5.39 (95 % CI: 3.00;9.67), 6.79 (95 % CI: 2.55; 18.1), and 2.10 (95 % CI: 0.37;11.8) for neglect, physical and sexual abuse, respectively, in individuals with mood disorder symptoms. The adjusted RRR for neglect and physical and sexual abuse in individuals with psychotic symptoms was 0.99 (95 % CI: 0.37; 2.59), 2.92 (95 % CI: 0.35; 24.4), and 10.5 (95 % CI: 0.99; 112), respectively. CONCLUSION Although the AL index in childhood was not directly associated with the later development of psychotic and mood disorder symptoms in this cohort, females in the higher tertiles of the AL index measured at 9 years of age had an elevated risk of mental health symptoms as young adults. In line with previous work, a strong association was identified between childhood adversity and mental health symptoms in young adulthood. These results highlight the importance of considering the impact of early stress on biological embedding and the later emergence of mental health problems, especially in females.
Collapse
Affiliation(s)
- Sabine Finlay
- Laboratory of Psychiatric Neuroscience, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia; College of Medicine and Dentistry, James Cook University, Queensland, Australia
| | - Oyelola Adegboye
- Menzies, School of Health Research, Charles Darwin University, Casuarina, Northern Territory, Australia
| | - Brett McDermott
- Child and Adolescent Mental Health Service, Hobart, Tasmania, Australia; Tasmanian Centre for Mental Health Service Innovation, Hobart, Tasmania, Australia
| | - Donna Rudd
- College of Medicine and Dentistry, James Cook University, Queensland, Australia
| | - Zoltán Sarnyai
- Laboratory of Psychiatric Neuroscience, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia; College of Medicine and Dentistry, James Cook University, Queensland, Australia.
| |
Collapse
|
13
|
Xie X, Wu F, Zhuang Z, Xu A. The bidirectional relationships between social isolation and cognitive function among older adults in China: separating between-person effects from within-person effects. Sci Rep 2024; 14:31832. [PMID: 39738602 DOI: 10.1038/s41598-024-83130-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025] Open
Abstract
This study investigates the bidirectional relationship between social isolation and cognitive function among older adults in China, utilizing data from the Chinese Longitudinal Healthy Longevity Survey (CLHLS). The baseline survey, which commenced in 2008, tracked 1,662 participants over four waves of data collection spanning a decade. We employed Cross-Lagged Panel Models (CLPM) and Random Intercept Cross-Lagged Panel Models (RI-CLPM) to analyze these relationships at both the between-person and within-person levels. CLPM results indicated significant cross-lagged effects between social isolation (β = - 0.119, p < 0.001; β = - 0.162, p < 0.001) and cognitive function (β = - 0.073, p < 0.001; β = - 0.091, p < 0.001) at the between-person level over the last three waves. Specifically, higher prior levels of social isolation were associated with a significant decline in subsequent cognitive function, and vice versa. Furthermore, RI-CLPM results showed that, after controlling for random intercepts and covariates, only social isolation had a significant negative impact on cognitive function across all waves (β = - 0.051, p < 0.05; β = - 0.047, p < 0.05; β = - 0.028, p < 0.05). Overall, this study demonstrates that, when considering both between-person and within-person effects, social isolation exerts a stronger lag effect on cognitive function among older adults in China. This suggests that, over a specific timeframe, reducing social isolation is crucial for promoting healthy aging in this population.
Collapse
Affiliation(s)
- Xinlong Xie
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing, China
| | - Fanfan Wu
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhiruo Zhuang
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing, China
| | - Aijun Xu
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing, China.
- Jiangsu Research Center for Major Health Risk Management and TCM Control Policy, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
14
|
Coleman EM, White M, Antonoudiou P, Weiss GL, Scarpa G, Stone B, Maguire J. Early life stress influences epilepsy outcomes in mice. Epilepsy Behav 2024; 163:110217. [PMID: 39689578 DOI: 10.1016/j.yebeh.2024.110217] [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: 09/18/2024] [Revised: 10/21/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024]
Abstract
Stress is a common seizure trigger that has been implicated in worsening epilepsy outcomes, which encompasses psychiatric and cognitive comorbidities and sudden unexpected death in epilepsy (SUDEP) risk. The neuroendocrine response to stress is mediated by the hypothalamic-pituitary-adrenal (HPA) axis and HPA axis dysfunction worsens epilepsy outcomes, increasing seizure burden, behavioral comorbidities, and risk for SUDEP in mice. Early life stress (ELS) reprograms the HPA axis into adulthood, impacting both the basal and stress-induced activity. Thus, we propose that ELS may influence epilepsy outcomes by influencing the function of the HPA axis. To test this hypothesis, we utilized the maternal separation paradigm and examined the impact on seizure susceptibility. We show that ELS exerts a sex dependent effect on seizure susceptibility in response to acute administration of the chemoconvulsant, kainic acid, which is associated with an altered relationship between seizure activity and HPA axis function. To further examine the impact of ELS on epilepsy outcomes, we utilized the intrahippocampal kainic acid model of chronic epilepsy in mice previously exposed to maternal separation. We find that the relationship between corticosterone levels and the extent of epileptiform activity is altered in mice subjected to ELS. We demonstrate that ELS impacts behavioral outcomes associated with chronic epilepsy in a sex-dependent manner, with females being more affected. We also observe reduced mortality (presumed SUDEP) in female mice subjected to ELS, consistent with previous findings suggesting a role for HPA axis dysfunction in SUDEP risk. These data demonstrate for the first time that ELS influences epilepsy outcomes and suggest that previous life experiences may impact the trajectory of epilepsy.
Collapse
Affiliation(s)
- Emanuel M Coleman
- Tufts University School of Medicine, Neuroscience Department, Boston, MA, USA
| | - Maya White
- Tufts University School of Medicine, Neuroscience Department, Boston, MA, USA
| | | | - Grant L Weiss
- Tufts University School of Medicine, Neuroscience Department, Boston, MA, USA
| | - Garrett Scarpa
- Tufts University School of Medicine, Neuroscience Department, Boston, MA, USA
| | - Bradly Stone
- Tufts University School of Medicine, Neuroscience Department, Boston, MA, USA
| | - Jamie Maguire
- Tufts University School of Medicine, Neuroscience Department, Boston, MA, USA.
| |
Collapse
|
15
|
Trousselard M. Neurophysiology of adaptative and maladaptive stress: Relations with psychology of stress. Neurophysiol Clin 2024; 55:103036. [PMID: 39674057 DOI: 10.1016/j.neucli.2024.103036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/01/2024] [Accepted: 12/01/2024] [Indexed: 12/16/2024] Open
Abstract
OBJECTIVES The stress reaction is an integrated response to a change in the environment that enables each individual to adapt to demand. While this response is physiologically coordinated by the brain, its phenomenology is expressed in the field of psychology and psychopathology. This interrelation between neurophysiological mechanisms and psychological processes is complex as dynamic interpersonal, biological, and psychocognitive systems interact with contextual and environmental factors to shape adaptation over the life constraints. METHOD This article aims to present the actors of the adjusted stress response, such as coping and coping flexibility, mindfulness and resilience, and their respective neurophysiology. RESULTS A model of the relationship between resilience, mindfulness and coping was proposed for optimizing adaptation to stress response. DISCUSSION These focuses are prerequisites for understanding and supporting human adaptation in the everyday environment and promoting efficient management of stress for mental and physical health.
Collapse
Affiliation(s)
- Marion Trousselard
- Institut de Recherche Biomédicale des Armées; Brétigny-sur-Orge, France; University of Lorraine, Inserm, INSPIIRE, F-54000, Nancy, France; UMR7268, University of Aix-Marseille, Marseille, France.
| |
Collapse
|
16
|
Mazaheri M, Radahmadi M, Sharifi MR. Effects of chronic empathic stress on synaptic efficacy, as well as short-term and long-term plasticity at the Schaffer collateral/commissural- CA1 synapses in the dorsal hippocampus of rats. Metab Brain Dis 2024; 40:54. [PMID: 39636524 DOI: 10.1007/s11011-024-01487-5] [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: 09/13/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
Empathy, the ability to comprehend and share others' emotional states, impacts brain functions. This in vivo electrophysiological study explored the influence of chronic empathic stress on synaptic efficacy, as well as short-term and long-term plasticity at the Schaffer collateral/Commissural - CA1 synapses in the dorsal hippocampus of rats, in situations of social equality and inequality. Forty-eight male rats were randomized into six groups: control, pseudo-observer, pseudo-demonstrator, observer, demonstrator, and co-demonstrator (Co, Pse-Ob, Pse-De, Ob, De, Co-De) groups. Stress induction (2h/day, 21 days) was performed in situations of equality and inequality. Serum corticosterone levels, slope, amplitude, and area under the curve (AUC) of field excitatory postsynaptic potentials (fEPSPs) were assessed in the hippocampal CA1 area using input-output (I/O) functions, paired-pulse (PP) responses with different interpulse intervals (IPIs), and long-term potentiation (LTP) after high-frequency stimulation (HFS). The fEPSP slope, amplitude, and AUC significantly decreased in all stress groups, especially in the De and Pse-De groups. These parameters were significantly increased in the Co-De and Ob groups compared to the De group. Notably, the corticosterone levels strongly confirmed the electrophysiological findings. Chronic empathic stress could disrupt synaptic efficacy and plasticity in the CA1 area. Empathic stress, involving the presence of cagemates in situations of social equality and inequality, can modify long-term plasticity and serum corticosterone levels in demonstrators and co-demonstrators. Under empathic stress related to situations of inequality, freely moving observers may influence the demonstrators' stress experience. Therefore, the presence of a conspecific in the social inequality conditions had significant suppressive effects on long-term plasticity, while conversely, under equality conditions, long-term plasticity was favorably improved through social buffering.
Collapse
Affiliation(s)
- Mohammad Mazaheri
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Radahmadi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohammad Reza Sharifi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
17
|
Okuda Y, Li D, Maruyama Y, Sonobe H, Mano T, Tainaka K, Shinohara R, Furuyashiki T. The activation of the piriform cortex to lateral septum pathway during chronic social defeat stress is crucial for the induction of behavioral disturbance in mice. Neuropsychopharmacology 2024:10.1038/s41386-024-02034-7. [PMID: 39638863 DOI: 10.1038/s41386-024-02034-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/17/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024]
Abstract
Chronic stress induces neural dysfunctions and risks mental illnesses. Clinical and preclinical studies have established the roles of brain regions underlying emotional and cognitive functions in stress and depression. However, neural pathways to perceive sensory stimuli as stress to cause behavioral disturbance remain unknown. Using whole-brain imaging of Arc-dVenus neuronal response reporter mice and machine learning analysis, here we unbiasedly demonstrated different patterns of contribution of widely distributed brain regions to neural responses to acute and chronic social defeat stress (SDS). Among these brain regions, multiple sensory cortices, especially the piriform (olfactory) cortex, primarily contributed to classifying neural responses to chronic SDS. Indeed, SDS-induced activation of the piriform cortex was augmented with repetition of SDS, accompanied by impaired odor discrimination. Axonal tracing and chemogenetic manipulation showed that excitatory neurons in the piriform cortex directly project to the lateral septum and activate it in response to chronic SDS, thereby inducing behavioral disturbance. These results pave the way for identifying a spatially defined sequence of neural consequences of stress and the roles of sensory pathways in perceiving chronic stress in mental illness pathology.
Collapse
Affiliation(s)
- Yuki Okuda
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Dongrui Li
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Yuzuki Maruyama
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Hirokazu Sonobe
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Tomoyuki Mano
- Computational Neuroethology Unit, Okinawa Institute of Science and Technology (OIST) Graduate University, Okinawa, 904-0412, Japan
| | - Kazuki Tainaka
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Ryota Shinohara
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan.
| | - Tomoyuki Furuyashiki
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan.
| |
Collapse
|
18
|
Thomou C, Nussbaumer M, Grammenou E, Komini C, Vlaikou AM, Papageorgiou MP, Filiou MD. Early Handling Exerts Anxiolytic Effects and Alters Brain Mitochondrial Dynamics in Adult High Anxiety Mice. Mol Neurobiol 2024; 61:10593-10612. [PMID: 38761326 PMCID: PMC11584496 DOI: 10.1007/s12035-024-04116-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 03/09/2024] [Indexed: 05/20/2024]
Abstract
Early handling (EH), the brief separation of pups from their mother during early life, has been shown to exert beneficial effects. However, the impact of EH in a high anxiety background as well as the role of brain mitochondria in shaping EH-driven responses remain elusive.Here, we used a high (HAB) vs. normal (NAB) anxiety-related behavior mouse model to study how EH affects pup and dam behavior in divergent anxiety backgrounds. We also investigated EH-induced effects at the protein and mRNA levels in adult male HAB mice in the hypothalamus, the prefrontal cortex, and the hippocampus by examining the same mitochondrial/energy pathways and mitochondrial dynamics mechanisms (fission, fusion, biogenesis, and mitophagy) in all three brain regions.EH exerts anxiolytic effects in adult HAB but not NAB male mice and does not affect HAB or NAB maternal behavior, although basal HAB vs. NAB maternal behaviors differ. In adult HAB male mice, EH does not impact oxidative phosphorylation (OXPHOS) and oxidative stress in any of the brain regions studied but leads to increased protein expression of glycolysis enzymes and a correlation of anxiety-related behavior with Krebs cycle enzymes in HAB mice in the hypothalamus. Intriguingly, EH alters mitochondrial dynamics by increasing hypothalamic DRP1, OPA1, and PGC1a protein levels. At the mRNA level, we observe altered, EH-driven mitochondrial dynamics mRNA signatures which predominantly affect the prefrontal cortex.Taken together, our results show that EH exerts anxiolytic effects in adulthood in high anxiety and modulates mitochondrial dynamics pathways in a brain region-specific manner.
Collapse
Affiliation(s)
- Christina Thomou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| | - Markus Nussbaumer
- Laboratory of Biochemistry, Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| | - Eleni Grammenou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| | - Chrysoula Komini
- Laboratory of Biochemistry, Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| | - Angeliki-Maria Vlaikou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| | - Maria P Papageorgiou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| | - Michaela D Filiou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece.
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece.
- Institute of Biosciences, University of Ioannina, Ioannina, Greece.
| |
Collapse
|
19
|
Page CE, Epperson CN, Novick AM, Duffy KA, Thompson SM. Beyond the serotonin deficit hypothesis: communicating a neuroplasticity framework of major depressive disorder. Mol Psychiatry 2024; 29:3802-3813. [PMID: 38816586 PMCID: PMC11692567 DOI: 10.1038/s41380-024-02625-2] [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: 11/17/2023] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
The serotonin deficit hypothesis explanation for major depressive disorder (MDD) has persisted among clinicians and the general public alike despite insufficient supporting evidence. To combat rising mental health crises and eroding public trust in science and medicine, researchers and clinicians must be able to communicate to patients and the public an updated framework of MDD: one that is (1) accessible to a general audience, (2) accurately integrates current evidence about the efficacy of conventional serotonergic antidepressants with broader and deeper understandings of pathophysiology and treatment, and (3) capable of accommodating new evidence. In this article, we summarize a framework for the pathophysiology and treatment of MDD that is informed by clinical and preclinical research in psychiatry and neuroscience. First, we discuss how MDD can be understood as inflexibility in cognitive and emotional brain circuits that involves a persistent negativity bias. Second, we discuss how effective treatments for MDD enhance mechanisms of neuroplasticity-including via serotonergic interventions-to restore synaptic, network, and behavioral function in ways that facilitate adaptive cognitive and emotional processing. These treatments include typical monoaminergic antidepressants, novel antidepressants like ketamine and psychedelics, and psychotherapy and neuromodulation techniques. At the end of the article, we discuss this framework from the perspective of effective science communication and provide useful language and metaphors for researchers, clinicians, and other professionals discussing MDD with a general or patient audience.
Collapse
Affiliation(s)
- Chloe E Page
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - C Neill Epperson
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Family Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Helen and Arthur E. Johnson Depression Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew M Novick
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Korrina A Duffy
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Scott M Thompson
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
20
|
Grey DK, Purcell JB, Buford KN, Schuster MA, Elliott MN, Emery ST, Mrug S, Knight DC. Discrimination Exposure, Neural Reactivity to Stress, and Psychological Distress. Am J Psychiatry 2024; 181:1112-1126. [PMID: 39473266 DOI: 10.1176/appi.ajp.20220884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2024]
Abstract
OBJECTIVE Discrimination exposure has a detrimental impact on mental health, increasing the risk of depression, anxiety, and posttraumatic stress. The impact discrimination exposure has on mental health is likely mediated by neural processes associated with emotion expression and regulation. However, the specific neural processes that mediate the relationship between discrimination exposure and mental health remain to be determined. The present study investigated the relationship adolescent discrimination exposure has with stress-elicited brain activity and mental health symptoms in young adulthood. METHODS A total of 301 participants completed the Montreal Imaging Stress Task while functional MRI data were collected. Discrimination exposure was measured four times from ages 11 to 19, and stress-elicited brain activity and psychological distress (depression, anxiety, posttraumatic stress) were assessed in young adulthood (age 20). RESULTS Stress-elicited dorsolateral and dorsomedial prefrontal cortex (PFC), inferior parietal lobule (IPL), and hippocampal activity varied with discrimination exposure. Activity within these brain regions varied with the cumulative amount and trajectory of discrimination exposure across adolescence (initial exposure, change in exposure, and acceleration of exposure). Depression, anxiety, and posttraumatic stress symptoms varied with discrimination exposure. Stress-elicited activity within the dorsolateral PFC and the IPL statistically mediated the relationship between discrimination exposure and psychological distress. CONCLUSIONS The findings suggest that adolescent discrimination exposure may alter the neural response to future stressors (i.e., within regions associated with emotion expression and regulation), which may in turn modify susceptibility and resilience to psychological distress. Thus, differences in stress-elicited neural reactivity may represent an important neurobiological mechanism underlying discrimination-related mental health disparities.
Collapse
Affiliation(s)
- Devon K Grey
- Department of Psychology, University of Alabama at Birmingham (Grey, Purcell, Buford, Mrug, Knight); Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA (Schuster); RAND Corporation, Santa Monica, CA (Elliott); UTHealth Houston School of Public Health, Houston (Emery)
| | - Juliann B Purcell
- Department of Psychology, University of Alabama at Birmingham (Grey, Purcell, Buford, Mrug, Knight); Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA (Schuster); RAND Corporation, Santa Monica, CA (Elliott); UTHealth Houston School of Public Health, Houston (Emery)
| | - Kristen N Buford
- Department of Psychology, University of Alabama at Birmingham (Grey, Purcell, Buford, Mrug, Knight); Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA (Schuster); RAND Corporation, Santa Monica, CA (Elliott); UTHealth Houston School of Public Health, Houston (Emery)
| | - Mark A Schuster
- Department of Psychology, University of Alabama at Birmingham (Grey, Purcell, Buford, Mrug, Knight); Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA (Schuster); RAND Corporation, Santa Monica, CA (Elliott); UTHealth Houston School of Public Health, Houston (Emery)
| | - Marc N Elliott
- Department of Psychology, University of Alabama at Birmingham (Grey, Purcell, Buford, Mrug, Knight); Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA (Schuster); RAND Corporation, Santa Monica, CA (Elliott); UTHealth Houston School of Public Health, Houston (Emery)
| | - Susan Tortolero Emery
- Department of Psychology, University of Alabama at Birmingham (Grey, Purcell, Buford, Mrug, Knight); Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA (Schuster); RAND Corporation, Santa Monica, CA (Elliott); UTHealth Houston School of Public Health, Houston (Emery)
| | - Sylvie Mrug
- Department of Psychology, University of Alabama at Birmingham (Grey, Purcell, Buford, Mrug, Knight); Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA (Schuster); RAND Corporation, Santa Monica, CA (Elliott); UTHealth Houston School of Public Health, Houston (Emery)
| | - David C Knight
- Department of Psychology, University of Alabama at Birmingham (Grey, Purcell, Buford, Mrug, Knight); Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA (Schuster); RAND Corporation, Santa Monica, CA (Elliott); UTHealth Houston School of Public Health, Houston (Emery)
| |
Collapse
|
21
|
Rodriguez P, López-Landa A, Romo-Parra H, Rubio-Osornio M, Rubio C. Unraveling the ozone impact and oxidative stress on the nervous system. Toxicology 2024; 509:153973. [PMID: 39423999 DOI: 10.1016/j.tox.2024.153973] [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: 09/13/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Ozone (O₃), a potent oxidant, can penetrate the body through breathing, generating reactive oxygen species (ROS) and triggering inflammatory processes. Oxidative stress, an imbalance between the production of ROS and the body's antioxidant capacity, plays a crucial role in the pathophysiology of various neurodegenerative diseases. This phenomenon can negatively impact the Central Nervous System (CNS), inducing structural and functional alterations that contribute to the development of neurological pathologies. This review examines how O₃-induced oxidative stress affects the nervous system by analyzing existing literature on the involved molecular mechanisms and potential antioxidant systems to mitigate its effects. Through a comprehensive review of experimental studies, our objective is to shed light on the interaction between O₃ and the nervous system, as well as its signaling pathways and altered genes, providing a foundation for future research in this field. Several studies have demonstrated that prolonged exposure to O₃ leads to increased expression of reactive oxygen species, causing alterations in the blood-brain barrier and damage to astrocytes and microglia. These effects can lead to an increase in the production of proinflammatory cytokines, neurotoxins, and genes, exacerbating neuronal damage and accelerating the progression of neurodegenerative diseases such as Alzheimer's, Parkinson's, and other neurological disorders. The results of this review suggest that exposure to O₃ may induce oxidative damage to the nervous system, which could have significant implications for public health.
Collapse
Affiliation(s)
- Paola Rodriguez
- Neurophysiology Department, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Mexico, Mexico
| | - Alejandro López-Landa
- Neurophysiology Department, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Mexico, Mexico; Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Héctor Romo-Parra
- Neurophysiology Department, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Mexico, Mexico; Psychology Department, Universidad Iberoamericana, Mexico, Mexico
| | - Moisés Rubio-Osornio
- Neurochemistry Department, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Mexico, Mexico
| | - Carmen Rubio
- Neurophysiology Department, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Mexico, Mexico.
| |
Collapse
|
22
|
Moradi Khankani A, Hossein Meftahi G. Pretreatment with 4-methylumbilliferon improves anxiety-like behaviors and memory impairment in stressed rats via modulation of neuronal cell death and oxidative stress. Brain Res 2024; 1844:149196. [PMID: 39181223 DOI: 10.1016/j.brainres.2024.149196] [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: 05/18/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
This work was done to investigate the ameliorating impact of 4-methylumbilliferon (4-MU) on spatial learning and memory dysfunction and restraint stress (STR)-induced anxiety-like behaviors in male Wistar rats and the underlying mechanisms. Thirty-two animals were assigned into 4 cohorts: control, 4-MU, STR, and STR+4-MU. Animals were exposed to STR for 4 h per day for 14 consecutive days or kept in normal conditions (healthy animals without exposure to stress). 4-MU (25 mg/kg) was intraperitoneally administered once daily to STR rats before restraint stress for 14 consecutive days. The behavioral tests were performed through Morris water maze tests and elevated-plus maze to examine learning/memory function, and anxiety levels, respectively. The levels of the antioxidant defense biomarkers (GPX, SOD) and MDA as an oxidant molecule in the brain tissues were measured using commercial ELISA kits. Neuronal loss or density of neurons was evaluated using Nissl staining. STR exposure could cause significant alterations in the levels of the antioxidant defense biomarkers (MDA, GPX, and SOD) in the prefrontal cortex and hippocampus, induce anxiety, and impair spatial learning and memory function. Treatment with 4-MU markedly reduced anxiety levels and improved spatial learning and memory dysfunction via restoring the antioxidant defense biomarkers to normal values and reducing MDA levels. Moreover, more intact cells with normal morphologies were detected in STR-induced animals treated with 4-MU. 4-MU could attenuate the STR-induced anxiety-like behaviors and spatial learning and memory dysfunction by reducing oxidative damage and neuronal loss in the prefrontal cortex and hippocampus region. Taken together, our findings provide new insights regarding the potential therapeutic effects of 4-MU against neurobehavioral disorders induced by STR.
Collapse
Affiliation(s)
| | - Gholam Hossein Meftahi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Roszkowicz-Ostrowska K, Młotkowska P, Marciniak E, Szlis M, Barszcz M, Misztal T. Activation of BDNF-TrkB Signaling in Specific Structures of the Sheep Brain by Kynurenic Acid. Cells 2024; 13:1928. [PMID: 39682677 DOI: 10.3390/cells13231928] [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: 10/13/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Fluctuations in kynurenic acid (KYNA) and brain-derived neurotrophic factor (BDNF) levels in the brain reflect its neurological status. The aim of the study was to investigate the effect of transiently elevated KYNA concentrations in the cerebroventricular circulation on the expression of BDNF and its high-affinity tropomyosin-related kinase receptor B (TrkB) in specific structures of the sheep brain. Intracerebroventricularly cannulated anestrous sheep were subjected to a series of four 30 min infusions of KYNA: 4 × 5 μg/60 μL/30 min (KYNA20, n = 6) and 4 × 25 μg/60 μL/30 min (KYNA100, n = 6) or a control infusion (n = 6), at 30 min intervals. Sections of the hippocampal CA3 field, amygdala (AMG), prefrontal cortex (PCx), and the hypothalamic medial-basal (MBH) and preoptic (POA) areas were dissected from the brain immediately after the experiment. The highest concentration of BDNF protein was found in the CA3 field (p < 0.001), which was 8-fold higher than in the AMG and 12-fold higher than that in the PCx (MBH and POA were not analyzed). The most pronounced BDNF mRNA expression was observed in the MBH, followed by the PCx, POA, AMG and CA3, while the highest abundance of TrkB mRNA was recorded in the AMG, followed by the MBH, PCx, CA3, and POA. KYNA increased (p < 0.05-p < 0.01) BDNF protein levels and the expression of its gene in the brain structures were examined, with the effect varying by dose and brain region. KYNA, particularly at the KYNA100 dose, also increased (p < 0.01) TrkB gene expression, except for the AMG, where the lower KYNA20 dose was more effective (p < 0.01). These findings suggest a positive relationship between KYNA levels in the cerebroventricular circulation and BDNF-TrkB expression in specific brain regions in a sheep model. This indicates that a transient increase in the CSF KYNA concentration can potentially restore BDNF production, for which deficiency underlies numerous neurological disorders.
Collapse
Affiliation(s)
- Katarzyna Roszkowicz-Ostrowska
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3 Str., 05-110 Jabłonna, Poland
| | - Patrycja Młotkowska
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3 Str., 05-110 Jabłonna, Poland
| | - Elżbieta Marciniak
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3 Str., 05-110 Jabłonna, Poland
| | - Michał Szlis
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3 Str., 05-110 Jabłonna, Poland
| | - Marcin Barszcz
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3 Str., 05-110 Jabłonna, Poland
| | - Tomasz Misztal
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3 Str., 05-110 Jabłonna, Poland
| |
Collapse
|
24
|
Heagy FK, Clements KN, Adams CL, Blain E, Issa FA. Socially induced plasticity of the posterior tuberculum and motor behavior in zebrafish (Danio rerio). J Exp Biol 2024; 227:jeb248148. [PMID: 39422204 PMCID: PMC11626077 DOI: 10.1242/jeb.248148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Social dominance is prevalent throughout the animal kingdom. It facilitates the stabilization of social relationships and allows animals to divide resources according to social rank. Zebrafish form stable dominance relationships that consist of dominants and subordinates. Although social status-dependent differences in behavior must arise as a result of neural plasticity, mechanisms by which neural circuits are reconfigured to cope with social dominance are poorly described. Here, we describe how the posterior tuberculum nucleus (PTN), which integrates sensory social information to modulate spinal motor circuits, is morphologically and functionally influenced by social status. We combined non-invasive behavioral monitoring of motor activity (startle escape and swim) and histological approaches to investigate how social dominance affects the morphological structure, axosomatic synaptic connectivity and functional activity of the PTN in relation to changes in motor behavior. We show that dopaminergic cell number significantly increases in dominants compared with subordinates, while PTN synaptic interconnectivity, demonstrated with PSD-95 expression, is higher in subordinates than in dominants. Secondly, these socially induced morphological differences emerge after 1 week of dominance formation and correlate with differences in cellular activities illustrated with higher phosphor-S6 ribosomal protein expression in dominants compared with subordinates. Thirdly, these morphological differences are reversible as the social environment evolves and correlate with adaptations in startle escape and swim behaviors. Our results provide new insights into the neural bases of social behavior that may be applicable to other social species with similar structural and functional organization.
Collapse
Affiliation(s)
- Faith K. Heagy
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Katie N. Clements
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Carrie L. Adams
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Elena Blain
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Fadi A. Issa
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| |
Collapse
|
25
|
Niu Y, Camacho MC, Wu S, Humphreys KL. The Impact of Early Life Experiences on Stress Neurobiology and the Development of Anxiety. Curr Top Behav Neurosci 2024. [PMID: 39531200 DOI: 10.1007/7854_2024_542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
We examine the association between stress exposure during early development (i.e., the prenatal period through the first two postnatal years) and variation in brain structure and function relevant to anxiety. Evidence of stress-related effects occurring in regions essential for emotional processing and regulation may increase susceptibility to anxiety.
Collapse
Affiliation(s)
- Yanbin Niu
- Vanderbilt University, Nashville, TN, USA
| | | | - Shuang Wu
- Vanderbilt University, Nashville, TN, USA
| | | |
Collapse
|
26
|
Blache D, Kho EA, Tilbrook AJ, Tomas K, Plush K, D'Souza D, Maloney SK, Cozzolino D. Near or mid-infra-red spectroscopy of the prefrontal cortex to identify previous stressful experience in an animal. Sci Rep 2024; 14:27594. [PMID: 39528591 PMCID: PMC11555064 DOI: 10.1038/s41598-024-79171-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
Measuring the quality of life of an animal in a production system is difficult, time-consuming, and expensive. We tested the ability of both NIR and MIR spectroscopy, each combined with machine learning, to predict the prior exposure of pigs to long- and short-term life challenges when they are raised in an intensive system. Samples were obtained post-mortem from two locations in the prefrontal cortex. The analysis showed a clear separation between the gray and white matter from the prefrontal cortex with MIR spectroscopy. Exposure to long-term challenge was poorly predicted by the MIR or NIR spectra (< 45% correct classifications). By contrast, the correct classification of samples according to the exposure to a short-tern challenge before death was higher than 65%. These rates of classification, considering the complexity of the stimulus and the small sample size, support that vibrational spectroscopy could be used to assess the exposure to challenging events of an animal on post-mortem brain tissue.
Collapse
Affiliation(s)
- Dominique Blache
- School of Agriculture and Environment, UWA Institute of Agriculture, The University of Western Australia, 35 Stirling Highway, Crawley, WA, 6009, Australia.
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province, P.R. China.
| | - Elise A Kho
- Centre for Animal Science, The Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Alan J Tilbrook
- Centre for Animal Science, The Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
- School of Veterinary Science, The University of Queensland, Gatton, QLD, 4343, Australia
| | - Katelyn Tomas
- Centre for Animal Science, The Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
- School of Veterinary Science, The University of Queensland, Gatton, QLD, 4343, Australia
| | - Kate Plush
- SunPork Group, Eagle Farm, QLD, 4009, Australia
| | | | - Shane K Maloney
- School of Human Sciences, UWA Institute of Agriculture, The University of Western Australia, 35 Stirling Highway, Crawley, WA, 6009, Australia
| | - Daniel Cozzolino
- Centre for Nutrition and Food Sciences, The Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
| |
Collapse
|
27
|
Herzberg MP, Smyser CD. Prenatal Social Determinants of Health: Narrative review of maternal environments and neonatal brain development. Pediatr Res 2024; 96:1417-1428. [PMID: 38961164 DOI: 10.1038/s41390-024-03345-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024]
Abstract
The Social Determinants of Health, a set of social factors including socioeconomic status, community context, and neighborhood safety among others, are well-known predictors of mental and physical health across the lifespan. Recent research has begun to establish the importance of these social factors at the earliest points of brain development, including during the prenatal period. Prenatal socioeconomic status, perceived stress, and neighborhood safety have all been reported to impact neonatal brain structure and function, with exploratory work suggesting subsequent effects on infant and child behavior. Secondary effects of the Social Determinants of Health, such as maternal sleep and psychopathology during pregnancy, have also been established as important predictors of infant brain development. This research not only establishes prenatal Social Determinants of Health as important predictors of future outcomes but may be effectively applied even before birth. Future research replicating and extending the effects in this nascent literature has great potential to produce more specific and mechanistic understanding of the social factors that shape early neurobehavioral development. IMPACT: This review synthesizes the research to date examining the effects of the Social Determinants of Health during the prenatal period and neonatal brain outcomes. Structural, functional, and diffusion-based imaging methodologies are included along with the limited literature assessing subsequent infant behavior. The degree to which results converge between studies is discussed, in combination with the methodological and sampling considerations that may contribute to divergence in study results. Several future directions are identified, including new theoretical approaches to assessing the impact of the Social Determinants of Health during the perinatal period.
Collapse
Affiliation(s)
- Max P Herzberg
- Department of Psychiatry, Washington University in St. Louis, Saint Louis, MO, USA
| | - Christopher D Smyser
- Department of Neurology, Pediatrics, and Radiology, Washington University in St. Louis, Saint Louis, MO, USA.
| |
Collapse
|
28
|
Perry RN, Ethier-Gagnon MA, Helmick C, Spinella TC, Tibbo PG, Stewart SH, Barrett SP. The impact of cannabidiol placebo on amygdala-based neural responses to an acute stressor. J Psychopharmacol 2024; 38:935-948. [PMID: 39400103 PMCID: PMC11528970 DOI: 10.1177/02698811241287557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
BACKGROUND Cannabidiol (CBD) impacts brain regions implicated in anxiety reactivity and stress reactivity (e.g., amygdala, anterior cingulate cortex (ACC), anterior insula (AI)); however, placebo-controlled studies are mixed regarding CBD's anxiolytic effects. We previously reported that CBD expectancy alone can alter subjective, physiological, and endocrine markers of stress/anxiety; however, it is unclear whether these findings reflect altered brain reactivity. This study evaluated whether CBD expectancy independently alters amygdala resting-state functional connectivity (rsFC) with the ACC and AI following acute stress. METHOD Thirty-eight (20 females) healthy adults were randomly assigned to receive accurate or inaccurate information regarding the CBD content of a CBD-free oil administered during a single experimental session. Following a baseline resting state MRI scan, participants administered their assigned oil sublingually, engaged in a stress task (serial subtraction with negative feedback) inside the scanner, and underwent another resting state MRI scan. Amygdala rsFC with the ACC and AI was measured during each scan, and the subjective state was assessed at six time points. Outcomes were analyzed using ANCOVA. RESULTS CBD expectancy (vs CBD-free expectancy) was associated with significantly weaker rsFC between the left amygdala and right ACC (p = 0.042), but did not systematically alter amygdala-AI rsFC (p-values > 0.05). We also replicated our previously reported CBD expectancy effects on subjective stress/anxiety in the scanner context. CONCLUSION CBD placebo effects may be sufficient to alter neural responses relevant to its purported anxiolytic and stress-relieving properties. Future work is needed to replicate these results and determine whether CBD expectancy and pharmacology interact to alter neural anxiety reactivity and stress reactivity.
Collapse
Affiliation(s)
- Robin N Perry
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, Canada
| | | | - Carl Helmick
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Toni C Spinella
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Philip G Tibbo
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, Canada
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Sherry H Stewart
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, Canada
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Sean P Barrett
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, Canada
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
29
|
Baram TZ, Birnie MT. Enduring memory consequences of early-life stress / adversity: Structural, synaptic, molecular and epigenetic mechanisms. Neurobiol Stress 2024; 33:100669. [PMID: 39309367 PMCID: PMC11415888 DOI: 10.1016/j.ynstr.2024.100669] [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: 03/19/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Adverse early life experiences are strongly associated with reduced cognitive function throughout life. The link is strong in many human studies, but these do not enable assigning causality, and the limited access to the live human brain can impede establishing the mechanisms by which early-life adversity (ELA) may induce cognitive problems. In experimental models, artificially imposed chronic ELA/stress results in deficits in hippocampus dependent memory as well as increased vulnerability to the deleterious effects of adult stress on memory. This causal relation of ELA and life-long memory impairments provides a framework to probe the mechanisms by which ELA may lead to human cognitive problems. Here we focus on the consequences of a one-week exposure to adversity during early postnatal life in the rodent, the spectrum of the ensuing memory deficits, and the mechanisms responsible. We highlight molecular, cellular and circuit mechanisms using convergent trans-disciplinary approaches aiming to enable translation of the discoveries in experimental models to the clinic.
Collapse
Affiliation(s)
- Tallie Z. Baram
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA
- Department of Neurology, University of California-Irvine, Irvine, CA, USA
| | - Matthew T. Birnie
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
30
|
Savva C, Vlassakev I, Bunney BG, Bunney WE, Massier L, Seldin M, Sassone-Corsi P, Petrus P, Sato S. Resilience to Chronic Stress Is Characterized by Circadian Brain-Liver Coordination. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100385. [PMID: 39387094 PMCID: PMC11462208 DOI: 10.1016/j.bpsgos.2024.100385] [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: 03/14/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 10/12/2024] Open
Abstract
Background Chronic stress has a profound impact on circadian regulation of physiology. In turn, disruption of circadian rhythms increases the risk of developing both psychiatric and metabolic disorders. To explore the role of chronic stress in modulating the links between neural and metabolic rhythms, we characterized the circadian transcriptional regulation across different brain regions and the liver as well as serum metabolomics in mice exposed to chronic social defeat stress, a validated model for studying depressive-like behaviors. Methods Male C57BL/6J mice underwent chronic social defeat stress, and subsequent social interaction screening identified distinct behavioral phenotypes associated with stress resilience and susceptibility. Stressed mice and their control littermates were sacrificed every 4 hours over the circadian cycle for comprehensive analyses of the circadian transcriptome in the hypothalamus, hippocampus, prefrontal cortex, and liver together with assessments of the circadian circulatory metabolome. Results Our data demonstrate that stress adaptation was characterized by reprogramming of the brain as well as the hepatic circadian transcriptome. Stress resiliency was associated with an increase in cyclic transcription in the hypothalamus, hippocampus, and liver. Furthermore, cross-tissue analyses revealed that resilient mice had enhanced transcriptional coordination of circadian pathways between the brain and liver. Conversely, susceptibility to social stress resulted in a loss of cross-tissue coordination. Circadian serum metabolomic profiles corroborated the transcriptome data, highlighting that stress-resilient mice gained circadian rhythmicity of circulating metabolites, including bile acids and sphingomyelins. Conclusions This study reveals that resilience to stress is characterized by enhanced metabolic rhythms and circadian brain-liver transcriptional coordination.
Collapse
Affiliation(s)
- Christina Savva
- Department of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Ivan Vlassakev
- Department of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Blynn G. Bunney
- Department of Psychiatry and Human Behavior, School of Medicine, University of California, Irvine, Irvine, California
| | - William E. Bunney
- Department of Psychiatry and Human Behavior, School of Medicine, University of California, Irvine, Irvine, California
| | - Lucas Massier
- Department of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Marcus Seldin
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California
| | - Paolo Sassone-Corsi
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California
| | - Paul Petrus
- Department of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California
| | - Shogo Sato
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California
- Center for Biological Clocks Research, Department of Biology, Texas A&M University, College Station, Texas
| |
Collapse
|
31
|
Bączyńska E, Zaręba-Kozioł M, Ruszczycki B, Krzystyniak A, Wójtowicz T, Bijata K, Pochwat B, Magnowska M, Roszkowska M, Figiel I, Masternak J, Pytyś A, Dzwonek J, Worch R, Olszyński K, Wardak A, Szymczak P, Labus J, Radwańska K, Jahołkowski P, Hogendorf A, Ponimaskin E, Filipkowski R, Szewczyk B, Bijata M, Włodarczyk J. Stress resilience is an active and multifactorial process manifested by structural, functional, and molecular changes in synapses. Neurobiol Stress 2024; 33:100683. [PMID: 39524934 PMCID: PMC11543545 DOI: 10.1016/j.ynstr.2024.100683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Stress resilience is the ability of neuronal networks to maintain their function despite the stress exposure. Using a mouse model we investigate stress resilience phenomenon. To assess the resilient and anhedonic behavioral phenotypes developed after the induction of chronic unpredictable stress, we quantitatively characterized the structural and functional plasticity of excitatory synapses in the hippocampus using a combination of proteomic, electrophysiological, and imaging methods. Our results indicate that stress resilience is an active and multifactorial process manifested by structural, functional, and molecular changes in synapses. We reveal that chronic stress influences palmitoylation of synaptic proteins, whose profiles differ between resilient and anhedonic animals. The changes in palmitoylation are predominantly related with the glutamate receptor signaling thus affects synaptic transmission and associated structures of dendritic spines. We show that stress resilience is associated with structural compensatory plasticity of the postsynaptic parts of synapses in CA1 subregion of the hippocampus.
Collapse
Affiliation(s)
- E. Bączyńska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, Warsaw, 02-781, Poland
| | - M. Zaręba-Kozioł
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
| | - B. Ruszczycki
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
- AGH University of Krakow, Faculty of Physics and Applied Computer Science, Department of Medical Physics and Biophysics, al. A. Mickiewicza 30, 30-059, Krakow, Poland
| | - A. Krzystyniak
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
| | - T. Wójtowicz
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
| | - K. Bijata
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
| | - B. Pochwat
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Cracow, Poland
| | - M. Magnowska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
| | - M. Roszkowska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
| | - I. Figiel
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
| | - J. Masternak
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
| | - A. Pytyś
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
| | - J. Dzwonek
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
| | - R. Worch
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
| | - K.H. Olszyński
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5, 02-106, Warsaw, Poland
| | - A.D. Wardak
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5, 02-106, Warsaw, Poland
| | - P. Szymczak
- Institute of Theoretical Physics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093, Warsaw, Poland
| | - J. Labus
- Cellular Neurophysiology, Center of Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - K. Radwańska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
| | - P. Jahołkowski
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Kirkeveien 166, 0424, Oslo, Norway
| | - A. Hogendorf
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Cracow, Poland
| | - E. Ponimaskin
- Cellular Neurophysiology, Center of Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - R.K. Filipkowski
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5, 02-106, Warsaw, Poland
| | - B. Szewczyk
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Cracow, Poland
| | - M. Bijata
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
| | - J. Włodarczyk
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, Warsaw, 02-093, Poland
| |
Collapse
|
32
|
Sarmiento LF, Ríos-Flórez JA, Rincón Uribe FA, Rodrigues Lima R, Kalenscher T, Gouveia A, Nitsch FJ. Do stress hormones influence choice? A systematic review of pharmacological interventions on the HPA axis and/or SAM system. Soc Cogn Affect Neurosci 2024; 19:nsae069. [PMID: 39363151 PMCID: PMC11498176 DOI: 10.1093/scan/nsae069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 08/08/2024] [Accepted: 10/03/2024] [Indexed: 10/05/2024] Open
Abstract
The hypothalamus-pituitary-adrenal axis (HPA axis) and the sympathetic-adrenal-medullary system (SAM system), two neuroendocrine systems associated with the stress response, have often been implicated to modulate decision-making in various domains. This systematic review summarizes the scientific evidence on the effects of pharmacological HPA axis and SAM system modulation on decision-making. We found 6375 references, of which 17 studies fulfilled our inclusion criteria. We quantified the risk of bias in our results with respect to missing outcome data, measurements, and selection of the reported results. The included studies administered hydrocortisone, fludrocortisone (HPA axis stimulants), yohimbine, reboxetine (SAM system stimulants), and/or propranolol (SAM system inhibitor). Integrating the evidence, we found that SAM system stimulation had no impact on risk aversion, loss aversion or intertemporal choice, while SAM system inhibition showed a tentative reduction in sensitivity to losses. HPA axis stimulation had no effect on loss aversion or reward anticipation but likely a time-dependent effect on decision under risk. Lastly, combined stimulation of both systems exhibited inconsistent results that could be explained by dose differences (loss aversion) and sex differences (risk aversion). Future research should address time-, dose-, and sex-dependencies of pharmacological effects on decision-making.
Collapse
Affiliation(s)
- Luis Felipe Sarmiento
- BiotechMed Center, BME Lab, Multimedia Systems Department, Faculty of Electronics, Telecommunications, and Informatics, Gdansk University of Technology, Gdansk 80-233, Poland
| | - Jorge Alexander Ríos-Flórez
- Professor at the Faculty of Law and Forensic Sciences, Tecnológico de Antioquia University Institution, Medellín 050034, Colombia
| | | | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural and Biology, Federal University from Pará, Belém 66075-110, Brazil
| | - Tobias Kalenscher
- Comparative Psychology, Institute of Experimental Psychology, Heinrich Heine University Düsseldorf, 40225, Germany
| | - Amauri Gouveia
- Laboratory of Neuroscience and Behavior, Federal University from Pará, Belém 66075-110, Brazil
| | - Felix Jan Nitsch
- Marketing Area, INSEAD, Fontainebleau 77300, France
- Paris Brain Institute (ICM), Sorbonne University, Paris 75013, France
| |
Collapse
|
33
|
Sandoval KE, Witt KA. Somatostatin: Linking Cognition and Alzheimer Disease to Therapeutic Targeting. Pharmacol Rev 2024; 76:1291-1325. [PMID: 39013601 PMCID: PMC11549939 DOI: 10.1124/pharmrev.124.001117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024] Open
Abstract
Over 4 decades of research support the link between Alzheimer disease (AD) and somatostatin [somatotropin-releasing inhibitory factor (SRIF)]. SRIF and SRIF-expressing neurons play an essential role in brain function, modulating hippocampal activity and memory formation. Loss of SRIF and SRIF-expressing neurons in the brain rests at the center of a series of interdependent pathological events driven by amyloid-β peptide (Aβ), culminating in cognitive decline and dementia. The connection between the SRIF and AD further extends to the neuropsychiatric symptoms, seizure activity, and inflammation, whereas preclinical AD investigations show SRIF or SRIF receptor agonist administration capable of enhancing cognition. SRIF receptor subtype-4 activation in particular presents unique attributes, with the potential to mitigate learning and memory decline, reduce comorbid symptoms, and enhance enzymatic degradation of Aβ in the brain. Here, we review the links between SRIF and AD along with the therapeutic implications. SIGNIFICANCE STATEMENT: Somatostatin and somatostatin-expressing neurons in the brain are extensively involved in cognition. Loss of somatostatin and somatostatin-expressing neurons in Alzheimer disease rests at the center of a series of interdependent pathological events contributing to cognitive decline and dementia. Targeting somatostatin-mediated processes has significant therapeutic potential for the treatment of Alzheimer disease.
Collapse
Affiliation(s)
- Karin E Sandoval
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| | - Ken A Witt
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| |
Collapse
|
34
|
Yi W, Chen W, Lan B, Yan L, Hu X, Wu J. A U-shaped relationship between chronic academic stress and the dynamics of reward processing. Neuroimage 2024; 300:120849. [PMID: 39265955 DOI: 10.1016/j.neuroimage.2024.120849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/29/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024] Open
Abstract
Despite the potential link between stress-induced reward dysfunctions and the development of mental problems, limited human research has investigated the specific impacts of chronic stress on the dynamics of reward processing. Here we aimed to investigate the relationship between chronic academic stress and the dynamics of reward processing (i.e., reward anticipation and reward consumption) using event-related potential (ERP) technology. Ninety healthy undergraduates who were preparing for the National Postgraduate Entrance Examination (NPEE) participated in the study and completed a two-door reward task, their chronic stress levels were assessed via the Perceived Stress Scale (PSS). The results showed that a lower magnitude of reward elicited more negative amplitudes of cue-N2 during the anticipatory phase, and reward omission elicited more negative amplitudes of FRN compared to reward delivery especially in high reward conditions during the consummatory phase. More importantly, the PSS score exhibited a U-shaped relationship with cue-N2 amplitudes regardless of reward magnitude during the anticipatory phase; and FRN amplitudes toward reward omission in high reward condition during the consummatory phase. These findings suggest that individuals exposed to either low or high levels of chronic stress, as opposed to moderate stress levels, exhibited a heightened reward anticipation, and an augmented violation of expectations or affective response when faced with relatively more negative outcomes.
Collapse
Affiliation(s)
- Wei Yi
- School of Psychology, Shenzhen University, 3688#, Nanhai Avenue, Nanshan District, Shenzhen 518060, China
| | - Wangxiao Chen
- School of Psychology, Shenzhen University, 3688#, Nanhai Avenue, Nanshan District, Shenzhen 518060, China
| | - Biqi Lan
- School of Psychology, Shenzhen University, 3688#, Nanhai Avenue, Nanshan District, Shenzhen 518060, China
| | - Linlin Yan
- Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Xiaoqing Hu
- Department of Psychology, The University of Hong Kong, Room 6.62, Jocky Club Tower, Pokfulam, Hong Kong, China
| | - Jianhui Wu
- School of Psychology, Shenzhen University, 3688#, Nanhai Avenue, Nanshan District, Shenzhen 518060, China.
| |
Collapse
|
35
|
Mohammadkhanizadeh A, Hosseini Y, Nikbakht F, Parvizi M, Khodabandehloo F. Evaluating the potential effects of apigenin on memory, anxiety, and social interaction amelioration after social isolation stress. Int J Dev Neurosci 2024. [PMID: 39367711 DOI: 10.1002/jdn.10380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/11/2024] [Accepted: 09/09/2024] [Indexed: 10/06/2024] Open
Abstract
Vigorous research confirmed the anti-inflammatory, antioxidant, and antidementia effects of apigenin (Api). The present study evaluated the beneficial impacts of Api administration on behaviour, brain-derived neurotrophic factor (BDNF), Interleukin 6 (IL-6), oxidative stress, and inflammation induced by social isolation (SI) stress in rats. For this purpose, rats underwent a 28-day SI period followed by a 4-week oral Api treatment (50 mg/kg/day, PO). On Day 56, behaviour tests were performed, including an elevated plus maze (EPM), Morris water maze (MWM), and three-chamber social tests. The oxidative stress markers, IL-6, and BDNF levels were measured in the hippocampus. Our results showed that SI stress caused an increase in anxiety and a decrease in spatial memory, sociability, and social preference index. In addition, SI stress increased hippocampal levels of IL-6 and malondialdehyde (MDA) content, whereas it reduced the hippocampal BDNF level and superoxide dismutase (SOD) activities. Our study indicated that Api attenuates anxiety and causes improvements in spatial memory and social interaction. These desirable effects of Api might be related to amelioration in the BDNF level, IL-6, and oxidative stress biomarkers in the hippocampus.
Collapse
Affiliation(s)
- Ali Mohammadkhanizadeh
- Behavioural and Cognitive Science Research Centre, AJA University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center and Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Yasaman Hosseini
- Behavioural and Cognitive Science Research Centre, AJA University of Medical Sciences, Tehran, Iran
| | - Farnaz Nikbakht
- Cellular and Molecular Research Center and Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Parvizi
- Behavioural and Cognitive Science Research Centre, AJA University of Medical Sciences, Tehran, Iran
- Department of Physiology, faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Fatemeh Khodabandehloo
- Department of Genetic and Advanced Medicine Technology, faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Lugenbühl JF, Viho EMG, Binder EB, Daskalakis NP. Stress Molecular Signaling in Interaction With Cognition. Biol Psychiatry 2024:S0006-3223(24)01628-7. [PMID: 39368530 DOI: 10.1016/j.biopsych.2024.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/02/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024]
Abstract
Exposure to stressful life events is associated with a high risk of developing psychiatric disorders with a wide variety of symptoms. Cognitive symptoms in stress-related psychiatric disorders can be particularly challenging to understand, both for those experiencing them and for health care providers. To gain insights, it is important to capture stress-induced structural, epigenomic, transcriptomic, and proteomic changes in relevant brain regions such as the amygdala, hippocampus, locus coeruleus, and prefrontal cortex that result in long-lasting alterations in brain function. In this review, we will emphasize a subset of stress molecular mechanisms that alter neuroplasticity, neurogenesis, and balance between excitatory and inhibitory neurons. Then, we discuss how to identify genetic risk factors that may accelerate stress-driven or stress-induced cognitive impairment. Despite the development of new technologies such as single-cell resolution sequencing, our understanding of the molecular effects of stress in the brain remains to be deepened. A better understanding of the diversity of stress effects in different brain regions and cell types is a prerequisite to open new avenues for mechanism-informed prevention and treatment of stress-related cognitive symptoms.
Collapse
Affiliation(s)
- Justina F Lugenbühl
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts; Department of Psychiatry and Neuropsychology, School for Mental Health, and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Eva M G Viho
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Elisabeth B Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany.
| | - Nikolaos P Daskalakis
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| |
Collapse
|
37
|
Kuhn AM, Bosis KE, Wohleb ES. Looking Back to Move Forward: Research in Stress, Behavior, and Immune Function. Neuroimmunomodulation 2024; 31:211-229. [PMID: 39369707 DOI: 10.1159/000541592] [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: 05/15/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND From the original studies investigating the effects of adrenal gland secretion to modern high-throughput multidimensional analyses, stress research has been a topic of scientific interest spanning just over a century. SUMMARY The objective of this review was to provide historical context for influential discoveries, surprising findings, and preclinical models in stress-related neuroimmune research. Furthermore, we summarize this work and present a current understanding of the stress pathways and their effects on the immune system and behavior. We focus on recent work demonstrating stress-induced immune changes within the brain and highlight studies investigating stress effects on microglia. Lastly, we conclude with potential areas for future investigation concerning microglia heterogeneity, bone marrow niches, and sex differences. KEY MESSAGES Stress is a phenomenon that ties together not only the central and peripheral nervous system, but the immune system as well. The cumulative effects of stress can enhance or suppress immune function, based on the intensity and duration of the stressor. These stress-induced immune alterations are associated with neurobiological changes, including structural remodeling of neurons and decreased neurogenesis, and these contribute to the development of behavioral and cognitive deficits. As such, research in this field has revealed important insights into neuroimmune communication as well as molecular and cellular mediators of complex behaviors relevant to psychiatric disorders.
Collapse
Affiliation(s)
- Alexander M Kuhn
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kelly E Bosis
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Eric S Wohleb
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
38
|
Strömbäck M, Wiik E, Hägglund Oja M, Kakko J. Learning to be mindful ─ experiences of mindfulness-based stress reduction for young adults with moderate to severe mental disorders. J Bodyw Mov Ther 2024; 40:2074-2081. [PMID: 39593567 DOI: 10.1016/j.jbmt.2024.10.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/30/2024] [Accepted: 10/14/2024] [Indexed: 11/28/2024]
Abstract
INTRODUCTION Mental disorders among young adults are a major health challenge. Mindfulness-based stress reduction (MBSR) has in previous research shown positive effects on mental health. However, research evaluating MBSR for young adults with mental disorders is insufficient and qualitative research exploring experiences of the programme is scarce. The aim of this study was to explore experiences of MBSR as a supplementary treatment in psychiatric outpatient care among young adults with moderate to severe mental disorders, from a short- and long-term perspective. METHOD Eleven young adults (aged 18-27 years) who had participated in an MBSR programme were interviewed approximately three months or two years later. The analytic method was Grounded Theory. RESULTS The analysis resulted in a theoretical model including one core category and five categories. The model illustrated a learning process, explaining the participants' short- and long-term experiences of learning to pay attention to the body, regulate emotions, a change of attitude towards oneself, and still using skills two years after the programme. DISCUSSION The MBSR programme was experienced as a learning process which improved the longstanding ability to cope with emotional stress in daily life. Tailoring MBSR to individual needs may further enhance its effectiveness. CONCLUSION The experienced short- and long-term health-promotion benefits of an MBSR programme among young adults with mental disorders support its implementation in psychiatric and primary health care. Future research should investigate how to optimise implementation and tailor to individual needs.
Collapse
Affiliation(s)
- Maria Strömbäck
- Department of Clinical Science, Psychiatry, Umeå University, Umeå, Sweden; Department of Community Medicine and Rehabilitation, Physiotherapy, Umeå University, Umeå, Sweden.
| | - Erica Wiik
- Department of Community Medicine and Rehabilitation, Physiotherapy, Umeå University, Umeå, Sweden
| | - Malin Hägglund Oja
- University Hospital of Umeå, Psychiatric Care, Region of Västerbotten, Umeå, Sweden
| | - Johan Kakko
- Department of Clinical Science, Psychiatry, Umeå University, Umeå, Sweden
| |
Collapse
|
39
|
Mazurie Z, Branchereau P, Cattaert D, Henkous N, Savona-Baron C, Vouimba RM. Acute stress differently modulates interneurons excitability and synaptic plasticity in the primary motor cortex of wild-type and SOD1 G93A mouse model of ALS. J Physiol 2024; 602:4987-5015. [PMID: 39216080 DOI: 10.1113/jp285210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 07/12/2024] [Indexed: 09/04/2024] Open
Abstract
Primary motor cortex (M1) network stability depends on activity of inhibitory interneurons, for which susceptibility to stress was previously demonstrated in limbic regions. Hyperexcitability in M1 following changes in the excitatory/inhibitory balance is a key pathological hallmark of amyotrophic lateral sclerosis (ALS). Using electrophysiological approaches, we assessed the impact of acute restraint stress on inhibitory interneurons excitability and global synaptic plasticity in M1 of the SOD1G93A ALS mouse model at a late pre-symptomatic stage (10-12.5 weeks). Based on their firing type (continuous, discontinuous, with accommodation or not) and electrophysiological characteristics (resting potential, rheobase, firing frequency), interneurons from M1 slices were separated into four clusters, labelled from 1 to 4. Among them, only interneurons from the first cluster, presenting continuous firing with few accommodations, tended to show increased excitability in wild-type (WT) and decreased excitability in SOD1G93A animals following stress. In vivo analyses of evoked field potentials showed that stress suppressed the theta burst-induced plasticity of an excitatory component (N1) recorded in the superficial layers of M1 in WT, with no impact on an inhibitory complex (N2-P1) from the deeper layers. In SOD1G93A mice, stress did not affect N1 but suppressed the N2-P1 plasticity. These data suggest that stress can alter M1 network functioning in a different manner in WT and SOD1G93A mice, possibly through changes of inhibitory interneurons excitability and synaptic plasticity. This suggests that stress-induced activity changes in M1 may therefore influence ALS outcomes. KEY POINTS: Disruption of the excitatory/inhibitory balance in the primary motor cortex (M1) has been linked to cortical hyperexcitability development, a key pathological hallmark of amyotrophic lateral sclerosis (ALS). Psychological stress was reported to influence excitatory/inhibitory balance in limbic regions, but very little is known about its influence on the M1 functioning under physiological or pathological conditions. Our study revealed that acute stress influences the excitatory/inhibitory balance within the M1, through changes in interneurons excitability along with network plasticity. Such changes were different in pathological (SOD1G93A ALS mouse model) vs. physiological (wild-type) conditions. The results of our study help us to better understand how stress modulates the M1 and highlight the need to further characterize stress-induced motor cortex changes because it may be of importance when evaluating ALS outcomes.
Collapse
Affiliation(s)
- Zoé Mazurie
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Pascal Branchereau
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Daniel Cattaert
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Nadia Henkous
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Catherine Savona-Baron
- Present address: BoRdeaux Institute of onCology (BRIC), INSERM U1312, University of Bordeaux, Bordeaux, France
| | - Rose-Marie Vouimba
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| |
Collapse
|
40
|
Higa GSV, Viana FJC, Francis-Oliveira J, Cruvinel E, Franchin TS, Marcourakis T, Ulrich H, De Pasquale R. Serotonergic neuromodulation of synaptic plasticity. Neuropharmacology 2024; 257:110036. [PMID: 38876308 DOI: 10.1016/j.neuropharm.2024.110036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/15/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Synaptic plasticity constitutes a fundamental process in the reorganization of neural networks that underlie memory, cognition, emotional responses, and behavioral planning. At the core of this phenomenon lie Hebbian mechanisms, wherein frequent synaptic stimulation induces long-term potentiation (LTP), while less activation leads to long-term depression (LTD). The synaptic reorganization of neuronal networks is regulated by serotonin (5-HT), a neuromodulator capable of modify synaptic plasticity to appropriately respond to mental and behavioral states, such as alertness, attention, concentration, motivation, and mood. Lately, understanding the serotonergic Neuromodulation of synaptic plasticity has become imperative for unraveling its impact on cognitive, emotional, and behavioral functions. Through a comparative analysis across three main forebrain structures-the hippocampus, amygdala, and prefrontal cortex, this review discusses the actions of 5-HT on synaptic plasticity, offering insights into its role as a neuromodulator involved in emotional and cognitive functions. By distinguishing between plastic and metaplastic effects, we provide a comprehensive overview about the mechanisms of 5-HT neuromodulation of synaptic plasticity and associated functions across different brain regions.
Collapse
Affiliation(s)
- Guilherme Shigueto Vilar Higa
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil; Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Felipe José Costa Viana
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - José Francis-Oliveira
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Emily Cruvinel
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Thainá Soares Franchin
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Tania Marcourakis
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Roberto De Pasquale
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
41
|
Chen CW, Yeh WL, Charoensaensuk V, Lin C, Yang LY, Chen MK, Yeh T, Tsai CF, Lu DY. Oral administration of osthole mitigates maladaptive behaviors through PPARα activation in mice subjected to repeated social defeat stress. Neurochem Int 2024; 179:105811. [PMID: 39053771 DOI: 10.1016/j.neuint.2024.105811] [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: 05/15/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 07/27/2024]
Abstract
Psychological stress induces neuroinflammatory responses, which are associated with the pathogenesis of various psychiatric disorders, such as posttraumatic stress disorder and anxiety. Osthole-a natural coumarin isolated from the seeds of the Chinese herb Cnidium monnieri-exerts anti-inflammatory and antioxidative effects on the central nervous system. However, the therapeutic benefits of osthole against psychiatric disorders remain largely unknown. We previously demonstrated that mice subjected to repeated social defeat stress (RSDS) in the presence of aggressor mice exhibited symptoms of posttraumatic stress disorder, such as social avoidance and anxiety-like behaviors. In this study, we investigated the therapeutic effects of osthole and the underlying molecular mechanisms. Osthole exerted therapeutic effects on cognitive behaviors, mitigating anxiety-like behaviors and social avoidance in a mouse model of RSDS. The anti-inflammatory response induced by the oral administration of osthole was strengthened through the upregulation of heme oxygenase-1 expression. The expression of PPARα was inhibited in mice subjected to RSDS. Nonetheless, osthole treatment reversed the inhibition of PPARα expression. We identified a positive correlation between heme oxygenase-1 expression and PPARα expression in osthole-treated mice. In conclusion, osthole has potential as a Chinese herbal medicine for anxiety disorders. When designing novel drugs for psychiatric disorders, researchers should consider targeting the activation of PPARα.
Collapse
Affiliation(s)
- Chao-Wei Chen
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung, Taiwan
| | - Wei-Lan Yeh
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung, Taiwan; Department of Biochemistry, School of Medicine, China Medical University, Taichung, Taiwan
| | - Vichuda Charoensaensuk
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chingju Lin
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Liang-Yo Yang
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Mao-Kai Chen
- Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan
| | - Tong Yeh
- Department of Medicine, School of Medicine, China Medical University, Taichung, Taiwan
| | - Cheng-Fang Tsai
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan.
| | - Dah-Yuu Lu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
42
|
Butto T, Chongtham MC, Mungikar K, Hartwich D, Linke M, Ruffini N, Radyushkin K, Schweiger S, Winter J, Gerber S. Characterization of transcriptional profiles associated with stress-induced neuronal activation in Arc-GFP mice. Mol Psychiatry 2024; 29:3010-3023. [PMID: 38649752 PMCID: PMC11449785 DOI: 10.1038/s41380-024-02555-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 03/21/2024] [Accepted: 04/05/2024] [Indexed: 04/25/2024]
Abstract
Chronic stress has become a predominant factor associated with a variety of psychiatric disorders, such as depression and anxiety, in both human and animal models. Although multiple studies have looked at transcriptional changes after social defeat stress, these studies primarily focus on bulk tissues, which might dilute important molecular signatures of social interaction in activated cells. In this study, we employed the Arc-GFP mouse model in conjunction with chronic social defeat (CSD) to selectively isolate activated nuclei (AN) populations in the ventral hippocampus (vHIP) and prefrontal cortex (PFC) of resilient and susceptible animals. Nuclear RNA-seq of susceptible vs. resilient populations revealed distinct transcriptional profiles linked predominantly with neuronal and synaptic regulation mechanisms. In the vHIP, susceptible AN exhibited increased expression of genes related to the cytoskeleton and synaptic organization. At the same time, resilient AN showed upregulation of cell adhesion genes and differential expression of major glutamatergic subunits. In the PFC, susceptible mice exhibited upregulation of synaptotagmins and immediate early genes (IEGs), suggesting a potentially over-amplified neuronal activity state. Our findings provide a novel view of stress-exposed neuronal activation and the molecular response mechanisms in stress-susceptible vs. resilient animals, which may have important implications for understanding mental resilience.
Collapse
Affiliation(s)
- Tamer Butto
- Institute for Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University, 55128, Mainz, Germany
| | | | - Kanak Mungikar
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Dewi Hartwich
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Matthias Linke
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Nicolas Ruffini
- Leibniz Institute for Resilience Research, Wallstr 7, 55122, Mainz, Germany
| | | | - Susann Schweiger
- Leibniz Institute for Resilience Research, Wallstr 7, 55122, Mainz, Germany
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Jennifer Winter
- Leibniz Institute for Resilience Research, Wallstr 7, 55122, Mainz, Germany.
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Susanne Gerber
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| |
Collapse
|
43
|
Goodman EJ, DiSabato DJ, Sheridan JF, Godbout JP. Novel microglial transcriptional signatures promote social and cognitive deficits following repeated social defeat. Commun Biol 2024; 7:1199. [PMID: 39341879 PMCID: PMC11438916 DOI: 10.1038/s42003-024-06898-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
Chronic stress is associated with anxiety and cognitive impairment. Repeated social defeat (RSD) in mice induces anxiety-like behavior driven by microglia and the recruitment of inflammatory monocytes to the brain. Nonetheless, it is unclear how microglia communicate with other cells to modulate the physiological and behavioral responses to stress. Using single-cell (sc)RNAseq, we identify novel, to the best of our knowledge, stress-associated microglia in the hippocampus defined by RNA profiles of cytokine/chemokine signaling, cellular stress, and phagocytosis. Microglia depletion with a CSF1R antagonist (PLX5622) attenuates the stress-associated profile of leukocytes, endothelia, and astrocytes. Furthermore, RSD-induced social withdrawal and cognitive impairment are microglia-dependent, but social avoidance is microglia-independent. Furthermore, single-nuclei (sn)RNAseq shows robust responses to RSD in hippocampal neurons that are both microglia-dependent and independent. Notably, stress-induced CREB, oxytocin, and glutamatergic signaling in neurons are microglia-dependent. Collectively, these stress-associated microglia influence transcriptional profiles in the hippocampus related to social and cognitive deficits.
Collapse
Affiliation(s)
- Ethan J Goodman
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio, 43210, USA
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Damon J DiSabato
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio, 43210, USA
| | - John F Sheridan
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, Ohio, 43210, USA.
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, Ohio, 43210, USA.
| | - Jonathan P Godbout
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio, 43210, USA.
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, Ohio, 43210, USA.
| |
Collapse
|
44
|
Otten J, Dan S, Rostin L, Profetto AE, Lardenoije R, Klengel T. Spatial transcriptomics reveals modulation of transcriptional networks across brain regions after auditory threat conditioning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614979. [PMID: 39386587 PMCID: PMC11463379 DOI: 10.1101/2024.09.25.614979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Prior research has demonstrated genome-wide transcriptional changes related to fear and anxiety across species, often focusing on individual brain regions or cell types. However, the extent of gene expression differences across brain regions and how these changes interact at the level of transcriptional connectivity remains unclear. To address this, we performed spatial transcriptomics RNAseq analyses in an auditory threat conditioning paradigm in mice. We generated a spatial transcriptomic atlas of a coronal mouse brain section covering cortical and subcortical regions, corresponding to histologically defined regions. Our finding revealed widespread transcriptional responses across all brain regions examined, particularly in the medial and lateral habenula, and the choroid plexus. Network analyses highlighted altered transcriptional connectivity between cortical and subcortical regions, emphasizing the role of steroidogenic factor 1. These results provide new insights into the transcriptional networks involved in auditory threat conditioning, enhancing our understanding of molecular and neural mechanisms underlying fear and anxiety disorders.
Collapse
|
45
|
Crocetta A, Liloia D, Costa T, Duca S, Cauda F, Manuello J. From gut to brain: unveiling probiotic effects through a neuroimaging perspective-A systematic review of randomized controlled trials. Front Nutr 2024; 11:1446854. [PMID: 39360283 PMCID: PMC11444994 DOI: 10.3389/fnut.2024.1446854] [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: 06/10/2024] [Accepted: 09/02/2024] [Indexed: 10/04/2024] Open
Abstract
The gut-brain axis, a bidirectional communication network between the gastrointestinal system and the brain, significantly influences mental health and behavior. Probiotics, live microorganisms conferring health benefits, have garnered attention for their potential to modulate this axis. However, their effects on brain function through gut microbiota modulation remain controversial. This systematic review examines the effects of probiotics on brain activity and functioning, focusing on randomized controlled trials using both resting-state and task-based functional magnetic resonance imaging (fMRI) methodologies. Studies investigating probiotic effects on brain activity in healthy individuals and clinical populations (i.e., major depressive disorder and irritable bowel syndrome) were identified. In healthy individuals, task-based fMRI studies indicated that probiotics modulate brain activity related to emotional regulation and cognitive processing, particularly in high-order areas such as the amygdala, precuneus, and orbitofrontal cortex. Resting-state fMRI studies revealed changes in connectivity patterns, such as increased activation in the Salience Network and reduced activity in the Default Mode Network. In clinical populations, task-based fMRI studies showed that probiotics could normalize brain function in patients with major depressive disorder and irritable bowel syndrome. Resting-state fMRI studies further suggested improved connectivity in mood-regulating networks, specifically in the subcallosal cortex, amygdala and hippocampus. Despite promising findings, methodological variability and limited sample sizes emphasize the need for rigorous, longitudinal research to clarify the beneficial effects of probiotics on the gut-brain axis and mental health.
Collapse
Affiliation(s)
- Annachiara Crocetta
- Department of Psychology, Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, University of Turin, Turin, Italy
- Department of Psychology, GCS fMRI, Koelliker Hospital, University of Turin, Turin, Italy
| | - Donato Liloia
- Department of Psychology, Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, University of Turin, Turin, Italy
- Department of Psychology, GCS fMRI, Koelliker Hospital, University of Turin, Turin, Italy
| | - Tommaso Costa
- Department of Psychology, Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, University of Turin, Turin, Italy
- Department of Psychology, GCS fMRI, Koelliker Hospital, University of Turin, Turin, Italy
- Neuroscience Institute of Turin (NIT), University of Turin, Turin, Italy
| | - Sergio Duca
- Department of Psychology, GCS fMRI, Koelliker Hospital, University of Turin, Turin, Italy
| | - Franco Cauda
- Department of Psychology, Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, University of Turin, Turin, Italy
- Department of Psychology, GCS fMRI, Koelliker Hospital, University of Turin, Turin, Italy
- Neuroscience Institute of Turin (NIT), University of Turin, Turin, Italy
| | - Jordi Manuello
- Department of Psychology, Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, University of Turin, Turin, Italy
- Department of Psychology, GCS fMRI, Koelliker Hospital, University of Turin, Turin, Italy
- Move’N’Brains Lab, Department of Psychology, University of Turin, Turin, Italy
| |
Collapse
|
46
|
McManus E, Haroon H, Duncan NW, Elliott R, Muhlert N. Hippocampal and limbic microstructure changes associated with stress across the lifespan: a UK biobank study. Sci Rep 2024; 14:21735. [PMID: 39289386 PMCID: PMC11408494 DOI: 10.1038/s41598-024-71965-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 09/02/2024] [Indexed: 09/19/2024] Open
Abstract
Experiencing highly stressful events can have detrimental and lasting effects on brain morphology. The current study explores the effects of stress during childhood and adulthood on grey matter macro- and microstructure using a sub-sample of 720 participants from the UK Biobank with very high or very low childhood and adulthood stress scores. We used T1-weighted and diffusion MRI data to assess grey matter macro- and microstructure within bilateral hippocampus, amygdala and thalamus. Findings showed that childhood stress is associated with changes in microstructural measures bilaterally within the hippocampus and amygdala. No effects of adulthood stress on brain microstructure were found. No interaction effects between sex and stress (either childhood or adulthood) were observed for any brain imaging measure. Analysis of sub-segments of the hippocampus showed that childhood stress predominantly impacted the bilateral heads of the hippocampus. Overall, these findings suggest that highly stressful experiences during childhood, but not adulthood, have lasting impact on brain microstructure. The effects of these experiences in childhood appear to persist regardless of experiences of high or low stress in adulthood.
Collapse
Affiliation(s)
- Elizabeth McManus
- School of Health Sciences, The University of Manchester, H.18 Coupland 1 Building, Oxford Rd, Manchester, M13 9PL, UK.
| | - Hamied Haroon
- School of Health Sciences, The University of Manchester, H.18 Coupland 1 Building, Oxford Rd, Manchester, M13 9PL, UK
| | - Niall W Duncan
- Graduate Institute of Mind, Brain and Consciousness, Taipei Medical University, Taipei, Taiwan
- Brain and Consciousness Research Centre, TMU Shuang Ho Hospital, New Taipei City, Taiwan
| | - Rebecca Elliott
- School of Health Sciences, The University of Manchester, H.18 Coupland 1 Building, Oxford Rd, Manchester, M13 9PL, UK
| | - Nils Muhlert
- School of Health Sciences, The University of Manchester, H.18 Coupland 1 Building, Oxford Rd, Manchester, M13 9PL, UK
| |
Collapse
|
47
|
Coleman EM, White M, Antonoudiou P, Weiss GL, Scarpa G, Stone B, Maguire J. Early life stress influences epilepsy outcomes in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612052. [PMID: 39314367 PMCID: PMC11419006 DOI: 10.1101/2024.09.09.612052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Stress is a common seizure trigger that has been implicated in worsening epilepsy outcomes. The neuroendocrine response to stress is mediated by the hypothalamic-pituitary-adrenal (HPA) axis and HPA axis dysfunction worsens epilepsy outcomes, increasing seizure burden, behavioral comorbidities, and risk for sudden unexpected death in epilepsy (SUDEP) in mice. Early life stress (ELS) reprograms the HPA axis into adulthood, impacting both the basal and stress-induced activity. Thus, we propose that ELS may influence epilepsy outcomes by influencing the function of the HPA axis. To test this hypothesis, we utilized the maternal separation paradigm and examined the impact on seizure susceptibility. We show that ELS exerts a sex dependent effect on seizure susceptibility in response to acute administration of the chemoconvulsant, kainic acid, which is associated with an altered relationship between seizure activity and HPA axis function. To further examine the impact of ELS on epilepsy outcomes, we utilized the intrahippocampal kainic acid model of chronic epilepsy in mice previously exposed to maternal separation. We find that the relationship between corticosterone levels and the extent of epileptiform activity is altered in mice subjected to ELS. We demonstrate that ELS impacts behavioral outcomes associated with chronic epilepsy in a sex-dependent manner, with females being more affected. We also observe reduced mortality (presumed SUDEP) in female mice subjected to ELS, consistent with previous findings suggesting a role for HPA axis dysfunction in SUDEP risk. These data demonstrate for the first time that ELS influences epilepsy outcomes and suggest that previous life experiences may impact the trajectory of epilepsy.
Collapse
|
48
|
Bonaz B, Sinniger V, Pellissier S. Role of stress and early-life stress in the pathogeny of inflammatory bowel disease. Front Neurosci 2024; 18:1458918. [PMID: 39319312 PMCID: PMC11420137 DOI: 10.3389/fnins.2024.1458918] [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: 07/03/2024] [Accepted: 08/23/2024] [Indexed: 09/26/2024] Open
Abstract
Numerous preclinical and clinical studies have shown that stress is one of the main environmental factor playing a significant role in the pathogeny and life-course of bowel diseases. However, stressful events that occur early in life, even during the fetal life, leave different traces within the central nervous system, in area involved in stress response and autonomic network but also in emotion, cognition and memory regulation. Early-life stress can disrupt the prefrontal-amygdala circuit thus favoring an imbalance of the autonomic nervous system and the hypothalamic-pituitary adrenal axis, resulting in anxiety-like behaviors. The down regulation of vagus nerve and cholinergic anti-inflammatory pathway favors pro-inflammatory conditions. Recent data suggest that emotional abuse at early life are aggravating risk factors in inflammatory bowel disease. This review aims to unravel the mechanisms that explain the consequences of early life events and stress in the pathophysiology of inflammatory bowel disease and their mental co-morbidities. A review of therapeutic potential will also be covered.
Collapse
Affiliation(s)
- Bruno Bonaz
- Université Grenoble Alpes, Service d'Hépato-Gastroentérologie, Grenoble Institut Neurosciences, Grenoble, France
| | - Valérie Sinniger
- Université Grenoble Alpes, Service d'Hépato-Gastroentérologie, Grenoble Institut Neurosciences, Grenoble, France
| | - Sonia Pellissier
- Université Savoie Mont Blanc, Université Grenoble Alpes, LIP/PC2S, Chambéry, France
| |
Collapse
|
49
|
Montes-Rodríguez CJ, Hernández-Reyes ED, Piña-Díaz V, Muñoz-Torres Z, Pérez-Zarazúa I, Urteaga-Urías E, Prospéro-García O. Activity-Dependent Synaptic Plasticity in the Medial Prefrontal Cortex of Male Rats Underlies Resilience-Related Behaviors to Social Adversity. J Neurosci Res 2024; 102:e25377. [PMID: 39275861 DOI: 10.1002/jnr.25377] [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: 01/18/2024] [Revised: 07/02/2024] [Accepted: 08/11/2024] [Indexed: 09/16/2024]
Abstract
Individuals considered resilient can overcome adversity, achieving normal physical and psychological development, while those deemed vulnerable may not. Adversity promotes structural and functional alterations in the medial prefrontal cortex (mPFC) and hippocampus. Moreover, activity-dependent synaptic plasticity is intricately linked to neuronal shaping resulting from experiences. We hypothesize that this plasticity plays a crucial role in resilience processes. However, there is a notable absence of studies investigating this plasticity and behavioral changes following social adversity at different life stages. Consequently, we evaluated the impact of social adversity during early postnatal development (maternal separation [MS]), adulthood (social defeat [SD]), and a combined exposure (MS + SD) on behavioral outcomes (anxiety, motivation, anhedonia, and social interaction). We also examined cFos expression induced by social interaction in mPFC and hippocampus of adult male rats. Behavioral analyses revealed that SD-induced anhedonia, whereas MS + SD increased social interaction and mitigated SD-induced anhedonia. cFos evaluation showed that social interaction heightened plasticity in the prelimbic (PrL) and infralimbic (IL) cortices, dentate gyrus (DG), CA3, and CA1. Social interaction-associated plasticity was compromised in IL and PrL cortices of the MS and SD groups. Interestingly, social interaction-induced plasticity was restored in the MS + SD group. Furthermore, plasticity was impaired in DG by all social stressors, and in CA3 was impaired by SD. Our findings suggest in male rats (i) two adverse social experiences during development foster resilience; (ii) activity-dependent plasticity in the mPFC is a foundation for resilience to social adversity; (iii) plasticity in DG is highly susceptible to social adversity.
Collapse
Affiliation(s)
- Corinne J Montes-Rodríguez
- Centro de Investigación en Ciencias, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Erika D Hernández-Reyes
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Facultad de Psicología, UNAM, Mexico City, Mexico
| | - Vanessa Piña-Díaz
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Facultad de Psicología, UNAM, Mexico City, Mexico
| | - Zeidy Muñoz-Torres
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Facultad de Psicología, UNAM, Mexico City, Mexico
| | - Itzel Pérez-Zarazúa
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Facultad de Psicología, UNAM, Mexico City, Mexico
| | - Emiliano Urteaga-Urías
- Academia de Cultura Científica y Humanística, Universidad Autónoma de la Ciudad de México, Mexico City, Mexico
| | - Oscar Prospéro-García
- Laboratorio de Cannabinoides, Departamento de Fisiología, Facultad de Medicina, UNAM, Mexico City, Mexico
| |
Collapse
|
50
|
Colodete DA, Grace AA, Guimarães FS, Gomes FV. Degradation of Perineuronal Nets in the Ventral Hippocampus of Adult Rats Recreates an Adolescent-Like Phenotype of Stress Susceptibility. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100338. [PMID: 39099729 PMCID: PMC11295568 DOI: 10.1016/j.bpsgos.2024.100338] [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: 03/20/2024] [Revised: 05/08/2024] [Accepted: 05/12/2024] [Indexed: 08/06/2024] Open
Abstract
Background Psychiatric disorders often emerge during late adolescence/early adulthood, a period with increased susceptibility to socioenvironmental factors that coincides with incomplete parvalbumin interneuron (PVI) development. Stress during this period causes functional loss of PVIs in the ventral hippocampus (vHip), which has been associated with dopamine system overdrive. This vulnerability persists until the appearance of perineuronal nets (PNNs) around PVIs. We assessed the long-lasting effects of adolescent or adult stress on behavior, ventral tegmental area dopamine neuron activity, and the number of PVIs and their associated PNNs in the vHip. Additionally, we tested whether PNN removal in the vHip of adult rats, proposed to reset PVIs to a juvenile-like state, would recreate an adolescent-like phenotype of stress susceptibility. Methods Male rats underwent a 10-day stress protocol during adolescence or adulthood. Three to 4 weeks poststress, we evaluated behaviors related to anxiety, sociability, and cognition, ventral tegmental area dopamine neuron activity, and the number of PV+ and PNN+ cells in the vHip. Furthermore, adult animals received intra-vHip infusion of ChABC (chondroitinase ABC) to degrade PNNs before undergoing stress. Results Unlike adult stress, adolescent stress induced anxiety responses, reduced sociability, cognitive deficits, ventral tegmental area dopamine system overdrive, and decreased PV+ and PNN+ cells in the vHip. However, intra-vHip ChABC infusion caused the adult stress to produce changes similar to the ones observed after adolescent stress. Conclusions Our findings underscore adolescence as a period of heightened vulnerability to the long-lasting impact of stress and highlight the protective role of PNNs against stress-induced damage in PVIs.
Collapse
Affiliation(s)
- Débora A.E. Colodete
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Anthony A. Grace
- Departments of Neuroscience, Psychiatry, and Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Francisco S. Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Felipe V. Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|