1
|
Ye J, Duan C, Han J, Chen J, Sun N, Li Y, Yuan T, Peng D. Peripheral mitochondrial DNA as a neuroinflammatory biomarker for major depressive disorder. Neural Regen Res 2025; 20:1541-1554. [PMID: 38934398 PMCID: PMC11688552 DOI: 10.4103/nrr.nrr-d-23-01878] [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: 01/03/2024] [Revised: 03/09/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
In the pathogenesis of major depressive disorder, chronic stress-related neuroinflammation hinders favorable prognosis and antidepressant response. Mitochondrial DNA may be an inflammatory trigger, after its release from stress-induced dysfunctional central nervous system mitochondria into peripheral circulation. This evidence supports the potential use of peripheral mitochondrial DNA as a neuroinflammatory biomarker for the diagnosis and treatment of major depressive disorder. Herein, we critically review the neuroinflammation theory in major depressive disorder, providing compelling evidence that mitochondrial DNA release acts as a critical biological substrate, and that it constitutes the neuroinflammatory disease pathway. After its release, mitochondrial DNA can be carried in the exosomes and transported to extracellular spaces in the central nervous system and peripheral circulation. Detectable exosomes render encaged mitochondrial DNA relatively stable. This mitochondrial DNA in peripheral circulation can thus be directly detected in clinical practice. These characteristics illustrate the potential for mitochondrial DNA to serve as an innovative clinical biomarker and molecular treatment target for major depressive disorder. This review also highlights the future potential value of clinical applications combining mitochondrial DNA with a panel of other biomarkers, to improve diagnostic precision in major depressive disorder.
Collapse
Affiliation(s)
- Jinmei Ye
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cong Duan
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaxin Han
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jinrong Chen
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Ning Sun
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Yuan Li
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tifei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Daihui Peng
- Division of Mood Disorder, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Li Q, Xie Y, Lin J, Li M, Gu Z, Xin T, Zhang Y, Lu Q, Guo Y, Xing Y, Wang W. Microglia Sing the Prelude of Neuroinflammation-Associated Depression. Mol Neurobiol 2025; 62:5311-5332. [PMID: 39535682 DOI: 10.1007/s12035-024-04575-w] [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: 06/14/2023] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Major depressive disorder (MDD) is a psychiatric condition characterized by sadness and anhedonia and is closely linked to chronic low-grade neuroinflammation, which is primarily induced by microglia. Nonetheless, the mechanisms by which microglia elicit depressive symptoms remain uncertain. This review focuses on the mechanism linking microglia and depression encompassing the breakdown of the blood-brain barrier, the hypothalamic-pituitary-adrenal axis, the gut-brain axis, the vagus and sympathetic nervous systems, and the susceptibility influenced by epigenetic modifications on microglia. These pathways may lead to the alterations of microglia in cytokine levels, as well as increased oxidative stress. Simultaneously, many antidepressant treatments can alter the immune phenotype of microglia, while anti-inflammatory treatments can also have antidepressant effects. This framework linking microglia, neuroinflammation, and depression could serve as a reference for targeting microglia to treat depression.
Collapse
Affiliation(s)
- Qingqing Li
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Ying Xie
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Jinyi Lin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Miaomiao Li
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Ziyan Gu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Tianli Xin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Yang Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Qixia Lu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Yihui Guo
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Yanhong Xing
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China.
| | - Wuyang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
3
|
Connolly MG, Johnson ZV, Chu L, Johnson ND, Buhr TJ, McNeill EM, Clark PJ, Rhodes JS. Single-Nucleus RNA Sequencing Reveals Enduring Signatures of Acute Stress and Chronic Exercise in Striatal Microglia. GENES, BRAIN, AND BEHAVIOR 2025; 24:e70019. [PMID: 40045485 PMCID: PMC11882474 DOI: 10.1111/gbb.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 03/09/2025]
Abstract
Acute stress has enduring effects on the brain and motivated behavior across species. For example, acute stress produces persisting decreases in voluntary physical activity as well as molecular changes in the striatum, a brain region that regulates voluntary physical activity and other motivated behaviors. Microglia, the primary immune cells of the central nervous system, are positioned at the interface between neural responses to stress and neural coordination of voluntary activity in that they respond to stress, sense molecular changes in the striatum, and modulate neuronal activity. However, the role of striatal microglia in stress-induced long-term suppression of voluntary activity is unknown. Here, we employ single-nucleus RNA sequencing to investigate how stress and exercise impact the biology of microglia in the striatum. We find that striatal microglia display altered activation profiles 6 weeks after an acute stressor. Furthermore, we show that access to a running wheel is associated with an additional and distinct microglial activation profile characterized by upregulation of genes related to complement components and phagocytosis pathways. Finally, we find that distinct gene sets show expression changes associated with general access to a running wheel versus variation in running levels. Taken together, our results deepen our understanding of the diverse molecular states that striatal microglia assume in response to stress and exercise and suggest that microglia exhibit a broader range of functional states than previously thought.
Collapse
Affiliation(s)
- Meghan G. Connolly
- Neuroscience ProgramUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Zachary V. Johnson
- Department of Psychiatry and Behavioral SciencesEmory UniversityAtlantaGeorgiaUSA
| | - Lynna Chu
- Department of StatisticsIowa State UniversityAmesIowaUSA
| | | | - Trevor J. Buhr
- Department of Food Science and Human NutritionIowa State UniversityAmesIowaUSA
- Neuroscience Graduate ProgramIowa State UniversityAmesIowaUSA
| | | | - Peter J. Clark
- Department of Food Science and Human NutritionIowa State UniversityAmesIowaUSA
- Neuroscience Graduate ProgramIowa State UniversityAmesIowaUSA
| | - Justin S. Rhodes
- Neuroscience ProgramUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Department of PsychologyUniversity of Illinois Urbana‐ChampaignChampaignIllinoisUSA
| |
Collapse
|
4
|
Soares AR, Garcia-Rivas V, Fai C, Thomas M, Zheng X, Picciotto MR, Mineur YS. Sex differences in the microglial response to stress and chronic alcohol exposure in mice. Biol Sex Differ 2025; 16:19. [PMID: 40038827 DOI: 10.1186/s13293-025-00701-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Women are more susceptible to stress-induced alcohol drinking, and preclinical data suggest that stress can increase alcohol intake in female rodents; however, a comprehensive understanding of the neurobiological processes underlying this sex difference is still emerging. Neuroimmune signaling, particularly by microglia, the brain's macrophages, is known to contribute to dysregulation of limbic circuits following stress and alcohol exposure. Females exhibit heightened immune reactivity, so we set out to characterize sex differences in the microglial response to stress and alcohol exposure. METHODS Male and female C57BL/6J mice were administered alcohol over 15 or 22 trials of a modified Drinking in the Dark paradigm, with repeated exposure to inescapable footshock stress and the stress-paired context. Mice were perfused immediately after drinking and we performed immunohistochemical analyses of microglial density, morphology, and protein expression in subregions of the amygdala and hippocampus. RESULTS We observed dynamic sex differences in microglial phenotypes at baseline and in response to stress and alcohol. Microglia in the hippocampus displayed more prominent sex differences and heightened reactivity to stress and alcohol. Chronic alcohol exposure decreased density of amygdala microglia and lysosomal expression. CONCLUSION We analyzed multiple measures of microglial activation, resulting in a comprehensive assessment of microglial changes mediated by sex, stress, and alcohol. These findings highlight the complexity of microglial contributions to the development of AUD and comorbid mood and stress disorders in men and women.
Collapse
Affiliation(s)
- Alexa R Soares
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
- Yale Interdepartmental Neuroscience Program, New Haven, CT, USA
| | - Vernon Garcia-Rivas
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Caroline Fai
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Merrilee Thomas
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Xiaoying Zheng
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Marina R Picciotto
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA.
- Yale Interdepartmental Neuroscience Program, New Haven, CT, USA.
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street - 3rd Floor Research, New Haven, CT, 06508, USA.
| | - Yann S Mineur
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| |
Collapse
|
5
|
Campanile AA, Eckel LA. Intermittent Overconsumption of High Fat Diet Promotes Microglial Reactivity in the Hypothalamus and Hindbrain of Female Rats. Cells 2025; 14:233. [PMID: 39937024 PMCID: PMC11817838 DOI: 10.3390/cells14030233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Elevated proinflammatory cytokines were reported in binge eating spectrum disorders characterized by intermittent overconsumption during periods of otherwise normal or restricted food intake. It is unknown whether binge eating promotes neuroinflammation, similar to that observed following chronic overconsumption of a high fat diet (HFD) in rodents. Here, we used a rodent model of binge-like eating to test the hypothesis that intermittent overconsumption of HFD promotes microglial reactivity in brain areas that control food intake. To promote overconsumption, one group of rats received chow plus intermittent access to HFD (INT). Control groups received either chow only (CHOW) or chow plus continuous access to HFD (CONT). Following behavioral testing, brains were processed to visualize ionized calcium-binding adaptor molecule 1 (Iba1), a microglial marker. INT rats consumed more calories than the control rats on days when the HFD was available, and fewer calories than the control rats on days when they only had access to chow. Despite consuming fewer total calories and 50% fewer fat calories, lean INT rats developed a pattern of microglial reactivity in feeding-relevant brain areas similar to obese CONT rats. We conclude that intermittent overconsumption of HFD, without diet-induced weight gain, promotes microglial reactivity in brain regions that control feeding.
Collapse
Affiliation(s)
- Alexis A. Campanile
- Program in Neuroscience, Department of Psychology, Florida State University, Tallahassee, FL 32304, USA;
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Lisa A. Eckel
- Program in Neuroscience, Department of Psychology, Florida State University, Tallahassee, FL 32304, USA;
| |
Collapse
|
6
|
Zhao R, Wang J, Chung SK, Xu B. New insights into anti-depression effects of bioactive phytochemicals. Pharmacol Res 2025; 212:107566. [PMID: 39746497 DOI: 10.1016/j.phrs.2024.107566] [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: 08/18/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
Depression is one of the most common psychological disorders, and due to its high prevalence and mortality rates, it imposes a significant disease burden. Contemporary treatments for depression involve various synthetic drugs, which have limitations such as side effects, single targets, and slow onset of action. Unlike synthetic medications, phytochemicals offer the benefits of a multi-target and multi-pathway mode of treatment for depression. In this literature review, we describe the pharmacological actions, experimental models, and clinical trials of the antidepressant effects of various phytochemicals. Additionally, we summarize the potential mechanisms by which these phytochemicals prevent depression, including regulating neurotransmitters and their receptors, the HPA axis, inflammatory responses, managing oxidative stress, neuroplasticity, and the gut microbiome. Phytochemicals exert therapeutic effects through multiple pathways and targets, making traditional Chinese medicine (TCM) a promising adjunctive antidepressant for the prevention, alleviation, and treatment of depression. Therefore, this review aims to provide robust evidence for subsequent research into developing phytochemical resources as effective antidepressant agents.
Collapse
Affiliation(s)
- Ruohan Zhao
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, Guangdong 519087, China
| | - Jingwen Wang
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, Guangdong 519087, China
| | - Sookja Kim Chung
- Faculty of Medicine, Macau University of Science and Technology, Macau, China.
| | - Baojun Xu
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, Guangdong 519087, China.
| |
Collapse
|
7
|
Murani E, Trakooljul N, Hadlich F, Wimmers K. Transcriptional signature of a hypersensitive glucocorticoid receptor variant in the neuroendocrine system suggests enhanced vulnerability to brain disorders. Brain Behav Immun 2025; 124:335-346. [PMID: 39674558 DOI: 10.1016/j.bbi.2024.12.004] [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: 05/27/2024] [Revised: 11/27/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024] Open
Abstract
The natural substitution Ala610Val in the porcine glucocorticoid receptor (GRAla610Val) leads to a profound compensatory downregulation of the hypothalamic-pituitary-adrenal (HPA) axis in early ontogeny. In this study, we leveraged this unique animal model to explore mechanisms of HPA axis regulation and consequences of its genetically-based persistent hypoactivity. To this end, we examined transcriptional signature of GRAla610Val in the hypothalamus, hippocampus, amygdala and adrenal gland in resting conditions (i.e. baseline glucocorticoid level) using mRNA sequencing. In addition, we studied transcriptome responses to two different doses of dexamethasone in the hypothalamus and hippocampus, depending on GRAla610Val. Across tissues, GRAla610Val consistently influenced the expression of several clustered protocadherins, particularly PCDHB7. Clustered protocadherins play an important role in neuronal connectivity and are implicated in different neurobiological disorders. Moreover, in line with our previous findings in blood immune cells, we found higher expression of pro-inflammatory genes, including canonical members of the TLR4 signaling pathway, in the brain of Val carriers. While the pro-inflammatory priming occurs already at resting conditions in the amygdala, in hypothalamus and hippocampus this seems to be associated with a stronger downregulation of several marker genes of homeostatic microglia, such as SALL1, by dexamethasone in Val carriers. Regarding the regulation of the HPA axis, GRAla610Val showed a dose-dependent effect on the central regulator of the axis, CRH, suggesting a dynamic adaptation to the glucocorticoid hypersensitivity of the Val variant. In the adrenal gland, GRAla610Val appears to downregulate cortisol production by impairing mitochondrial function. Overall, the transcriptional signature of GRAla610Val provides strong evidence that GR hypersensitivity leads to increased susceptibility to brain disorders.
Collapse
Affiliation(s)
- Eduard Murani
- Competence Field Genetics and Genomics, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| | - Nares Trakooljul
- Competence Field Genetics and Genomics, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Frieder Hadlich
- Competence Field Genetics and Genomics, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Klaus Wimmers
- Competence Field Genetics and Genomics, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| |
Collapse
|
8
|
Campos-Sánchez JC, Meseguer J, Guardiola FA. Fish microglia: Beyond the resident macrophages of the central nervous system - A review of their morphofunctional characteristics. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 162:105274. [PMID: 39341477 DOI: 10.1016/j.dci.2024.105274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
From classical to modern literature on microglia, the importance of the potential and variability of these immune cells in vertebrates has been pointed out. Recent aspects such as relationships and interactions between microglia and neurons in both normal and injured neural tissues, as well as their nexus with other organs and with the microbiota, or how these cells are modulated during development and adulthood are current topics of major interest. State-of-the-art research methodologies, including microscopy and potent in vivo imaging techniques, genomic and proteomic methods, current culture conditions together with the easy maintenance and manipulation of some fish embryos and adult specimens such as zebrafish (Danio rerio), have emerged and adapted to the phylogenetic position of some fish species. Furthermore, these advancements have facilitated the development of successful protocols aimed at addressing significant hypotheses and unresolved questions regarding vertebrate glia. The present review aims to analyse the available information on fish microglia, mainly the most recent one concerning teleosts, to establish an overview of their structural and immune functional features as a basis for their potentialities, heterogeneity, diversification, involvement, and relationships with neurons under normal and pathological conditions.
Collapse
Affiliation(s)
- Jose Carlos Campos-Sánchez
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - José Meseguer
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - Francisco A Guardiola
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain.
| |
Collapse
|
9
|
Nakagoshi N, Locatelli FM, Kitamura S, Hirota S, Kawano T. The impact of preoperative stress on age-related cognitive dysfunction after abdominal surgery: a study using a rat model. BMC Res Notes 2024; 17:369. [PMID: 39702453 DOI: 10.1186/s13104-024-07023-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] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024] Open
Abstract
OBJECTIVE This study examines the impact of preoperative stress on postoperative neuroinflammation and associated cognitive dysfunction, with a focus on aged individuals. The goal is to determine whether managing preoperative stress can enhance postoperative outcomes and lower the risk of cognitive impairment. RESULTS In aged rats, preoperative restraint stress significantly worsened neuroinflammation and cognitive deficits following abdominal surgery. Elevated levels of pro-inflammatory cytokines were observed in the hippocampus and medial prefrontal cortex two days post-surgery, and these effects persisted for twenty-eight days. In contrast, adult rats did not show significant changes in neuroinflammation or cognitive function due to preoperative restraint stress. An ex vivo analysis indicated that hippocampal microglia from aged rats exhibited an intensified proinflammatory response to lipopolysaccharide stimulation, further heightened by preoperative restraint stress. These findings suggest that managing preoperative stress could mitigate these adverse effects, leading to better postoperative recovery and cognitive health in elderly patients.
Collapse
Affiliation(s)
- Natsuki Nakagoshi
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Oko-cho, Kohasu, Nankoku, 783-8505, Kochi, Japan
| | - Fabricio M Locatelli
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Oko-cho, Kohasu, Nankoku, 783-8505, Kochi, Japan
| | - Sonoe Kitamura
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Oko-cho, Kohasu, Nankoku, 783-8505, Kochi, Japan
| | - Seiji Hirota
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Oko-cho, Kohasu, Nankoku, 783-8505, Kochi, Japan
| | - Takashi Kawano
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Oko-cho, Kohasu, Nankoku, 783-8505, Kochi, Japan.
| |
Collapse
|
10
|
Ontiveros-Ángel P, Vega-Torres JD, Simon TB, Williams V, Inostroza-Nives Y, Alvarado-Crespo N, Gonzalez YV, Pompolius M, Katzka W, Lou J, Sharafeddin F, De la Peña I, Dong T, Gupta A, Viet CT, Febo M, Obenaus A, Nair A, Figueroa JD. Early-life obesogenic environment integrates immunometabolic and epigenetic signatures governing neuroinflammation. Brain Behav Immun Health 2024; 42:100879. [PMID: 39430879 PMCID: PMC11490928 DOI: 10.1016/j.bbih.2024.100879] [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: 09/24/2024] [Accepted: 09/29/2024] [Indexed: 10/22/2024] Open
Abstract
Childhood overweight/obesity is associated with stress-related psychopathology, yet the pathways connecting childhood obesity to stress susceptibility are poorly understood. We employed a systems biology approach with 62 adolescent Lewis rats fed a Western-like high-saturated fat diet (WD, 41% kcal from fat) or a control diet (CD, 13% kcal from fat). A subset of rats underwent a 31-day model of predator exposures and social instability (PSS). Effects were assessed using behavioral tests, DTI (diffusion tensor imaging), NODDI (neurite orientation dispersion and density imaging), 16S rRNA gene sequencing for gut microbiome profiling, hippocampal microglia analysis, and targeted gene methylation. Parallel experiments on human microglia cells (HMC3) examined how palmitic acid influences cortisol-related inflammatory responses. Rats exposed to WD and PSS exhibited deficits in sociability, increased fear/anxiety-like behaviors, food consumption, and body weight. WD/PSS altered hippocampal microstructure (subiculum, CA1, dentate gyrus), and microbiome analysis showed a reduced abundance of members of the phylum Firmicutes. WD/PSS synergistically promoted neuroinflammatory changes in hippocampal microglia, linked with microbiome shifts and altered Fkbp5 expression/methylation. In HMC3, palmitate disrupted cortisol responses, affecting morphology, phagocytic markers, and cytokine release, partially mediated by FKBP5. This study identifies gene-environment interactions that influence microglia biology and may contribute to the connection between childhood obesity and stress-related psychopathology later in life.
Collapse
Affiliation(s)
- Perla Ontiveros-Ángel
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, USA
| | - Julio David Vega-Torres
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, USA
| | - Timothy B. Simon
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, USA
| | - Vivianna Williams
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, USA
| | - Yaritza Inostroza-Nives
- Department of Biochemistry and Pharmacology, San Juan Bautista School of Medicine, Caguas, Puerto Rico, USA
| | - Nashareth Alvarado-Crespo
- Department of Biochemistry and Pharmacology, San Juan Bautista School of Medicine, Caguas, Puerto Rico, USA
| | - Yarimar Vega Gonzalez
- Department of Biochemistry and Pharmacology, San Juan Bautista School of Medicine, Caguas, Puerto Rico, USA
| | - Marjory Pompolius
- Translational Research Imaging Laboratory, Department of Psychiatry, Department of Neuroscience, College of Medicine, University of Florida Health, Gainesville, FL, USA
| | - William Katzka
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA Microbiome Center, University of California, Los Angeles, CA, USA
| | - John Lou
- Loma Linda University Health School of Behavioral Health, Loma Linda, CA, USA
| | - Fransua Sharafeddin
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, USA
| | - Ike De la Peña
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA, USA
| | - Tien Dong
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA Microbiome Center, University of California, Los Angeles, CA, USA
| | - Arpana Gupta
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA Microbiome Center, University of California, Los Angeles, CA, USA
| | - Chi T. Viet
- Department of Oral & Maxillofacial Surgery, Loma Linda University Health School of Dentistry, Loma Linda, CA, USA
| | - Marcelo Febo
- Translational Research Imaging Laboratory, Department of Psychiatry, Department of Neuroscience, College of Medicine, University of Florida Health, Gainesville, FL, USA
| | - Andre Obenaus
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | - Aarti Nair
- Department of Psychology, Loma Linda University, Loma Linda, CA, USA
| | - Johnny D. Figueroa
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
11
|
Viola MF, Franco Taveras E, Mass E. Developmental programming of tissue-resident macrophages. Front Immunol 2024; 15:1475369. [PMID: 39575254 PMCID: PMC11578957 DOI: 10.3389/fimmu.2024.1475369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Macrophages are integral components of the innate immune system that colonize organs early in development and persist into adulthood through self-renewal. Their fate, whether they are replaced by monocytes or retain their embryonic origin, depends on tissue type and integrity. Macrophages are influenced by their environment, a phenomenon referred to as developmental programming. This influence extends beyond the local tissue microenvironment and includes soluble factors that can reach the macrophage niche. These factors include metabolites, antibodies, growth factors, and cytokines, which may originate from maternal diet, lifestyle, infections, or other developmental triggers and perturbations. These influences can alter macrophage transcriptional, epigenetic, and metabolic profiles, affecting cell-cell communication and tissue integrity. In addition to their crucial role in tissue immunity, macrophages play vital roles in tissue development and homeostasis. Consequently, developmental programming of these long-lived cells can modulate tissue physiology and pathology throughout life. In this review, we discuss the ontogeny of macrophages, the necessity of developmental programming by the niche for macrophage identity and function, and how developmental perturbations can affect the programming of macrophages and their subtissular niches, thereby influencing disease onset and progression in adulthood. Understanding these effects can inform targeted interventions or preventive strategies against diseases. Finally, understanding the consequences of developmental programming will shed light on how maternal health and disease may impact the well-being of future generations.
Collapse
Affiliation(s)
| | | | - Elvira Mass
- Developmental Biology of the Immune System, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
12
|
Xing WQ, Piao XJ, Han Q, Shi HY, Wu WC, Si F, Lu JJ, Zhou TZ, Guo JR, Li SZ, Xu B. SIRT2 regulates high mobility group protein B1 nucleoplasmic shuttle and degradation via deacetylation in microglia. J Cell Physiol 2024; 239:e31364. [PMID: 39129208 DOI: 10.1002/jcp.31364] [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: 03/02/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 08/13/2024]
Abstract
High mobility group protein B1 (HMGB1) acts as a pathogenic inflammatory response to mediate ranges of conditions such as epilepsy, septic shock, ischemia, traumatic brain injury, Parkinson's disease, Alzheimer's disease and mass spectrometry. HMGB1 promotes inflammation during sterile and infectious damage and plays a crucial role in disease development. Mobilization from the nucleus to the cytoplasm is the first important step in the release of HMGB1 from activated immune cells. Here, we demonstrated that Sirtuin 2 (SIRT2) physically interacts with and deacetylates HMGB1 at 43 lysine residue at nuclear localization signal locations, strengthening its interaction with HMGB1 and causing HMGB1 to be localized in the cytoplasm. These discoveries are the first to shed light on the SIRT2 nucleoplasmic shuttle, which influences HMGB1 and its degradation, hence revealing novel therapeutic targets and avenues for neuroinflammation treatment.
Collapse
Affiliation(s)
- Wan-Qun Xing
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Xian-Ji Piao
- The Fifth Affiliated Hospital of Harbin Medical University, Daqing, China
| | - Qi Han
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Hui-Ying Shi
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Wen-Cong Wu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Fan Si
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jing-Jing Lu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Tie-Zhong Zhou
- College of Animal Husbandry and Veterinary Medicine, Jinzhou Medical University, Jinzhou, China
| | - Jing-Ru Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shi-Ze Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Bin Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| |
Collapse
|
13
|
Burke MR, Sotiropoulos I, Waites CL. The multiple roles of chronic stress and glucocorticoids in Alzheimer's disease pathogenesis. Trends Neurosci 2024; 47:933-948. [PMID: 39307629 PMCID: PMC11563862 DOI: 10.1016/j.tins.2024.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/22/2024] [Accepted: 08/29/2024] [Indexed: 11/15/2024]
Abstract
Chronic stress and the accompanying long-term elevation of glucocorticoids (GCs), the stress hormones of the body, increase the risk and accelerate the progression of Alzheimer's disease (AD). Signatures of AD include intracellular tau (MAPT) tangles, extracellular amyloid β (Aβ) plaques, and neuroinflammation. A growing body of work indicates that stress and GCs initiate cellular processes underlying these pathologies through dysregulation of protein homeostasis and trafficking, mitochondrial bioenergetics, and response to damage-associated stimuli. In this review, we integrate findings from mechanistic studies in rodent and cellular models, wherein defined chronic stress protocols or GC administration have been shown to elicit AD-related pathology. We specifically discuss the effects of chronic stress and GCs on tau pathogenesis, including hyperphosphorylation, aggregation, and spreading, amyloid precursor protein (APP) processing and trafficking culminating in Aβ production, immune priming by proinflammatory cytokines and disease-associated molecular patterns, and alterations to glial cell and blood-brain barrier (BBB) function.
Collapse
Affiliation(s)
- Mia R Burke
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and Aging Brain, Columbia University Irving Medical Center, New York, NY, USA; Pathobiology and Mechanisms of Disease Graduate Program, Columbia University Irving Medical Center, New York, NY, USA
| | - Ioannis Sotiropoulos
- Institute of Biosciences and Applications, National Centre for Scientific Research (NCSR) Demokritos, Agia Paraskevi, Greece
| | - Clarissa L Waites
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and Aging Brain, Columbia University Irving Medical Center, New York, NY, USA; Department of Neuroscience, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
14
|
Aguilar-Delgadillo A, Cruz-Mendoza F, Luquin-de Andais teh S, Ruvalcaba-Delgadillo Y, Jáuregui-Huerta F. Stress-induced c-fos expression in the medial prefrontal cortex differentially affects the main residing cell phenotypes. Heliyon 2024; 10:e39325. [PMID: 39498004 PMCID: PMC11532284 DOI: 10.1016/j.heliyon.2024.e39325] [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: 10/04/2023] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 11/07/2024] Open
Abstract
Stress poses a challenge to the body's equilibrium and triggers a series of responses that enable organisms to adapt to stressful stimuli. The medial prefrontal cortex (mPFC), particularly in acute stress conditions, undergoes significant physiological changes to cope with the demands associated with cellular activation. The proto-oncogene c-fos and its protein product c-Fos have long been utilized to investigate the effects of external factors on the central nervous system (CNS). While c-Fos expression has traditionally been attributed to neurons, emerging evidence suggests its potential expression in glial cells. In this study, our main objective was to explore the expression of c-Fos in glial cells and examine how acute stress influences these activity patterns. We conducted our experiments on male Wistar rats, subjecting them to acute stress and sacrificing them 2 h after the stressor initiation. Using double-labelling fluorescent immunohistochemistry targeting c-Fos, along with markers such as GFAP, Iba-1, Olig2, NG2, and NeuN, we analyzed 35 μm brain slices obtained from the mPFC. Our findings compellingly demonstrate that c-Fos expression extends beyond neurons and is present in astrocytes, oligodendrocytes, microglia, and NG2 cells-the diverse population of glial cells. Moreover, we observed distinct regulation of c-Fos expression in response to stress across different subregions of the mPFC. These results emphasize the importance of considering glial cells and their perspective in studies investigating brain activity, highlighting c-Fos as a response marker in glial cells. By shedding light on the differential regulation of c-Fos expression in response to stress, our study contributes to the understanding of glial cell involvement in stress-related processes. This underscores the significance of including glial cells in investigations of brain activity and expands our knowledge of c-Fos as a potential marker for glial cell responses.
Collapse
Affiliation(s)
| | - Fernando Cruz-Mendoza
- Neurosciences Department, Health sciences center, University of Guadalajara, Guadalajara, Mexico
| | | | | | - Fernando Jáuregui-Huerta
- Neurosciences Department, Health sciences center, University of Guadalajara, Guadalajara, Mexico
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| |
Collapse
|
15
|
Bonomi R, Hillmer AT, Woodcock E, Bhatt S, Rusowicz A, Angarita GA, Carson RE, Davis MT, Esterlis I, Nabulsi N, Huang Y, Krystal JH, Pietrzak RH, Cosgrove KP. Microglia-mediated neuroimmune suppression in PTSD is associated with anhedonia. Proc Natl Acad Sci U S A 2024; 121:e2406005121. [PMID: 39172786 PMCID: PMC11363315 DOI: 10.1073/pnas.2406005121] [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: 03/26/2024] [Accepted: 07/16/2024] [Indexed: 08/24/2024] Open
Abstract
Dynamic brain immune function in individuals with posttraumatic stress disorder is rarely studied, despite evidence of peripheral immune dysfunction. Positron emission tomography brain imaging using the radiotracer [11C]PBR28 was used to measure the 18-kDa translocator protein (TSPO), a microglial marker, at baseline and 3 h after administration of lipopolysaccharide (LPS), a potent immune activator. Data were acquired in 15 individuals with PTSD and 15 age-matched controls. The PTSD group exhibited a significantly lower magnitude LPS-induced increase in TSPO availability in an a priori prefrontal-limbic circuit compared to controls. Greater anhedonic symptoms in the PTSD group were associated with a more suppressed neuroimmune response. In addition, while a reduced granulocyte-macrophage colony-stimulating factor response to LPS was observed in the PTSD group, other measured cytokine responses and self-reported sickness symptoms did not differ between groups; these findings highlight group differences in central-peripheral immune system relationships. The results of this study provide evidence of a suppressed microglia-mediated neuroimmune response to a direct immune system insult in individuals with PTSD that is associated with the severity of symptoms. They also provide further support to an emerging literature challenging traditional concepts of microglial and immune function in psychiatric disease.
Collapse
Affiliation(s)
- Robin Bonomi
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
| | - Ansel T. Hillmer
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT06520
- Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, CT06519
| | - Eric Woodcock
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
| | - Shivani Bhatt
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
| | | | | | - Richard E. Carson
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT06520
- Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, CT06519
| | - Margaret T. Davis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, Veterans Affairs Connecticut Healthcare System, West Haven, CT06516
| | - Irina Esterlis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT06520
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, Veterans Affairs Connecticut Healthcare System, West Haven, CT06516
| | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT06520
- Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, CT06519
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT06520
- Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, CT06519
| | - John H. Krystal
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, Veterans Affairs Connecticut Healthcare System, West Haven, CT06516
| | - Robert H. Pietrzak
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, Veterans Affairs Connecticut Healthcare System, West Haven, CT06516
| | - Kelly P. Cosgrove
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT06520
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, Veterans Affairs Connecticut Healthcare System, West Haven, CT06516
| |
Collapse
|
16
|
Joseph A, Baslet G, O'Neal MA, Polich G, Gonsalvez I, Christoforou AN, Dworetzky BA, Spagnolo PA. Prevalence of autoimmune diseases in functional neurological disorder: influence of psychiatric comorbidities and biological sex. J Neurol Neurosurg Psychiatry 2024; 95:865-869. [PMID: 38514177 DOI: 10.1136/jnnp-2023-332825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Functional neurological disorder (FND) is a common and disabling neuropsychiatric condition, which disproportionally affects women compared with men. While the etiopathogenesis of this disorder remains elusive, immune dysregulation is emerging as one potential mechanism. To begin to understand the role of immune dysfunctions in FND, we assessed the prevalence of several common autoimmune diseases (ADs) in a large cohort of patients with FND and examined the influence of psychiatric comorbidities and biological sex. METHODS Using a large biorepository database (Mass General Brigham Biobank), we obtained demographic and clinical data of a cohort of 643 patients diagnosed with FND between January 2015 and December 2021. The proportion of ADs was calculated overall, by sex and by the presence of psychiatric comorbidities. RESULTS The overall prevalence of ADs in our sample was 41.9%, with connective tissue and autoimmune endocrine diseases being the most commonly observed ADs. Among patients with FND and ADs, 27.7% had ≥2 ADs and 8% met criteria for multiple autoimmune syndrome. Rates of ADs were significantly higher in subjects with comorbid major depressive disorder and post-traumatic stress disorder (p= 0.02). Women represented the largest proportion of patients with concurrent ADs, both in the overall sample and in the subgroups of interest (p's < 0.05). CONCLUSIONS This study is unique in providing evidence of an association between FND and ADs. Future studies are needed to investigate the mechanisms underlying this association and to understand whether FND is characterised by distinct dysregulations in immune response.
Collapse
Affiliation(s)
- Anna Joseph
- Psychiatry, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Gaston Baslet
- Psychiatry, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Mary A O'Neal
- Harvard Medical School, Boston, Massachusetts, USA
- Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Ginger Polich
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Boston, Boston, Massachusetts, USA
| | - Irene Gonsalvez
- Psychiatry, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Andrea N Christoforou
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Boston, Boston, Massachusetts, USA
| | - Barbara A Dworetzky
- Harvard Medical School, Boston, Massachusetts, USA
- Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Primavera A Spagnolo
- Psychiatry, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Zhang M, Liang C, Chen X, Cai Y, Cui L. Interplay between microglia and environmental risk factors in Alzheimer's disease. Neural Regen Res 2024; 19:1718-1727. [PMID: 38103237 PMCID: PMC10960290 DOI: 10.4103/1673-5374.389745] [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: 06/14/2023] [Revised: 09/09/2023] [Accepted: 10/24/2023] [Indexed: 12/18/2023] Open
Abstract
Alzheimer's disease, among the most common neurodegenerative disorders, is characterized by progressive cognitive impairment. At present, the Alzheimer's disease main risk remains genetic risks, but major environmental factors are increasingly shown to impact Alzheimer's disease development and progression. Microglia, the most important brain immune cells, play a central role in Alzheimer's disease pathogenesis and are considered environmental and lifestyle "sensors." Factors like environmental pollution and modern lifestyles (e.g., chronic stress, poor dietary habits, sleep, and circadian rhythm disorders) can cause neuroinflammatory responses that lead to cognitive impairment via microglial functioning and phenotypic regulation. However, the specific mechanisms underlying interactions among these factors and microglia in Alzheimer's disease are unclear. Herein, we: discuss the biological effects of air pollution, chronic stress, gut microbiota, sleep patterns, physical exercise, cigarette smoking, and caffeine consumption on microglia; consider how unhealthy lifestyle factors influence individual susceptibility to Alzheimer's disease; and present the neuroprotective effects of a healthy lifestyle. Toward intervening and controlling these environmental risk factors at an early Alzheimer's disease stage, understanding the role of microglia in Alzheimer's disease development, and targeting strategies to target microglia, could be essential to future Alzheimer's disease treatments.
Collapse
Affiliation(s)
- Miaoping Zhang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, China
| | - Chunmei Liang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, China
| | - Xiongjin Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, China
| | - Yujie Cai
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, China
| | - Lili Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, China
| |
Collapse
|
18
|
Dar W. Aspartame-induced cognitive dysfunction: Unveiling role of microglia-mediated neuroinflammation and molecular remediation. Int Immunopharmacol 2024; 135:112295. [PMID: 38776852 DOI: 10.1016/j.intimp.2024.112295] [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: 03/04/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
Aspartame, an artificial sweetener, is consumed by millions of people globally. There are multiple reports of aspartame and its metabolites affecting cognitive functions in animal models and humans, which include learning problems, headaches, seizures, migraines, irritable moods, anxiety, depression, and insomnia. These cognitive deficits and associated symptoms are partly attributed to dysregulated excitatory and inhibitory neurotransmitter balance due to aspartate released from aspartame, resulting in an excitotoxic effect in neurons, leading to neuronal damage. However, microglia, a central immunocompetent cell type in brain tissue and a significant player in inflammation can contribute to the impact. Microglia rapidly respond to changes in CNS homeostasis. Aspartame consumption might affect the microglia phenotype directly via methanol-induced toxic effects and indirectly via aspartic acid-mediated excitotoxicity, exacerbating symptoms of cognitive decline. Long-term oral consumption of aspartame thus might change microglia's phenotype from ramified to activated, resulting in chronic or sustained activation, releasing excess pro-inflammatory molecules. This pro-inflammatory surge might lead to the degeneration of healthy neurons and other glial cells, impairing cognition. This review will deliberate on possible links and research gaps that need to be explored concerning aspartame consumption, ecotoxicity and microglia-mediated inflammatory cognitive impairment. The study covers a comprehensive analysis of the impact of aspartame consumption on cognitive function, considering both direct and indirect effects, including the involvement of microglia-mediated neuroinflammation. We also propose a novel intervention strategy involving tryptophan supplementation to mitigate cognitive decline symptoms in individuals with prolonged aspartame consumption, providing a potential solution to address the adverse effects of aspartame on cognitive function.
Collapse
Affiliation(s)
- Waseem Dar
- Translational Neurobiology and Disease Modelling Laboratory, Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence, Greater Noida, 201314, India.
| |
Collapse
|
19
|
Fanikos M, Kohn SA, Stamato R, Brenhouse HC, Gildawie KR. Impacts of age and environment on postnatal microglial activity: Consequences for cognitive function following early life adversity. PLoS One 2024; 19:e0306022. [PMID: 38917075 PMCID: PMC11198844 DOI: 10.1371/journal.pone.0306022] [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: 04/02/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
Early life adversity (ELA) increases the likelihood of later-life neuropsychiatric disorders and cognitive dysfunction. Importantly, ELA, neuropsychiatric disorders, and cognitive deficits all involve aberrant immune signaling. Microglia are the primary neuroimmune cells and regulate brain development. Microglia are particularly sensitive to early life insults, which can program their responses to future challenges. ELA in the form of maternal separation (MS) in rats alters later-life microglial morphology and the inflammatory profile of the prefrontal cortex, a region important for cognition. However, the role of microglial responses during MS in the development of later cognition is not known. Therefore, here we aimed to determine whether the presence of microglia during MS mediates long-term impacts on adult working memory. Clodronate liposomes were used to transiently deplete microglia from the brain, while empty liposomes were used as a control. We hypothesized that if microglia mediate the long-term impacts of ELA on working memory in adulthood, then depleting microglia during MS would prevent these deficits. Importantly, microglial function shifts throughout the neonatal period, so an exploratory investigation assessed whether depletion during the early versus late neonatal period had different effects on adult working memory. Surprisingly, empty liposome treatment during the early, but not late, postnatal period induced microglial activity changes that compounded with MS to impair working memory in females. In contrast, microglial depletion later in infancy impaired later life working memory in females, suggesting that microglial function during late infancy plays an important role in the development of cognitive function. Together, these findings suggest that microglia shift their sensitivity to early life insults across development. Our findings also highlight the potential for MS to impact some developmental processes only when compounded with additional neuroimmune challenges in a sex-dependent manner.
Collapse
Affiliation(s)
- Michaela Fanikos
- Department of Psychology, Northeastern University, Boston, Massachusetts, United States of America
| | - Skylar A. Kohn
- Department of Psychology, Northeastern University, Boston, Massachusetts, United States of America
| | - Rebecca Stamato
- Department of Psychology, Northeastern University, Boston, Massachusetts, United States of America
| | - Heather C. Brenhouse
- Department of Psychology, Northeastern University, Boston, Massachusetts, United States of America
| | - Kelsea R. Gildawie
- Department of Psychology, Northeastern University, Boston, Massachusetts, United States of America
| |
Collapse
|
20
|
Soares AR, Garcia-Rivas V, Fai C, Thomas MA, Zheng X, Picciotto MR, Mineur YS. Role of microglia in stress-induced alcohol intake in female and male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597614. [PMID: 38895217 PMCID: PMC11185719 DOI: 10.1101/2024.06.05.597614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Rates of alcohol use disorder (AUD) have escalated in recent years, with a particular increase among women. Women are more susceptible to stress-induced alcohol drinking, and preclinical data suggest that stress can increase alcohol intake in female rodents; however, a comprehensive understanding of sex-specific neurobiological substrates underlying this phenomenon is still emerging. Microglia, the resident macrophages of the brain, are essential for reshaping neuronal processes, and microglial activity contributes to overall neuronal plasticity. We investigated microglial dynamics and morphology in limbic brain structures of male and female mice following exposure to stress, alcohol or both challenges. In a modified paradigm of intermittent binge drinking (repeated "drinking in the dark"), we determined that female, but not male, mice increased their alcohol consumption after exposure to a physical stressor and re-exposure trials in the stress-paired context. Ethanol (EtOH) drinking and stress altered a number of microglial parameters, including overall number, in subregions of the amygdala and hippocampus, with effects that were somewhat more pronounced in female mice. We used the CSF1R antagonist PLX3397 to deplete microglia in female mice to determine whether microglia contribute to stress-induced escalation of EtOH intake. We observed that microglial depletion attenuated stress-induced alcohol intake with no effect in the unstressed group. These findings suggest that microglial activity can contribute to alcohol intake under stressful conditions, and highlight the importance of evaluating sex-specific mechanisms that could result in tailored interventions for AUD in women.
Collapse
|
21
|
Valenza M, Facchinetti R, Torazza C, Ciarla C, Bronzuoli MR, Balbi M, Bonanno G, Popoli M, Steardo L, Milanese M, Musazzi L, Bonifacino T, Scuderi C. Molecular signatures of astrocytes and microglia maladaptive responses to acute stress are rescued by a single administration of ketamine in a rodent model of PTSD. Transl Psychiatry 2024; 14:209. [PMID: 38796504 PMCID: PMC11127980 DOI: 10.1038/s41398-024-02928-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/28/2024] Open
Abstract
Stress affects the brain and alters its neuroarchitecture and function; these changes can be severe and lead to psychiatric disorders. Recent evidence suggests that astrocytes and microglia play an essential role in the stress response by contributing to the maintenance of cerebral homeostasis. These cells respond rapidly to all stimuli that reach the brain, including stressors. Here, we used a recently validated rodent model of post-traumatic stress disorder in which rats can be categorized as resilient or vulnerable after acute inescapable footshock stress. We then investigated the functional, molecular, and morphological determinants of stress resilience and vulnerability in the prefrontal cortex, focusing on glial and neuronal cells. In addition, we examined the effects of a single subanesthetic dose of ketamine, a fast-acting antidepressant recently approved for the treatment of resistant depression and proposed for other stress-related psychiatric disorders. The present results suggest a prompt glial cell response and activation of the NF-κB pathway after acute stress, leading to an increase in specific cytokines such as IL-18 and TNF-α. This response persists in vulnerable individuals and is accompanied by a significant change in the levels of critical glial proteins such as S100B, CD11b, and CX43, brain trophic factors such as BDNF and FGF2, and proteins related to dendritic arborization and synaptic architecture such as MAP2 and PSD95. Administration of ketamine 24 h after the acute stress event rescued many of the changes observed in vulnerable rats, possibly contributing to support brain homeostasis. Overall, our results suggest that pivotal events, including reactive astrogliosis, changes in brain trophic factors, and neuronal damage are critical determinants of vulnerability to acute traumatic stress and confirm the therapeutic effect of acute ketamine against the development of stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Marta Valenza
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Roberta Facchinetti
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Carola Torazza
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Claudia Ciarla
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Maria Rosanna Bronzuoli
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Matilde Balbi
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Maurizio Popoli
- Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy
| | - Luca Steardo
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Marco Milanese
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Laura Musazzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Caterina Scuderi
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy.
| |
Collapse
|
22
|
Kozlowska K, Scher S. Recent advances in understanding the neurobiology of pediatric functional neurological disorder. Expert Rev Neurother 2024; 24:497-516. [PMID: 38591353 DOI: 10.1080/14737175.2024.2333390] [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: 05/26/2023] [Accepted: 03/18/2024] [Indexed: 04/10/2024]
Abstract
INTRODUCTION Functional neurological disorder (FND) is a neuropsychiatric disorder that manifests in a broad array of functional motor, sensory, or cognitive symptoms, which arise from complex interactions between brain, mind, body, and context. Children with FND make up 10%-20% of presentations to neurology services in children's hospitals and up to 20% of adolescents admitted to hospital for the management of intractable seizures. AREAS COVERED The current review focuses on the neurobiology of pediatric FND. The authors present an overview of the small but growing body of research pertaining to the biological, emotion-processing, cognitive, mental health, physical health, and social system levels. EXPERT OPINION Emerging research suggests that pediatric FND is underpinned by aberrant changes within and between neuron-glial (brain) networks, with a variety of factors - on multiple system levels - contributing to brain network changes. In pediatric practice, adverse childhood experiences (ACEs) are commonly reported, and activation or dysregulation of stress-system components is a frequent finding. Our growing understanding of the neurobiology of pediatric FND has yielded important flow-on effects for assessing and diagnosing FND, for developing targeted treatment interventions, and for improving the treatment outcomes of children and adolescents with FND.
Collapse
Affiliation(s)
- Kasia Kozlowska
- The Children's Hospital at Westmead, Westmead, NSW, Australia
- Brain Dynamics Centre, Westmead Institute of Medical Research, Westmead, NSW, Australia
- University of Sydney Medical School, Camperdown, NSW, Australia
| | - Stephen Scher
- University of Sydney Medical School, Camperdown, NSW, Australia
- Department of Psychiatry, Harvard Medical School, Belmont, MA, USA
- McLean Hospital, Belmont, MA, USA
| |
Collapse
|
23
|
Chang J, Jiang T, Shan X, Zhang M, Li Y, Qi X, Bian Y, Zhao L. Pro-inflammatory cytokines in stress-induced depression: Novel insights into mechanisms and promising therapeutic strategies. Prog Neuropsychopharmacol Biol Psychiatry 2024; 131:110931. [PMID: 38176531 DOI: 10.1016/j.pnpbp.2023.110931] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/12/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
Stress-mediated depression is one of the common psychiatric disorders with a high prevalence and suicide rate, there is a lack of effective treatment. Accordingly, effective treatments with few adverse effects are urgently needed. Pro-inflammatory cytokines (PICs) may play a key role in stress-mediated depression. Thereupon, both preclinical and clinical studies have found higher levels of IL-1β, TNF-α and IL-6 in peripheral blood and brain tissue of patients with depression. Recent studies have found PICs cause depression by affecting neuroinflammation, monoamine neurotransmitters, hypothalamic pituitary adrenal axis and neuroplasticity. Moreover, they play an important role in the symptom, development and progression of depression, maybe a potential diagnostic and therapeutic marker of depression. In addition, well-established antidepressant therapies have some relief on high levels of PICs. Importantly, anti-inflammatory drugs relieve depressive symptoms by reducing levels of PICs. Collectively, reducing PICs may represent a promising therapeutic strategy for depression.
Collapse
Affiliation(s)
- Jun Chang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Tingcan Jiang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaoqian Shan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Mingxing Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yujiao Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xin Qi
- Department of Cardiology, Tianjin Union Medical Center, 300121, China
| | - Yuhong Bian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Lan Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
24
|
Da Silva DE, Richards CM, McRae SA, Riar I, Yang S(S, Zurfluh NE, Gibon J, Klegeris A. Extracellular mixed histones are neurotoxic and modulate select neuroimmune responses of glial cells. PLoS One 2024; 19:e0298748. [PMID: 38630734 PMCID: PMC11023449 DOI: 10.1371/journal.pone.0298748] [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: 05/15/2023] [Accepted: 01/29/2024] [Indexed: 04/19/2024] Open
Abstract
Although histone proteins are widely known for their intranuclear functions where they organize DNA, all five histone types can also be released into the extracellular space from damaged cells. Extracellular histones can interact with pattern recognition receptors of peripheral immune cells, including toll-like receptor 4 (TLR4), causing pro-inflammatory activation, which indicates they may act as damage-associated molecular patterns (DAMPs) in peripheral tissues. Very limited information is available about functions of extracellular histones in the central nervous system (CNS). To address this knowledge gap, we applied mixed histones (MH) to cultured cells modeling neurons, microglia, and astrocytes. Microglia are the professional CNS immunocytes, while astrocytes are the main support cells for neurons. Both these cell types are critical for neuroimmune responses and their dysregulated activity contributes to neurodegenerative diseases. We measured effects of extracellular MH on cell viability and select neuroimmune functions of microglia and astrocytes. MH were toxic to cultured primary murine neurons and also reduced viability of NSC-34 murine and SH-SY5Y human neuron-like cells in TLR4-dependent manner. MH did not affect the viability of resting or immune-stimulated BV-2 murine microglia or U118 MG human astrocytic cells. When applied to BV-2 cells, MH enhanced secretion of the potential neurotoxin glutamate, but did not modulate the release of nitric oxide (NO), tumor necrosis factor-α (TNF), C-X-C motif chemokine ligand 10 (CXCL10), or the overall cytotoxicity of lipopolysaccharide (LPS)- and/or interferon (IFN)-γ-stimulated BV-2 microglial cells towards NSC-34 neuron-like cells. We demonstrated, for the first time, that MH downregulated phagocytic activity of LPS-stimulated BV-2 microglia. However, MH also exhibited protective effect by ameliorating the cytotoxicity of LPS-stimulated U118 MG astrocytic cells towards SH-SY5Y neuron-like cells. Our data demonstrate extracellular MH could both damage neurons and alter neuroimmune functions of glial cells. These actions of MH could be targeted for treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Dylan E. Da Silva
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| | - Christy M. Richards
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| | - Seamus A. McRae
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| | - Ishvin Riar
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| | - Sijie (Shirley) Yang
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| | - Noah E. Zurfluh
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| | - Julien Gibon
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, University Way, Kelowna, British Columbia, Canada
| |
Collapse
|
25
|
Traetta ME, Chaves Filho AM, Akinluyi ET, Tremblay MÈ. Neurodevelopmental and Neuropsychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2024; 37:457-495. [PMID: 39207708 DOI: 10.1007/978-3-031-55529-9_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
This chapter will focus on microglial involvement in neurodevelopmental and neuropsychiatric disorders, particularly autism spectrum disorder (ASD), schizophrenia and major depressive disorder (MDD). We will describe the neuroimmune risk factors that contribute to the etiopathology of these disorders across the lifespan, including both in early life and adulthood. Microglia, being the resident immune cells of the central nervous system, could play a key role in triggering and determining the outcome of these disorders. This chapter will review preclinical and clinical findings where microglial morphology and function were examined in the contexts of ASD, schizophrenia and MDD. Clinical evidence points out to altered microglial morphology and reactivity, as well as increased expression of pro-inflammatory cytokines, supporting the idea that microglial abnormalities are involved in these disorders. Indeed, animal models for these disorders found altered microglial morphology and homeostatic functions which resulted in behaviours related to these disorders. Additionally, as microglia have emerged as promising therapeutic targets, we will also address in this chapter therapies involving microglial mechanisms for the treatment of neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
| | | | - Elizabeth Toyin Akinluyi
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology and Therapeutics, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Département de Médecine Moléculaire, Université Laval, Quebec City, QC, Canada.
- Axe Neurosciences, Center de Recherche du CHU de Québec, Université Laval, Quebec City, QC, Canada.
- Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
26
|
Chen K, Qi X, Zhu LL, Li ML, Cong B, Li YM. Quantitative analysis of microglia morphological changes in the hypothalamus of chronically stressed rats. Brain Res Bull 2024; 206:110861. [PMID: 38141789 DOI: 10.1016/j.brainresbull.2023.110861] [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: 06/19/2023] [Revised: 12/05/2023] [Accepted: 12/20/2023] [Indexed: 12/25/2023]
Abstract
Based on the successful establishment of a rat model of chronic restraint stress, we used multiple algorithms to quantify the morphological changes of rat hypothalamic microglia from various perspectives, providing a pathomorphological basis for the subsequent study of molecular mechanisms of hypothalamic stress injury, such as neuroinflammation. To verify the successful establishment of the chronic stress model, an enzyme-linked immunosorbent assay was performed to detect serum glucocorticoid levels. Microglia labeled with Iba1 in frozen sections of rat hypothalamus were scanned and photographed at multiple levels using confocal microscopy. Subsequently, images were processed for external contouring and skeletonization, and morphological indices of microglia were calculated and analyzed using fractal, skeleton, and Sholl analysis. In addition, the co-expression of CD68 (a marker that can reflect phagocytic activity) and Iba1 was observed by immunofluorescence technique. Compared with the control group, microglia in the chronic stress group displayed reduced fractal dimension and lacunarity, increased density and circularity, enlarged soma areas, and shortened and reduced branches. Sholl analysis confirmed the reduced complexity of microglia following chronic stress. Meanwhile, microglia CD68 increased significantly, indicating that the microglia in the chronic stress group have greater phagocytosis activity. In summary, chronic restraint stress promoted the conversion of microglia in the rat hypothalamus to a less complex form, manifested as larger soma, shorter and fewer branches, more uniform and dense texture, and increased circularity; indeed, the shape of these microglia resembled that of amoeba and they displayed strong phagocytosis activity.
Collapse
Affiliation(s)
- Ke Chen
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, No.361 Zhongshan Dong Road, 050017 Shijiazhuang, China
| | - Xin Qi
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, No.361 Zhongshan Dong Road, 050017 Shijiazhuang, China
| | - Lin-Lin Zhu
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, No.361 Zhongshan Dong Road, 050017 Shijiazhuang, China
| | - Mei-Li Li
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, No.361 Zhongshan Dong Road, 050017 Shijiazhuang, China
| | - Bin Cong
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, No.361 Zhongshan Dong Road, 050017 Shijiazhuang, China.
| | - Ying-Min Li
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Hebei Medical University, No.361 Zhongshan Dong Road, 050017 Shijiazhuang, China.
| |
Collapse
|
27
|
Gore IR, Gould E. Developmental and adult stress: effects of steroids and neurosteroids. Stress 2024; 27:2317856. [PMID: 38563163 PMCID: PMC11046567 DOI: 10.1080/10253890.2024.2317856] [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: 07/03/2023] [Accepted: 02/03/2024] [Indexed: 04/04/2024] Open
Abstract
In humans, exposure to early life adversity has profound implications for susceptibility to developing neuropsychiatric disorders later in life. Studies in rodents have shown that stress experienced during early postnatal life can have lasting effects on brain development. Glucocorticoids and sex steroids are produced in endocrine glands and the brain from cholesterol; these molecules bind to nuclear and membrane-associated steroid receptors. Unlike other steroids that can also be made in the brain, neurosteroids bind specifically to neurotransmitter receptors, not steroid receptors. The relationships among steroids, neurosteroids, and stress are multifaceted and not yet fully understood. However, studies demonstrating altered levels of progestogens, androgens, estrogens, glucocorticoids, and their neuroactive metabolites in both developmental and adult stress paradigms strongly suggest that these molecules may be important players in stress effects on brain circuits and behavior. In this review, we discuss the influence of developmental and adult stress on various components of the brain, including neurons, glia, and perineuronal nets, with a focus on sex steroids and neurosteroids. Gaining an enhanced understanding of how early adversity impacts the intricate systems of brain steroid and neurosteroid regulation could prove instrumental in identifying novel therapeutic targets for stress-related conditions.
Collapse
Affiliation(s)
- Isha R Gore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Elizabeth Gould
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| |
Collapse
|
28
|
Coleman PT, Costanza-Chavez GW, Martin HN, Amat J, Frank MG, Sanchez RJ, Potter GJ, Mellert SM, Carter RK, Bonnici GN, Maier SF, Baratta MV. Prior experience with behavioral control over stress facilitates social dominance. Neurobiol Stress 2024; 28:100597. [PMID: 38213318 PMCID: PMC10783635 DOI: 10.1016/j.ynstr.2023.100597] [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: 06/13/2023] [Revised: 11/13/2023] [Accepted: 12/01/2023] [Indexed: 01/13/2024] Open
Abstract
Dominance status has extensive effects on physical and mental health, and an individual's relative position can be shaped by experiential factors. A variety of considerations suggest that the experience of behavioral control over stressors should produce winning in dominance tests and that winning should blunt the impact of later stressors, as does prior control. To investigate the interplay between competitive success and stressor control, we first examined the impact of stressor controllability on subsequent performance in a warm spot competition test modified for rats. Prior experience of controllable, but not physically identical uncontrollable, stress increased later effortful behavior and occupation of the warm spot. Controllable stress subjects consistently ranked higher than did uncontrollable stress subjects. Pharmacological inactivation of the prelimbic (PL) cortex during behavioral control prevented later facilitation of dominance. Next, we explored whether repeated winning experiences produced later resistance against the typical sequelae of uncontrollable stress. To establish dominance status, triads of rats were given five sessions of warm spot competition. The development of stable dominance was prevented by reversible inactivation of the PL or NMDA receptor blockade in the dorsomedial striatum. Stable winning blunted the later stress-induced increase in dorsal raphe nucleus serotonergic activity, as well as prevented uncontrollable stress-induced social avoidance. In contrast, endocrine and neuroimmune responses to uncontrollable stress were unaffected, indicating a selective impact of prior dominance. Together, these data demonstrate that instrumental control over stress promotes later dominance, but also reveal that winning experiences buffer against the neural and behavioral outcomes of future adversity.
Collapse
Affiliation(s)
| | | | - Heather N. Martin
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, USA
| | - Jose Amat
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, USA
| | - Matthew G. Frank
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, USA
| | - Rory J. Sanchez
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, USA
| | - Garrett J. Potter
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, USA
| | - Simone M. Mellert
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, USA
| | - Rene K. Carter
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, USA
| | - Gianni N. Bonnici
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, USA
| | - Steven F. Maier
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, USA
| | - Michael V. Baratta
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, USA
| |
Collapse
|
29
|
Charney M, Foster S, Shukla V, Zhao W, Jiang SH, Kozlowska K, Lin A. Neurometabolic alterations in children and adolescents with functional neurological disorder. Neuroimage Clin 2023; 41:103557. [PMID: 38219534 PMCID: PMC10825645 DOI: 10.1016/j.nicl.2023.103557] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/22/2023] [Accepted: 12/18/2023] [Indexed: 01/16/2024]
Abstract
OBJECTIVES In vivo magnetic resonance spectroscopy (MRS) was used to investigate neurometabolic homeostasis in children with functional neurological disorder (FND) in three regions of interest: supplementary motor area (SMA), anterior default mode network (aDMN), and posterior default mode network (dDMN). Metabolites assessed included N-acetyl aspartate (NAA), a marker of neuron function; myo-inositol (mI), a glial-cell marker; choline (Cho), a membrane marker; glutamate plus glutamine (Glx), a marker of excitatory neurotransmission; γ-aminobutyric acid (GABA), a marker of inhibitor neurotransmission; and creatine (Cr), an energy marker. The relationship between excitatory (glutamate and glutamine) and inhibitory (GABA) neurotransmitter (E/I) balance was also examined. METHODS MRS data were acquired for 32 children with mixed FND (25 girls, 7 boys, aged 10.00 to 16.08 years) and 41 healthy controls of similar age using both short echo point-resolved spectroscopy (PRESS) and Mescher-Garwood point-resolved spectroscopy (MEGAPRESS) sequences in the three regions of interest. RESULTS In the SMA, children with FND had lower NAA/Cr, mI/Cr (trend level), and GABA/Cr ratios. In the aDMN, no group differences in metabolite ratios were found. In the pDMN, children with FND had lower NAA/Cr and mI/Cr (trend level) ratios. While no group differences in E/I balance were found (FND vs. controls), E/I balance in the aDMN was lower in children with functional seizures-a subgroup within the FND group. Pearson correlations found that increased arousal (indexed by higher heart rate) was associated with lower mI/Cr in the SMA and pDMN. CONCLUSIONS Our findings of multiple differences in neurometabolites in children with FND suggest dysfunction on multiple levels of the biological system: the neuron (lower NAA), the glial cell (lower mI), and inhibitory neurotransmission (lower GABA), as well as dysfunction in energy regulation in the subgroup with functional seizures.
Collapse
Affiliation(s)
- Molly Charney
- Department of Neurology, Columbia University Irving Medical Center, New York-Presbyterian, New York, NY, USA; Center for Clinical Spectroscopy, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sheryl Foster
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Radiology, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Vishwa Shukla
- Center for Clinical Spectroscopy, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wufan Zhao
- Center for Clinical Spectroscopy, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sam H Jiang
- Center for Clinical Spectroscopy, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kasia Kozlowska
- Department of Psychological Medicine, The Children's Hospital at Westmead, Westmead, NSW 2145, Australia; Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; Brain Dynamics Centre, Westmead Institute of Medical Research, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia.
| | - Alexander Lin
- Center for Clinical Spectroscopy, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Zhuang H, Li Q, Sun C, Xu D, Gan G, Zhang C, Chen C, Yuan Y, Liu L, Xiao Y, Yao X, Wang C, Kang X, Yang C, Zhao J, Chen W, Wang J, Li J, Luo C, Wang J, Jia X, Yu Z, Liu L. Voluntary wheel exercise ameliorates cognitive impairment, hippocampal neurodegeneration and microglial abnormalities preceded by demyelination in a male mouse model of noise-induced hearing loss. Brain Behav Immun 2023; 114:325-348. [PMID: 37683962 DOI: 10.1016/j.bbi.2023.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/23/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023] Open
Abstract
Acquired peripheral hearing loss (APHL) in midlife has been identified as the greatest modifiable risk factor for dementia; however, the pathophysiological neural mechanisms linking APHL with an increased risk of dementia remain to be elucidated. Here, in an adult male mouse model of noise-induced hearing loss (NIHL), one of the most common forms of APHL, we demonstrated accelerated age-related cognitive decline and hippocampal neurodegeneration during a 6-month follow-up period, accompanied by progressive hippocampal microglial aberrations preceded by immediate-onset transient elevation in serum glucocorticoids and delayed-onset sustained myelin disruption in the hippocampus. Pretreatment with the glucocorticoid receptor antagonist RU486 before stressful noise exposure partially mitigated the early activation of hippocampal microglia, which were present at 7 days post noise exposure (7DPN), but had no impact on later microglial aberrations, hippocampal neurodegeneration, or cognitive decline exhibited at 1 month post noise exposure (1MPN). One month of voluntary wheel exercise following noise exposure barely affected either the hearing threshold shift or hippocampal myelin changes but effectively countered cognitive impairment and the decline in hippocampal neurogenesis in NIHL mice at 1MPN, paralleled by the normalization of microglial morphology, which coincided with a reduction in microglial myelin inclusions and a restoration of microglial hypoxia-inducible factor-1α (HIF1α) expression. Our results indicated that accelerated cognitive deterioration and hippocampal neuroplastic decline following NIHL are most likely driven by the maladaptive response of hippocampal microglia to myelin damage secondary to hearing loss, and we also demonstrated the potential of voluntary physical exercise as a promising and cost-effective strategy to alleviate the detrimental impact of APHL on cognitive function and thus curtail the high and continuously increasing global burden of dementia. Furthermore, the findings of the present study highlight the contribution of myelin debris overload to microglial malfunction and identify the microglial HIF1α-related pathway as an attractive candidate for future comprehensive investigation to obtain a more definitive picture of the underlying mechanisms linking APHL and dementia.
Collapse
Affiliation(s)
- Hong Zhuang
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Qian Li
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Congli Sun
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Dan Xu
- School of Public Health, Southeast University, Nanjing 210009, China
| | - Guangming Gan
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Chenchen Zhang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Chen Chen
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Yang Yuan
- Department of Endocrinology, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China
| | - Linchen Liu
- Department of Rheumatology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yu Xiao
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiuting Yao
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Conghui Wang
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiaoming Kang
- School of Life Science and Technology, Southeast University, Nanjing 210009, China
| | - Chenxi Yang
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jingyi Zhao
- School of Life Science and Technology, Southeast University, Nanjing 210009, China
| | - Wenhao Chen
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jiatang Wang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jinyu Li
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Caichen Luo
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jie Wang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Xirui Jia
- School of Life Science and Technology, Southeast University, Nanjing 210009, China
| | - Zhehao Yu
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Lijie Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
31
|
Zhu H, Guan A, Liu J, Peng L, Zhang Z, Wang S. Noteworthy perspectives on microglia in neuropsychiatric disorders. J Neuroinflammation 2023; 20:223. [PMID: 37794488 PMCID: PMC10548593 DOI: 10.1186/s12974-023-02901-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/22/2023] [Indexed: 10/06/2023] Open
Abstract
Microglia are so versatile that they not only provide immune surveillance for central nervous system, but participate in neural circuitry development, brain blood vessels formation, blood-brain barrier architecture, and intriguingly, the regulation of emotions and behaviors. Microglia have a profound impact on neuronal survival, brain wiring and synaptic plasticity. As professional phagocytic cells in the brain, they remove dead cell debris and neurotoxic agents via an elaborate mechanism. The functional profile of microglia varies considerately depending on age, gender, disease context and other internal or external environmental factors. Numerous studies have demonstrated a pivotal involvement of microglia in neuropsychiatric disorders, including negative affection, social deficit, compulsive behavior, fear memory, pain and other symptoms associated with major depression disorder, anxiety disorder, autism spectrum disorder and schizophrenia. In this review, we summarized the latest discoveries regarding microglial ontogeny, cell subtypes or state spectrum, biological functions and mechanistic underpinnings of emotional and behavioral disorders. Furthermore, we highlight the potential of microglia-targeted therapies of neuropsychiatric disorders, and propose outstanding questions to be addressed in future research of human microglia.
Collapse
Affiliation(s)
- Hongrui Zhu
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Ao Guan
- School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Jiayuan Liu
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Li Peng
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Zhi Zhang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
- Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Sheng Wang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
32
|
Borgonetti V, Galeotti N. Novel Combination of Choline with Withania somnifera (L.) Dunal, and Bacopa monnieri (L.) Wetts Reduced Oxidative Stress in Microglia Cells, Promoting Neuroprotection. Int J Mol Sci 2023; 24:14038. [PMID: 37762339 PMCID: PMC10531461 DOI: 10.3390/ijms241814038] [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: 08/03/2023] [Revised: 08/28/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Memory deficit is one of the major negative outcomes of chronic stress. Cholinergic system modulates memory not only through the neuronal cells, but also via interactions with non-neuronal cells, suggesting that microglia can influence synaptic function and plasticity, contributing to cognition and memory function. Withania somnifera (L.) Dunal (WS) and Bacopa monnieri (L.) Wettst (BM), are traditional herbal medicinal products used for the temporary relief of symptoms of stress. The aim of this study was to investigate whether choline (CLN) activity could be enhanced via an association with adaptogens: WS and BM extracts. First, we optimized an in vitro model of corticotropin-releasing hormone (CRH)-induced oxidative stress on microglial BV2 cells. CRH 100 nM reduced BV2 cell viability and induced morphological changes and neurotoxicity after 24 h of microglia stimulation. Moreover, it induced an increase in the production of reactive oxygen species (ROS) and dysregulated antioxidant protein (i.e., SIRT-1 and NRF-2). The association between choline and adaptogens (CBW) 10 μg/mL counteracted the effect of CRH on BV2 cells and reduced the neurotoxicity produced by BV2 CRH-conditioned medium in the SH-SY5Y cell lines. CBW 200 mg/kg produced an ameliorative effect on recognition memory in the novel object recognition test (NORT) test in mice. In conclusion, combining choline with adaptogen plant extracts might represent a promising intervention in chronic stress associated with memory disturbances through the attenuation of microglia-induced oxidative stress.
Collapse
Affiliation(s)
| | - Nicoletta Galeotti
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy;
| |
Collapse
|
33
|
Ma L, Yan Y, Webb RJ, Li Y, Mehrabani S, Xin B, Sun X, Wang Y, Mazidi M. Psychological Stress and Gut Microbiota Composition: A Systematic Review of Human Studies. Neuropsychobiology 2023; 82:247-262. [PMID: 37673059 DOI: 10.1159/000533131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 07/10/2023] [Indexed: 09/08/2023]
Abstract
INTRODUCTION The associations between psychological stress and gut microbiota composition are not fully understood. This study investigated associations between psychological stress and gut microbiota composition and examined the potential modifying effects of age, sex, and ethnicity on such associations. METHODS A systematic literature search was conducted using PubMed, Web of Science, PsycINFO, and Embase databases for studies published until November 2021 which examined associations between psychological stress and gut microbiota composition. RESULTS During the search process, 10,790 studies were identified, and after screening, 13 met the eligibility criteria and were included. The median sample size was 70, and the median age of participants was 28.0 years. Most of the included studies did not report associations between measures of alpha- and beta diversity of the gut microbiota composition and psychological stress. A few studies reported that the Shannon index, Chao 1, Simpson index, and weighted UniFrac were negatively associated with psychological stress. Significant reductions in several taxa at the phyla-, family-, and genus-levels were observed in participants with higher psychological stress. At the phylum level, the abundance of Proteobacteria and Verrucomicrobia were negatively associated with psychological stress. At the family-level, no more than two studies reported associations of the same microbiota with psychological stress. At the genus level, the following results were found in more than two studies; psychological stress was negatively associated with the abundance of Lachnospira, Lachnospiraceae, Phascolarctobacterium, Sutterella, and Veillonella, and positively associated with the abundance of Methanobrevibacter, Rhodococcus, and Roseburia. However, it was not possible to determine the influence of age, sex, or ethnicity due to the limited studies included. CONCLUSION Our findings provide evidence that psychological stress is associated with changes in the abundance of the gut microbiota. Larger sample longitudinal studies are needed to determine the causal relationship between psychological stress and the gut microbiota.
Collapse
Affiliation(s)
- Lu Ma
- Global Health Institute, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yating Yan
- Global Health Institute, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China,
| | - Richard James Webb
- School of Health and Sports Sciences, Hope Park Campus, Liverpool Hope University, Liverpool, UK
| | - Ying Li
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Sanaz Mehrabani
- Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bao Xin
- School of Public Health, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xiaomin Sun
- Global Health Institute, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Youfa Wang
- Global Health Institute, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Mohsen Mazidi
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK
- Nuffield Department of Population Health, Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), University of Oxford, Oxford, UK
- Department of Twin Research and Genetic Epidemiology, King's College London, South Wing St Thomas', London, UK
| |
Collapse
|
34
|
Chen X, Cui QQ, Hu XH, Ye J, Liu ZC, Mei YX, Wang F, Hu ZL, Chen JG. CD200 in dentate gyrus improves depressive-like behaviors of mice through enhancing hippocampal neurogenesis via alleviation of microglia hyperactivation. J Neuroinflammation 2023; 20:157. [PMID: 37391731 DOI: 10.1186/s12974-023-02836-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/16/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND Neuroinflammation and microglia play critical roles in the development of depression. Cluster of differentiation 200 (CD200) is an anti-inflammatory glycoprotein that is mainly expressed in neurons, and its receptor CD200R1 is primarily in microglia. Although the CD200-CD200R1 pathway is necessary for microglial activation, its role in the pathophysiology of depression remains unknown. METHODS The chronic social defeat stress (CSDS) with behavioral tests were performed to investigate the effect of CD200 on the depressive-like behaviors. Viral vectors were used to overexpress or knockdown of CD200. The levels of CD200 and inflammatory cytokines were tested with molecular biological techniques. The status of microglia, the expression of BDNF and neurogenesis were detected with immunofluorescence imaging. RESULTS We found that the expression of CD200 was decreased in the dentate gyrus (DG) region of mice experienced CSDS. Overexpression of CD200 alleviated the depressive-like behaviors of stressed mice and inhibition of CD200 facilitated the susceptibility to stress. When CD200R1 receptors on microglia were knocked down, CD200 was unable to exert its role in alleviating depressive-like behavior. Microglia in the DG brain region were morphologically activated after exposure to CSDS. In contrast, exogenous administration of CD200 inhibited microglia hyperactivation, alleviated neuroinflammatory response in hippocampus, and increased the expression of BDNF, which in turn ameliorated adult hippocampal neurogenesis impairment in the DG induced by CSDS. CONCLUSIONS Taken together, these results suggest that CD200-mediated alleviation of microglia hyperactivation contributes to the antidepressant effect of neurogenesis in dentate gyrus in mice.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian-Qian Cui
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Hai Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Ye
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zi-Cun Liu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan-Xi Mei
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
- The Research Center for Depression, Tongji Medical College, Huazhong University of Science, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China
| | - Zhuang-Li Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China.
- The Research Center for Depression, Tongji Medical College, Huazhong University of Science, Wuhan, 430030, China.
- Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China.
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China.
- The Research Center for Depression, Tongji Medical College, Huazhong University of Science, Wuhan, 430030, China.
- Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China.
| |
Collapse
|
35
|
Infantes-López MI, Nieto-Quero A, Chaves-Peña P, Zambrana-Infantes E, Cifuentes M, Márquez J, Pedraza C, Pérez-Martín M. New insights into hypothalamic neurogenesis disruption after acute and intense stress: implications for microglia and inflammation. Front Neurosci 2023; 17:1190418. [PMID: 37425000 PMCID: PMC10327603 DOI: 10.3389/fnins.2023.1190418] [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: 03/20/2023] [Accepted: 05/23/2023] [Indexed: 07/11/2023] Open
Abstract
In recent years, the hypothalamus has emerged as a new neurogenic area, capable of generating new neurons after development. Neurogenesis-dependent neuroplasticity seems to be critical to continuously adapt to internal and environmental changes. Stress is a potent environmental factor that can produce potent and enduring effects on brain structure and function. Acute and chronic stress is known to cause alterations in neurogenesis and microglia in classical adult neurogenic regions such as the hippocampus. The hypothalamus is one of the major brain regions implicated in homeostatic stress and emotional stress systems, but little is known about the effect of stress on the hypothalamus. Here, we studied the impact of acute and intense stress (water immersion and restrain stress, WIRS), which may be considered as an inducer of an animal model of posttraumatic stress disorder, on neurogenesis and neuroinflammation in the hypothalamus of adult male mice, focusing on three nuclei: PVN, VMN and ARC, and also in the periventricular area. Our data revealed that a unique stressor was sufficient to provoke a significant impact on hypothalamic neurogenesis by inducing a reduction in the proliferation and number of immature neurons identified as DCX+ cells. These differences were accompanied by marked microglial activation in the VMN and ARC, together with a concomitant increase in IL-6 levels, indicating that WIRS induced an inflammatory response. To investigate the possible molecular mechanisms responsible for neuroplastic and inflammatory changes, we tried to identify proteomic changes. The data revealed that WIRS induced changes in the hypothalamic proteome, modifying the abundance of three and four proteins after 1 h or 24 h of stress application, respectively. These changes were also accompanied by slight changes in the weight and food intake of the animals. These results are the first to show that even a short-term environmental stimulus such as acute and intense stress can have neuroplastic, inflammatory, functional and metabolic consequences on the adult hypothalamus.
Collapse
Affiliation(s)
- María Inmaculada Infantes-López
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma Bionand, Málaga, Spain
| | - Andrea Nieto-Quero
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma Bionand, Málaga, Spain
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Málaga, Spain
| | - Patricia Chaves-Peña
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Málaga, Spain
| | - Emma Zambrana-Infantes
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma Bionand, Málaga, Spain
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Málaga, Spain
| | - Manuel Cifuentes
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma Bionand, Málaga, Spain
| | - Javier Márquez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma Bionand, Málaga, Spain
- Departamento de Biología Molecular y Bioquímica, Canceromics Lab, Universidad de Málaga, Málaga, Spain
| | - Carmen Pedraza
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma Bionand, Málaga, Spain
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Málaga, Spain
| | - Margarita Pérez-Martín
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma Bionand, Málaga, Spain
| |
Collapse
|
36
|
Coleman PT, Costanza-Chavez GW, Martin HN, Amat J, Frank MG, Sanchez RJ, Potter GJ, Mellert SM, Carter RK, Bonnici GN, Maier SF, Baratta MV. Prior experience with behavioral control over stress facilitates social dominance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543982. [PMID: 37333397 PMCID: PMC10274770 DOI: 10.1101/2023.06.06.543982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Dominance status has extensive effects on physical and mental health, and an individual's relative position can be shaped by experiential factors. A variety of considerations suggest that the experience of behavioral control over stressors should produce winning in dominance tests and that winning should blunt the impact of later stressors, as does prior control. To investigate the interplay between competitive success and stressor control, we first examined the impact of stressor controllability on subsequent performance in a warm spot competition test modified for rats. Prior experience of controllable, but not physically identical uncontrollable, stress increased later effortful behavior and occupation of the warm spot. Controllable stress subjects consistently ranked higher than did uncontrollable stress subjects. Pharmacological inactivation of the prelimbic (PL) cortex during behavioral control prevented later facilitation of dominance. Next, we explored whether repeated winning experiences produced later resistance against the typical sequelae of uncontrollable stress. To establish dominance status, triads of rats were given five sessions of warm spot competition. Reversible inactivation of the PL or NMDA receptor blockade in the dorsomedial striatum led to a long-term reduction in social rank. Stable dominance blunted the later stress-induced increase in dorsal raphe nucleus serotonergic activity, as well as prevented stress-induced social avoidance. In contrast, endocrine and neuroimmune responses to uncontrollable stress were unaffected, indicating a selective impact of prior dominance. Together, these data demonstrate that instrumental control over stress promotes later dominance, but also reveal that winning experiences buffer against the neural and behavioral outcomes of future adversity.
Collapse
|
37
|
Li Q, Kang X, Liu L, Xiao Y, Xu D, Zhuang H, Liu H, Zhao J, Zou H, Yang J, Zhan X, Li T, Wang X, Liu L. Adult mice with noise-induced hearing loss exhibited temporal ordering memory deficits accompanied by microglia-associated neuroplastic changes in the medial prefrontal cortex. Neurobiol Dis 2023:106181. [PMID: 37271287 DOI: 10.1016/j.nbd.2023.106181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 05/17/2023] [Accepted: 05/30/2023] [Indexed: 06/06/2023] Open
Abstract
Acquired peripheral hearing loss in midlife is considered the primary modifiable risk factor for dementia, while the underlying pathological mechanism remains poorly understood. Excessive noise exposure is the most common cause of acquired peripheral hearing loss in modern society. This study was designed to investigate the impact of noise-induced hearing loss (NIHL) on cognition, with a focus on the medial prefrontal cortex (mPFC), a brain region that is involved in both auditory and cognitive processes and is highly affected in patients with cognitive impairment. Adult C57BL/6 J mice were randomly assigned to a control group and seven noise groups: 0HPN, 12HPN, 1DPN, 3DPN, 7DPN, 14DPN, and 28DPN, which were exposed to broadband noise at a 123 dB sound pressure level (SPL) for 2 h and sacrificed immediately (0 h), 12 h, or 1, 3, 7, 14, or 28 days post-noise exposure (HPN, DPN), respectively. Hearing assessment, behavioral tests, and neuromorphological studies in the mPFC were performed in control and 28DPN mice. All experimental animals were included in the time-course analysis of serum corticosterone (CORT) levels and mPFC microglial morphology. The results illustrated that noise exposure induced early-onset transient serum CORT elevation and permanent moderate-to-severe hearing loss in mice. 28DPN mice, in which permanent NIHL has been verified, exhibited impaired performance in temporal order object recognition tasks concomitant with reduced structural complexity of mPFC pyramidal neurons. The time-course immunohistochemical analysis in the mPFC revealed significantly higher morphological microglial activation at 14 and 28 DPN, preceded by a remarkably higher amount of microglial engulfed postsynaptic marker PSD95 at 7 DPN. Additionally, lipid accumulation in microglia was observed in 7DPN, 14DPN and 28DPN mice, suggesting a driving role of lipid handling deficits following excessive phagocytosis of synaptic elements in delayed and sustained microglial abnormalities. These findings provide fundamentally novel information concerning mPFC-related cognitive impairment in mice with NIHL and empirical evidence suggesting the involvement of microglial malfunction in the mPFC neurodegenerative consequences of NIHL.
Collapse
Affiliation(s)
- Qian Li
- Medical College, Southeast University, Nanjing 210009, China
| | - Xiaomin Kang
- School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Linchen Liu
- Department of Rheumatology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Yu Xiao
- Medical College, Southeast University, Nanjing 210009, China
| | - Dan Xu
- School of Public Health, Southeast University, Nanjing 210009, China
| | - Hong Zhuang
- Medical College, Southeast University, Nanjing 210009, China
| | - Haiqing Liu
- School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Jingyi Zhao
- School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Han Zou
- Medical College, Southeast University, Nanjing 210009, China
| | - Jianing Yang
- Medical College, Southeast University, Nanjing 210009, China
| | - Xindi Zhan
- Medical College, Southeast University, Nanjing 210009, China
| | - Tianxiao Li
- Medical College, Southeast University, Nanjing 210009, China
| | - Xinchen Wang
- Medical College, Southeast University, Nanjing 210009, China
| | - Lijie Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
38
|
Seymour B, Crook RJ, Chen ZS. Post-injury pain and behaviour: a control theory perspective. Nat Rev Neurosci 2023; 24:378-392. [PMID: 37165018 PMCID: PMC10465160 DOI: 10.1038/s41583-023-00699-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2023] [Indexed: 05/12/2023]
Abstract
Injuries of various types occur commonly in the lives of humans and other animals and lead to a pattern of persistent pain and recuperative behaviour that allows safe and effective recovery. In this Perspective, we propose a control-theoretic framework to explain the adaptive processes in the brain that drive physiological post-injury behaviour. We set out an evolutionary and ethological view on how animals respond to injury, illustrating how the behavioural state associated with persistent pain and recuperation may be just as important as phasic pain in ensuring survival. Adopting a normative approach, we suggest that the brain implements a continuous optimal inference of the current state of injury from diverse sensory and physiological signals. This drives the various effector control mechanisms of behavioural homeostasis, which span the modulation of ongoing motivation and perception to drive rest and hyper-protective behaviours. However, an inherent problem with this is that these protective behaviours may partially obscure information about whether injury has resolved. Such information restriction may seed a tendency to aberrantly or persistently infer injury, and may thus promote the transition to pathological chronic pain states.
Collapse
Affiliation(s)
- Ben Seymour
- Institute for Biomedical Engineering, University of Oxford, Oxford, UK.
- Wellcome Centre for Integrative Neuroimaging, John Radcliffe Hospital, Headington, Oxford, UK.
| | - Robyn J Crook
- Department of Biology, San Francisco State University, San Francisco, CA, USA.
| | - Zhe Sage Chen
- Departments of Psychiatry, Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY, USA.
- Interdisciplinary Pain Research Program, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
39
|
Adkins AM, Colby EM, Kim WK, Wellman LL, Sanford LD. Stressor control and regional inflammatory responses in the brain: regulation by the basolateral amygdala. J Neuroinflammation 2023; 20:128. [PMID: 37244986 PMCID: PMC10225081 DOI: 10.1186/s12974-023-02813-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/20/2023] [Indexed: 05/29/2023] Open
Abstract
Increasing evidence has connected the development of certain neuropsychiatric disorders, as well as neurodegenerative diseases, to stress-induced dysregulation of the immune system. We have shown that escapable (ES) and inescapable (IS) footshock stress, and memories associated with ES or IS, can differentially alter inflammatory-related gene expression in brain in a region dependent manner. We have also demonstrated that the basolateral amygdala (BLA) regulates stress- and fear memory-induced alterations in sleep, and that differential sleep and immune responses in the brain to ES and IS appear to be integrated during fear conditioning and then reproduced by fear memory recall. In this study, we investigated the role of BLA in influencing regional inflammatory responses within the hippocampus (HPC) and medial prefrontal cortex (mPFC) by optogenetically stimulating or inhibiting BLA in male C57BL/6 mice during footshock stress in our yoked shuttlebox paradigm based on ES and IS. Then, mice were immediately euthanized and RNA extracted from brain regions of interest and loaded into NanoString® Mouse Neuroinflammation Panels for compilation of gene expression profiles. Results showed differential regional effects in gene expression and activated pathways involved in inflammatory-related signaling following ES and IS, and these differences were altered depending on amygdalar excitation or inhibition. These findings demonstrate that the stress-induced immune response, or "parainflammation", is affected by stressor controllability and that BLA influences regional parainflammation to ES or IS in HPC and mPFC. The study illustrates how stress-induced parainflammation can be regulated at the neurocircuit level and suggests that this approach can be useful for uncovering circuit and immune interactions in mediating differential stress outcomes.
Collapse
Affiliation(s)
- Austin M. Adkins
- Sleep Research Laboratory, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA 23507 USA
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA 23507 USA
- Pathology and Anatomy, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA 23507 USA
| | - Emily M. Colby
- Sleep Research Laboratory, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA 23507 USA
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA 23507 USA
- Pathology and Anatomy, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA 23507 USA
| | - Woong-Ki Kim
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA 23507 USA
- Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, P.O. Box 1980, VA 23507 Norfolk, USA
| | - Laurie L. Wellman
- Sleep Research Laboratory, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA 23507 USA
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA 23507 USA
- Pathology and Anatomy, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA 23507 USA
| | - Larry D. Sanford
- Sleep Research Laboratory, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA 23507 USA
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA 23507 USA
- Pathology and Anatomy, Eastern Virginia Medical School, P.O. Box 1980, Norfolk, VA 23507 USA
| |
Collapse
|
40
|
Fricchione G. Mind body medicine: a modern bio-psycho-social model forty-five years after Engel. Biopsychosoc Med 2023; 17:12. [PMID: 36997979 PMCID: PMC10060142 DOI: 10.1186/s13030-023-00268-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 03/31/2023] Open
Affiliation(s)
- Gregory Fricchione
- Benson-Henry Institute for Mind Body Medicine at Massachusetts General Hospital, Boston, USA.
- Department of Psychiatry, Massachusetts General Hospital, Boston, USA.
- Harvard Medical School, Boston, USA.
| |
Collapse
|
41
|
Wang C, Zhou Y, Feinstein A. Neuro-immune crosstalk in depressive symptoms of multiple sclerosis. Neurobiol Dis 2023; 177:106005. [PMID: 36680805 DOI: 10.1016/j.nbd.2023.106005] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Depressive disorders can occur in up to 50% of people with multiple sclerosis in their lifetime. If left untreated, comorbid major depressive disorders may not spontaneously remit and is associated with an increased morbidity and mortality. Conversely, epidemiological evidence supports increased psychiatric visit as a significant prodromal event prior to diagnosis of MS. Are there common molecular pathways that contribute to the co-development of MS and psychiatric illnesses? We discuss immune cells that are dysregulated in MS and how such dysregulation can induce or protect against depressive symptoms. This is not meant to be a comprehensive review of all molecular pathways but rather a framework to guide future investigations of immune responses in depressed versus euthymic people with MS. Currently, there is weak evidence supporting the use of antidepressant medication in comorbid MS patients. It is our hope that by better understanding the neuroimmune crosstalk in the context of depression in MS, we can enhance the potential for future therapeutic options.
Collapse
Affiliation(s)
- Chao Wang
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Yulin Zhou
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Anthony Feinstein
- Department of Psychiatry, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
42
|
Harvey AR. Injury, illness, and emotion: A review of the motivational continuum from trauma through recovery from an ecological perspective. Brain Behav Immun Health 2023; 27:100586. [PMID: 36655055 PMCID: PMC9841046 DOI: 10.1016/j.bbih.2022.100586] [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: 10/30/2022] [Revised: 12/15/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Image 1.
Collapse
|
43
|
Shin SH, Kim YK. Early Life Stress, Neuroinflammation, and Psychiatric Illness of Adulthood. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:105-134. [PMID: 36949308 DOI: 10.1007/978-981-19-7376-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Stress exposure during early stages of life elevates the risk of developing psychopathologies and psychiatric illness in later life. The brain and immune system are not completely developed by birth and therefore continue develop after birth; this post birth development is influenced by several psychosocial factors; hence, early life stress (ELS) exposure can alter brain structural development and function. A growing number of experimental animal and observational human studies have investigated the link between ELS exposure and increased risk of psychopathology through alternations in the immune system, by evaluating inflammation biomarkers. Recent studies, including brain imaging, have also shed light on the mechanisms by which both the innate and adaptive immune systems interact with neural circuits and neurotransmitters, which affect psychopathology. Herein, we discuss the link between the experience of stress in early life and lifelong alterations in the immune system, which subsequently lead to the development of various psychiatric illnesses.
Collapse
Affiliation(s)
- Sang Ho Shin
- Department of Psychiatry, College of Medicine, Korea University Ansan Hospital, Korea University, Ansan, Republic of Korea
| | - Yong-Ku Kim
- Department of Psychiatry, College of Medicine, Korea University Ansan Hospital, Korea University, Ansan, Republic of Korea.
| |
Collapse
|
44
|
Machado da Silva MC, Iglesias LP, Candelario-Jalil E, Khoshbouei H, Moreira FA, de Oliveira ACP. Role of Microglia in Psychostimulant Addiction. Curr Neuropharmacol 2023; 21:235-259. [PMID: 36503452 PMCID: PMC10190137 DOI: 10.2174/1570159x21666221208142151] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022] Open
Abstract
The use of psychostimulant drugs can modify brain function by inducing changes in the reward system, mainly due to alterations in dopaminergic and glutamatergic transmissions in the mesocorticolimbic pathway. However, the etiopathogenesis of addiction is a much more complex process. Previous data have suggested that microglia and other immune cells are involved in events associated with neuroplasticity and memory, which are phenomena that also occur in addiction. Nevertheless, how dependent is the development of addiction on the activity of these cells? Although the mechanisms are not known, some pathways may be involved. Recent data have shown psychoactive substances may act directly on immune cells, alter their functions and induce various inflammatory mediators that modulate synaptic activity. These could, in turn, be involved in the pathological alterations that occur in substance use disorder. Here, we extensively review the studies demonstrating how cocaine and amphetamines modulate microglial number, morphology, and function. We also describe the effect of these substances in the production of inflammatory mediators and a possible involvement of some molecular signaling pathways, such as the toll-like receptor 4. Although the literature in this field is scarce, this review compiles the knowledge on the neuroimmune axis that is involved in the pathogenesis of addiction, and suggests some pharmacological targets for the development of pharmacotherapy.
Collapse
Affiliation(s)
- Maria Carolina Machado da Silva
- Department of Pharmacology, Neuropharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil;
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lia Parada Iglesias
- Department of Pharmacology, Neuropsychopharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Habibeh Khoshbouei
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Fabrício Araujo Moreira
- Department of Pharmacology, Neuropsychopharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | |
Collapse
|
45
|
Olfactory Stimulation Successfully Modulates the Neurochemical, Biochemical and Behavioral Phenotypes of the Visceral Pain. Molecules 2022; 27:molecules27217659. [DOI: 10.3390/molecules27217659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Visceral pain (VP) is the organ-derived nociception in which increased inflammatory reaction and exaggerated activation of the central nucleus of the amygdala (CeA) may contribute to this deficiency. Considering the amygdala also serves as the integration center for olfaction, the present study aimed to determine whether olfactory stimulation (OS) would effectively depress over-activation and inflammatory reaction in CeA, and successfully relieve VP-induced abnormalities. Adult rats subjected to intraperitoneal injection of acetic acid inhaled lavender essential oil for 2 or 4 h. The potential benefits of OS were determined by measuring the pro-inflammatory cytokine level, intracellular potassium and the upstream small-conductance calcium-activated potassium (SK) channel expression, together with detecting the stress transmitters that participated in the modulation of CeA activity. Results indicated that in VP rats, strong potassium intensity, reduced SK channel protein level, and increased corticotropin-releasing factor, c-fos, and substance P immuno-reactivities were detected in CeA. Enhanced CeA activation corresponded well with increased inflammatory reaction and decreased locomotion, respectively. However, in rats subjected to VP and received OS, all above parameters were significantly returned to normal levels with higher change detected in treating OS of 4h. As OS successfully depresses inflammation and CeA over-activation, application of OS may serve as an alternative and effective strategy to efficiently relieve VP-induced deficiency.
Collapse
|
46
|
Schramm E, Waisman A. Microglia as Central Protagonists in the Chronic Stress Response. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:9/6/e200023. [PMID: 36357946 PMCID: PMC9453699 DOI: 10.1212/nxi.0000000000200023] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 07/06/2022] [Indexed: 05/15/2023]
Abstract
Chronic stress is a major risk factor for developing psychiatric conditions. In addition to elevating the levels of stress hormones released in the body, chronic stress activates the immune system, resulting in increased levels of proinflammatory cytokines and innate immune cells in the circulation of rodents and humans. Furthermore, exposure to chronic stress alters the phenotype of microglia, a population of innate immune cells that reside in the CNS parenchyma. In rodent models, chronic stress activates microglia in defined brain regions and induces changes in their phenotype and functional properties. In this review, we discussed how microglia are activated in stressful situations. Furthermore, we described how microglia affect the CNS environment during chronic stress, through the production of cytokines, the induction of reactive oxygen species, and phagocytosis. We suggested that, due to their strategic location as immune cells within the CNS, microglia are important players in the induction of psychopathologies after chronic stress.
Collapse
Affiliation(s)
- Eva Schramm
- From the Institute for Molecular Medicine (E.S., A.W.), University Medical Center, Johannes Gutenberg University Mainz; Focus Program Translational Neurosciences (A.W.), University Medical Center of the Johannes Gutenberg-University Mainz; and Research Center for Immunotherapy (A.W.), University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Ari Waisman
- From the Institute for Molecular Medicine (E.S., A.W.), University Medical Center, Johannes Gutenberg University Mainz; Focus Program Translational Neurosciences (A.W.), University Medical Center of the Johannes Gutenberg-University Mainz; and Research Center for Immunotherapy (A.W.), University Medical Center of the Johannes Gutenberg-University Mainz, Germany.
| |
Collapse
|
47
|
Zhang Y, Dong Y, Zhu Y, Sun D, Wang S, Weng J, Zhu Y, Peng W, Yu B, Jiang Y. Microglia-specific transcriptional repression of interferon-regulated genes after prolonged stress in mice. Neurobiol Stress 2022; 21:100495. [DOI: 10.1016/j.ynstr.2022.100495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/25/2022] Open
|
48
|
El-Azma MH, El-Beih NM, El-Shamy KA, Koriem KM, Elkassaby MI, El-Sayed WM. Pumpkin seed oil and zinc attenuate chronic mild stress perturbations in the cerebral cortex of rats. NUTRITION & FOOD SCIENCE 2022; 52:1070-1082. [DOI: 10.1108/nfs-10-2021-0315] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Purpose
This study aims to investigate the potential of pumpkin seed oil (PSO) and zinc to attenuate oxidative stress and neuroinflammation caused by chronic mild stress (CMS) in the cerebral cortex of male rats.
Design/methodology/approach
The rats were submitted to stress for six weeks and then the behavior of the rats was tested by forced swimming test (FST) and novel cage test. The treated groups were given venlafaxine (20 mg/kg), pumpkin seed oil (40 mg/kg) and zinc (4 mg/kg). The cortex homogenate was used for the detection of the oxidative stress parameters, the concentration of neurotransmitters, tumor necrosis factor-α (TNF-α) and interleukin 1β (IL-1β), Na+/K+-ATPase activity, and the expression of histamine N-methyltransferase (Hnmt) and tyrosine hydroxylase (Th).
Findings
CMS causes a significant increase in immobility time in the FST and a significant decrease in the number of rearing in the novel cage test. CMS group showed a significant increase in alanine aminotransferase (ALT) activity, levels of cortisol, TNF-α, IL-1β, nitric oxide and malondialdehyde. CMS caused a significant decrease in the concentrations of serotonin, GABA, norepinephrine, and the activities of glutathione peroxidase, catalase, superoxide dismutase and Na+/K+-ATPase. CMS caused a marked reduction in the expression of Hnmt and Th in the cortex. PSO and zinc attenuated the Na+/K+-ATPase activity, oxidative parameters and neuroinflammation induced by the CMS, and this was reflected by the elevation of the concentration of neurotransmitters and reduction of cortisol and ALT, in addition to the behavior normalization. PSO and zinc attenuated the CMS by improving the antioxidant milieu and anti-inflammatory status of the cerebral cortex.
Originality/value
There are no studies on the effect of pumpkin seed oil on depression
Collapse
|
49
|
Wu H, Bao H, Liu C, Zhang Q, Huang A, Quan M, Li C, Xiong Y, Chen G, Hou L. Extracellular Nucleosomes Accelerate Microglial Inflammation via C-Type Lectin Receptor 2D and Toll-Like Receptor 9 in mPFC of Mice With Chronic Stress. Front Immunol 2022; 13:854202. [PMID: 35844599 PMCID: PMC9276970 DOI: 10.3389/fimmu.2022.854202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 05/27/2022] [Indexed: 11/23/2022] Open
Abstract
Damage-associated molecular patterns (DAMPs) are the primary promoter of progressive neuroinflammation and are associated with chronic stress-related emotional disorders. The present study investigated the role and mechanism of extracellular nucleosomes and histones, the newly defined DAMPs, in mice with chronic stress. C57BL/6 mice were exposed to chronic unpredictable mild stress (CUMS) and corticosterone drinking, respectively, for 4 weeks. Negative emotional behaviors were comprehensively investigated. Microglial morphology, oxidative stress, and inflammation, as well as C-type lectin receptor 2D (Clec2d) and Toll-like receptor 9 (TLR9) expression in medial prefrontal cortex (mPFC) were assessed with flow cytometer and cell sorting. Specifically, microglial pro-inflammatory activation and inflammation were further investigated with stereotactic injection of recombinant nucleosomes and histones in mPFC and further evaluated with AAV-Clec2d knocking-down, DNase I, and activated protein C (APC) pretreatment. Moreover, the rescue effect by AAV-Clec2d knocking-down was observed in mice with chronic stress. Mice with chronic stress were presented as obviously depressive- and anxiety-like behaviors and accompanied with significant microglial oxidative stress and inflammation, indicating by reactive oxygen species (ROS) production, primed nuclear factor-κB (NF-κB) signaling pathway, activated NACHT, LRR, and PYD domain–containing protein 3 (NLRP3) inflammasome, and upregulated Clec2d and TLR9 in mPFC, together with histones dictation in cerebrospinal fluid and extracellular trap formation. Stereotactic injection of nucleosomes was contributed to promote microglial inflammation rather than histones in mPFC, indicating that the pro-inflammatory role was derived from extracellular histones-bound DNA but not freely histones. AAV-Clec2d knocking-down, DNase I, and APC were all effective to inhibit nucleosome-induced microglial oxidative stress and inflammation. Moreover, AAV-Clec2d knocking-down in mice with chronic stress exhibited reduced microglial inflammation and improved negative emotional behaviors. Our findings reveal a novel mechanism of DAMP-associated inflammation that extracellular nucleosomes accelerate microglial inflammation via Clec2d and TLR9, and then contribute to chronic stress-induced emotional disorders.
Collapse
Affiliation(s)
- Huanghui Wu
- Department of Anesthesiology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Han Bao
- Department of Anesthesiology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Cong Liu
- Department of Anesthesiology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Qiao Zhang
- Department of Anesthesiology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Ailing Huang
- Department of Anesthesiology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Minxue Quan
- Department of Anesthesiology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Chunhui Li
- Department of Anesthesiology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Ying Xiong
- Department of Anesthesiology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Guozhong Chen
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University, Shanghai, China
- *Correspondence: Guozhong Chen, ; Lichao Hou,
| | - Lichao Hou
- Department of Anesthesiology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Guozhong Chen, ; Lichao Hou,
| |
Collapse
|
50
|
Tidmarsh LV, Harrison R, Ravindran D, Matthews SL, Finlay KA. The Influence of Adverse Childhood Experiences in Pain Management: Mechanisms, Processes, and Trauma-Informed Care. FRONTIERS IN PAIN RESEARCH 2022; 3:923866. [PMID: 35756908 PMCID: PMC9226323 DOI: 10.3389/fpain.2022.923866] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/20/2022] [Indexed: 12/30/2022] Open
Abstract
Adverse childhood experiences (ACEs) increase the likelihood of reduced physical and psychological health in adulthood. Though understanding and psychological management of traumatic experiences is growing, the empirical exploration of ACEs and physical clinical outcomes remains under-represented and under-explored. This topical review aimed to highlight the role of ACEs in the experience of chronic pain, pain management services and clinical decision making by: (1) providing an overview of the relationship between ACEs and chronic pain; (2) identifying biopsychosocial mechanisms through which ACEs may increase risk of persistent pain; (3) highlighting the impact of ACEs on patient adherence and completion of pain management treatment; and (4) providing practical clinical implications for pain management. Review findings demonstrated that in chronic pain, ACEs are associated with increased pain complications, pain catastrophizing and depression and the combination of these factors further heightens the risk of early treatment attrition. The pervasive detrimental impacts of the COVID-19 pandemic on ACEs and their cyclical effects on pain are discussed in the context of psychological decline during long treatment waitlists. The review highlights how people with pain can be further supported in pain services by maintaining trauma-informed practices and acknowledging the impact of ACEs on chronic pain and detrimental health outcomes. Clinicians who are ACE-informed have the potential to minimize the negative influence of ACEs on treatment outcomes, ultimately optimizing the impact of pain management services.
Collapse
Affiliation(s)
- Lydia V. Tidmarsh
- School of Psychology and Clinical Language Sciences, University of Reading, Reading, United Kingdom
- *Correspondence: Lydia V. Tidmarsh
| | - Richard Harrison
- School of Psychology and Clinical Language Sciences, University of Reading, Reading, United Kingdom
| | | | - Samantha L. Matthews
- School of Psychology and Clinical Language Sciences, University of Reading, Reading, United Kingdom
| | - Katherine A. Finlay
- School of Psychology and Clinical Language Sciences, University of Reading, Reading, United Kingdom
| |
Collapse
|