1
|
Rawls A, Dziabis J, Nguyen D, Anbarci D, Clark M, Zhang G, Dzirasa K, Bilbo SD. Sex-specific regulation of microglial MyD88 in HMGB1-Induced anxiety phenotype in mice. Neurobiol Stress 2025; 36:100721. [PMID: 40236260 PMCID: PMC11997396 DOI: 10.1016/j.ynstr.2025.100721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 03/19/2025] [Accepted: 03/22/2025] [Indexed: 04/17/2025] Open
Abstract
Stress is a significant risk factor for the development and recurrence of anxiety disorders. Stress can profoundly impact the immune system, and lead to microglial functional alterations in the medial prefrontal cortex (mPFC), a brain region involved in the pathogenesis of anxiety. High mobility group box 1 protein (HMGB1) is a potent pro-inflammatory stimulus and danger-associated molecular pattern (DAMP) released from neuronal and non-neuronal cells following stress. HMGB1 provokes pro-inflammatory responses in the brain and, when administered locally, alters behavior in the absence of other stressors. In this study, we administered dsHMGB1 into the mPFC of male and female mice for 5 days to investigate the cellular and molecular mechanisms underlying HMGB1-induced behavioral dysfunction, with a focus on cell-type specificity and potential sex differences. Here, we demonstrate that dsHMGB1 infusion into the mPFC elicited behavior changes in both sexes but only altered microglial morphology robustly in female mice. Moreover, preventing microglial changes with cell-specific ablation of the MyD88 pathway prevented anxiety-like behaviors only in females. These results support the hypothesis that microglial MyD88 signaling is a critical mediator of HMGB1-induced stress responses, particularly in adult female mice.
Collapse
Affiliation(s)
- Ashleigh Rawls
- Department of Pharmacology, Duke University, Durham, NC, United States of America
| | - Julia Dziabis
- Department of Psychology and Neuroscience, Duke University, Durham, NC, United States of America
| | - Dang Nguyen
- Department of Psychology and Neuroscience, Duke University, Durham, NC, United States of America
| | - Dilara Anbarci
- Department of Cell Biology, Duke University, Durham, NC, United States of America
| | - Madeline Clark
- Department of Neurobiology, Duke University, Durham, NC, United States of America
| | - Grace Zhang
- Department of Psychology and Neuroscience, Duke University, Durham, NC, United States of America
| | - Kafui Dzirasa
- Department of Neurobiology, Duke University, Durham, NC, United States of America
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, United States of America
- Howard Hughes Medical Institute, Chevy Chase, MD, United States of America
| | - Staci D. Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, United States of America
- Department of Neurobiology, Duke University, Durham, NC, United States of America
| |
Collapse
|
2
|
Wang H, Lu X, Ye Y, Huang C, Fang Y, Yang R, Sun M, Ren J, Song R, Xu F, Su J, Hong H, Huang C. Stimulation of microglia leads to a rapid antidepressant effect by triggering astrocytic P2Y1Rs and promoting BDNF-mediated neurogenesis in the hippocampus. Brain Behav Immun 2025; 128:134-151. [PMID: 40194747 DOI: 10.1016/j.bbi.2025.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/29/2025] [Accepted: 04/03/2025] [Indexed: 04/09/2025] Open
Abstract
Reversing the decline of microglia in the dentate gyrus of stressed animals has antidepressant effects, but the molecular mechanisms are unclear. Since microglia normally interact with astrocytes and astrocytic purinergic 2Y1 receptor (P2Y1R) signaling plays an important role in regulating cellular crosstalk, we hypothesize that astrocytic P2Y1R signaling may mediate the antidepressant effects of microglia stimulation. Our results showed that a single injection of low-dose lipopolysaccharide (LPS) (100 μg/kg) elicited rapid antidepressant effects and a significant increase in adenosine triphosphate (ATP) levels in the dentate gyrus in chronically stressed mice, and that these effects of LPS were abolished by chemogenetic inhibition of microglia. Depletion of endogenous ATP, non-specific antagonization of purinergic receptors, or specific inhibition of P2Y1Rs, but not other purinergic receptors, by MRS2179 in the hippocampus abolished the antidepressant effects of low-dose LPS. Conditional gene knockout data showed that the antidepressant effect of low-dose LPS could not be observed in mice lacking P2Y1Rs in astrocytes but not in forebrain neurons. Chemogenetic inhibition of microglia in the dentate gyrus, specific deletion of P2Y1Rs in astrocytes and the absence of ATP abolished the increase in doublecortin (DCX)+ cells and brain-derived neurotrophic factor (BDNF) induced by a low dose of LPS in the dentate gyrus of stressed mice, and infusion of BDNF antibodies into the hippocampus simultaneously abolished the pro-neurogenesis and antidepressant effects of microglia stimulation in stressed mice. Taken together, these results suggest that ATP signaling mobilized by microglia stimulation has an antidepressant effect by triggering astrocytic P2Y1R-dependent synthesis of BDNF.
Collapse
Affiliation(s)
- Hanxiao Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001 Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001 Jiangsu, China
| | - Ying Ye
- Department of Ultrasound, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001 Jiangsu, China
| | - Chen Huang
- Department of Vascular Surgery, Affiliated Hospital of Nantong University, Jiangsu Province, #20 Xisi Road, Nantong 226001 Jiangsu, China
| | - Yunli Fang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001 Jiangsu, China
| | - Rongrong Yang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001 Jiangsu, China
| | - Micona Sun
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001 Jiangsu, China
| | - Jie Ren
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001 Jiangsu, China
| | - Rongrong Song
- Department of Emergency and Critical Care Medicine, Tongzhou People's Hospital, #999 Jianshe Road, Nantong 226300 Jiangsu, China
| | - Feng Xu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, First People's Hospital of Nantong City, #666 Shengli Road, Nantong 226006 Jiangsu, China
| | - Jianbin Su
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, First People's Hospital of Nantong City, #666 Shengli Road, Nantong 226006 Jiangsu, China
| | - Hongxiang Hong
- Department of Spine Surgery, Affiliated Hospital 2 of Nantong University, First People's Hospital of Nantong City, #666 Shengli Road, Nantong 226006 Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001 Jiangsu, China.
| |
Collapse
|
3
|
Bishnoi IR, Bordt EA. Sex and Region-Specific Differences in Microglial Morphology and Function Across Development. NEUROGLIA (BASEL, SWITZERLAND) 2025; 6:2. [PMID: 40181886 PMCID: PMC11967618 DOI: 10.3390/neuroglia6010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Microglia are exceptionally dynamic resident innate immune cells within the central nervous system, existing on a continuum of morphologies and functions throughout their lifespan. They play vital roles in response to injuries and infections, clearing cellular debris, and maintaining neural homeostasis throughout development. Emerging research suggests that microglia are strongly influenced by biological factors, including sex, developmental stage, and their local environment. This review synthesizes findings on sex differences in microglial morphology and function in key brain regions, including the frontal cortex, hippocampus, amygdala, hypothalamus, basal ganglia, and cerebellum, across the lifespan. Where available, we examine how gonadal hormones influence these microglial characteristics. Additionally, we highlight the limitations of relying solely on morphology to infer function and underscore the need for comprehensive, multimodal approaches to guide future research. Ultimately, this review aims to advance the dialogue on these spatiotemporally heterogeneous cells and their implications for sex differences in brain function and vulnerability to neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Indra R. Bishnoi
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Evan A. Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
4
|
Wang W, Liang W, Sun C, Liu S. Sex Differences in Depression: Insights from Multimodal Gray Matter Morphology and Peripheral Inflammatory Factors. Int J Mol Sci 2024; 25:13412. [PMID: 39769178 PMCID: PMC11677592 DOI: 10.3390/ijms252413412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Major depressive disorder (MDD) exhibits notable sex differences in prevalence and clinical and neurobiological manifestations. Though the relationship between peripheral inflammation and MDD-related brain changes is well studied, the role of sex as a modifying factor is underexplored. This study aims to assess how sex influences brain and inflammatory markers in MDD. We utilized voxel-based and surface-based morphometry to analyze gray matter (GM) structure, along with GM-based spatial statistics (GBSS) to examine GM microstructure among treatment-naive patients with depression (n = 174) and age-matched healthy controls (n = 133). We uncovered sex-by-diagnosis interactions in several limbic system structures, the frontoparietal operculum and temporal regions. Post hoc analyses revealed that male patients exhibit pronounced brain abnormalities, while no significant differences were noted in females despite their higher depressive scores. Additionally, heightened inflammation levels in MDD were observed in both sexes, with sex-specific effects on sex-specific brain phenotypes, particularly including a general negative correlation in males. Intriguingly, mediation analyses highlight the specific role of the parahippocampal gyrus (PHG) in mediating interleukin (IL)-8 and depression in men. The findings suggest that in clinical practice, it would be beneficial to prioritize sex-specific assessments and interventions for MDD. This includes recognizing the possibility that male patients may experience significant brain alterations, especially when identifying male patients who may underreport symptoms. Possible limitations encompass a small sample size and the cross-sectional design. In future research, the incorporation of longitudinal studies or diverse populations, while considering illness duration, will enhance our understanding of how inflammation interacts with brain changes in depression.
Collapse
Affiliation(s)
- Wenjun Wang
- Shandong Key Laboratory of Mental Disorders, Department of Anatomy and Neurobiology, Institute for Sectional Anatomy and Digital Human, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China
| | - Wenjia Liang
- Shandong Key Laboratory of Mental Disorders, Department of Anatomy and Neurobiology, Institute for Sectional Anatomy and Digital Human, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China
| | - Chenxi Sun
- Shandong Key Laboratory of Mental Disorders, Department of Anatomy and Neurobiology, Institute for Sectional Anatomy and Digital Human, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China
| | - Shuwei Liu
- Shandong Key Laboratory of Mental Disorders, Department of Anatomy and Neurobiology, Institute for Sectional Anatomy and Digital Human, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China
| |
Collapse
|
5
|
Abdulla ZI, Mineur YS, Crouse RB, Etherington IM, Yousuf H, Na JJ, Picciotto MR. Medial prefrontal cortex acetylcholine signaling mediates the ability to learn an active avoidance response following learned helplessness training. Neuropsychopharmacology 2024; 50:488-496. [PMID: 39362985 PMCID: PMC11631976 DOI: 10.1038/s41386-024-02003-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024]
Abstract
Increased brain levels of acetylcholine (ACh) have been observed in patients with depression, and increasing ACh levels pharmacologically can precipitate stress-related behaviors in humans and animals. Conversely, optimal ACh levels are required for cognition and memory. We hypothesize that excessive ACh signaling results in strengthening of negative encoding in which memory formation is aberrantly strengthened for stressful events. The medial prefrontal cortex (mPFC) is critical for both top-down control of stress-related circuits, and for encoding of sensory experiences. We therefore evaluated the role of ACh signaling in the mPFC in a learned helplessness task in which mice were exposed to repeated inescapable stressors followed by an active avoidance task. Using fiber photometry with a genetically-encoded ACh sensor, we found that ACh levels in the mPFC during exposure to inescapable stressors were positively correlated with later escape deficits in an active avoidance test in males, but not females. Consistent with these measurements, we found that both pharmacologically- and chemogenetically-induced increases in mPFC ACh levels resulted in escape deficits in both male and female mice, whereas chemogenetic inhibition of ACh neurons projecting to the mPFC improved escape performance in males, but impaired escape performance in females. These results highlight the adaptive role of ACh release in stress response, but also support the idea that sustained elevation of ACh contributes to maladaptive behaviors. Furthermore, mPFC ACh signaling may contribute to stress-based learning differentially in males and females.
Collapse
Affiliation(s)
- Zuhair I Abdulla
- Department of Psychiatry, Yale University, New Haven, CT, 06508, USA
| | - Yann S Mineur
- Department of Psychiatry, Yale University, New Haven, CT, 06508, USA
| | - Richard B Crouse
- Yale University Interdepartmental Neuroscience Program, New Haven, CT, USA
| | - Ian M Etherington
- Yale University Interdepartmental Neuroscience Program, New Haven, CT, USA
| | - Hanna Yousuf
- Department of Psychiatry, Yale University, New Haven, CT, 06508, USA
| | | | - Marina R Picciotto
- Department of Psychiatry, Yale University, New Haven, CT, 06508, USA.
- Yale University Interdepartmental Neuroscience Program, New Haven, CT, USA.
- Kavli Institute for Neuroscience at Yale, New Haven, CT, USA.
| |
Collapse
|
6
|
Smail MA, Lenz KM. Developmental functions of microglia: Impact of psychosocial and physiological early life stress. Neuropharmacology 2024; 258:110084. [PMID: 39025401 DOI: 10.1016/j.neuropharm.2024.110084] [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: 02/15/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Microglia play numerous important roles in brain development. From early embryonic stages through adolescence, these immune cells influence neuronal genesis and maturation, guide connectivity, and shape brain circuits. They also interact with other glial cells and structures, influencing the brain's supportive microenvironment. While this central role makes microglia essential, it means that early life perturbations to microglia can have widespread effects on brain development, potentially resulting in long-lasting behavioral impairments. Here, we will focus on the effects of early life psychosocial versus physiological stressors in rodent models. Psychosocial stress refers to perceived threats that lead to stress axes activation, including prenatal stress, or chronic postnatal stress, including maternal separation and resource scarcity. Physiological stress refers to physical threats, including maternal immune activation, postnatal infection, and traumatic brain injury. Differing sources of early life stress have varied impacts on microglia, and these effects are moderated by factors such as developmental age, brain region, and sex. Overall, these stressors appear to either 1) upregulate basal microglia numbers and activity throughout the lifespan, while possibly blunting their responsivity to subsequent stressors, or 2) shift the developmental curve of microglia, resulting in differential timing and function, impacting the critical periods they govern. Either could contribute to behavioral dysfunctions that occur after the resolution of early life stress. Exploring how different stressors impact microglia, as well as how multiple stressors interact to alter microglia's developmental functions, could deepen our understanding of how early life stress changes the brain's developmental trajectory. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Marissa A Smail
- Department of Psychology, Ohio State University, Columbus, OH, USA.
| | - Kathryn M Lenz
- Department of Psychology, Ohio State University, Columbus, OH, USA; Department of Neuroscience, Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, Ohio State University, Columbus, OH, USA
| |
Collapse
|
7
|
Grizzell JA, Clarity TT, Rodriguez RM, Marshall ZQ, Cooper MA. Effects of social dominance and acute social stress on morphology of microglia and structural integrity of the medial prefrontal cortex. Brain Behav Immun 2024; 122:353-367. [PMID: 39187049 PMCID: PMC11402560 DOI: 10.1016/j.bbi.2024.08.043] [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: 11/28/2023] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024] Open
Abstract
Chronic stress increases activity of the brain's innate immune system and impairs function of the medial prefrontal cortex (mPFC). However, whether acute stress triggers similar neuroimmune mechanisms is poorly understood. Across four studies, we used a Syrian hamster model to investigate whether acute stress drives changes in mPFC microglia in a time-, subregion-, and social status-dependent manner. We found that acute social defeat increased expression of ionized calcium binding adapter molecule 1 (Iba1) in the infralimbic (IL) and prelimbic (PL) and altered the morphology Iba1+ cells 1, 2, and 7 days after social defeat. We also investigated whether acute defeat induced tissue degeneration and reductions of synaptic plasticity 2 days post-defeat. We found that while social defeat increased deposition of cellular debris and reduced synaptophysin immunoreactivity in the PL and IL, treatment with minocycline protected against these cellular changes. Finally, we tested whether a reduced conditioned defeat response in dominant compared to subordinate hamsters was associated with changes in microglia reactivity in the IL and PL. We found that while subordinate hamsters and those without an established dominance relationships showed defeat-induced changes in morphology of Iba1+ cells and cellular degeneration, dominant hamsters showed resistance to these effects of social defeat. Taken together, these findings indicate that acute social defeat alters microglial morphology, increases markers of tissue degradation, and impairs structural integrity in the IL and PL, and that experience winning competitive interactions can specifically protect the IL and reduce stress vulnerability.
Collapse
Affiliation(s)
- J Alex Grizzell
- Neuroscience and Behavioral Biology Program, Emory University, United States; Department of Psychology, University of Tennessee Knoxville, United States; Department of Psychology and Neurosciences, University of Colorado Boulder, United States
| | - Thomas T Clarity
- Department of Psychology, University of Tennessee Knoxville, United States
| | - R Mason Rodriguez
- Department of Psychology, University of Tennessee Knoxville, United States
| | - Zachary Q Marshall
- Department of Psychology and Neurosciences, University of Colorado Boulder, United States
| | - Matthew A Cooper
- Department of Psychology, University of Tennessee Knoxville, United States.
| |
Collapse
|
8
|
Ye M, Zhu H, Lu X, Yang R, Wang H, Peng J, Pan H, Fang Y, Shi R, Li F, Chen Z, Hu W, Huang C. Central innate immunization induces tolerance against post-traumatic stress disorder-like behavior and neuroinflammatory responses in male mice. Brain Behav Immun 2024; 122:368-387. [PMID: 39197543 DOI: 10.1016/j.bbi.2024.08.049] [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: 01/07/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/01/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is a severe psychiatric disorder associated with abnormally elevated neuroinflammatory responses. Suppression of neuroinflammation is considered to be effective in ameliorating PTSD-like behaviors in rodents. Since pre-stimulation of microglia prior to stress exposure can prevent neuroinflammation, we hypothesized that pre-stimulation of microglia may prevent PTSD in animals. The results show that a single injection of a classical immune stimulant, lipopolysaccharide (LPS), at 50, 100 or 500, but not 10 μg/kg, one day before stress exposure, prevented the anxiety- and fear-like behaviors induced by modified single prolonged stress (mSPS). The time-dependent analysis shows that a single injection of LPS (100 μg/kg) either one or five, but not ten, days before stress prevented mSPS-induced anxiety- and fear-like behaviors. A second low-dose LPS injection 10 days after the first injection or a repeated LPS injection (4 × ) 10 days before stress induced tolerance to mSPS. Mechanistic studies show that a single injection of LPS one day before stress stimulation prevented mSPS-induced increases in levels of interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and IL-6 mRNA in the hippocampus and medial prefrontal cortex. Inhibition of microglia by pretreatment with minocycline or depletion of microglia by PLX3397 abolished the preventive effect of low-dose LPS pre-injection on mSPS-induced anxiety- and fear-like behavior and neuroinflammatory responses. These results suggest that pre-stimulation of microglia may prevent the development of PTSD-like behaviors by attenuating the development of neuroinflammatory responses. This could help to develop new strategies to prevent the damaging effects of harmful stress on the brain.
Collapse
Affiliation(s)
- Minxiu Ye
- Department of Pharmacy, Kunshan Hospital of Traditional Chinese Medicine, #388 Zuchongzhi South Road, Kunshan, Suzhou 215300, Jiangsu, China
| | - Haojie Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Rongrong Yang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China
| | - Hanxiao Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Jie Peng
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Hainan Pan
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Yunli Fang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Ruiting Shi
- Faculty of Humanities and Social Sciences, City University of Macau, Av. Parde Tomas Pereira, Macau, Taipa 999078, China
| | - Fu Li
- Department of Pharmacy, Changzhou Geriatric Hospital Affiliated to Soochow University, Changzhou No.7 People's Hospital, 288# Yanling East Road, Changzhou 213000, Jiangsu, China
| | - Zhuo Chen
- Invasive Technology Department, the Second Affiliated Hospital of Nantong University, First People's Hospital of Nantong City, #666 Shengli Road, Nantong 226006, Jiangsu, China
| | - Wenfeng Hu
- Department of Pharmacy, Affiliated Maternal and Child Health Hospital of Nantong University, #399 Shijidadao, Nantong 226007, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
9
|
Portugalov A, Peled G, Zorin S, Akirav I. Cannabidiol Modulates Neuroinflammatory Markers in a PTSD Model Conducted on Female Rats. Biomolecules 2024; 14:1384. [PMID: 39595561 PMCID: PMC11591736 DOI: 10.3390/biom14111384] [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: 08/23/2024] [Revised: 10/19/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is a debilitating neuropsychiatric condition closely linked to neuroinflammation, with a higher prevalence in women. Cannabidiol (CBD), a non-psychoactive cannabinoid, has shown promise as a potential treatment for PTSD. In this study, we used a PTSD model in which female rats were subjected to a severe foot shock followed by contextual situational reminders (SRs). Testing was conducted one month after exposure. The rats received daily CBD injections for three weeks during the SRs, from days 7 to 28. Two days after the final SR, the rats underwent five extinction trials, followed by the forced swim test (FST). After a five-day rest period, the rats were sacrificed, and brain tissues from the medial prefrontal cortex (mPFC) and ventral subiculum (vSUB) were analyzed for inflammatory markers. Chronic CBD treatment reversed impairments in fear extinction caused by shock and SR. It also reduced learned helplessness in the FST and decreased the upregulation of mPFC-il1β induced by shock and SRs. Additionally, exposure to shock and SRs downregulated mPFC-il6 while upregulating vSUB-il6. CBD treatment further downregulated il6 expression in the vSUB compared to the vehicle groups. Our findings show that CBD effectively inhibited the development of PTSD-like behaviors and suppressed neuroinflammation in the mPFC.
Collapse
Affiliation(s)
- Anna Portugalov
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel; (A.P.); (G.P.); (S.Z.)
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| | - Gaia Peled
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel; (A.P.); (G.P.); (S.Z.)
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| | - Sharon Zorin
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel; (A.P.); (G.P.); (S.Z.)
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| | - Irit Akirav
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel; (A.P.); (G.P.); (S.Z.)
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
10
|
Kuhn AM, Bosis KE, Wohleb ES. Looking Back to Move Forward: Research in Stress, Behavior, and Immune Function. Neuroimmunomodulation 2024; 31:211-229. [PMID: 39369707 DOI: 10.1159/000541592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND From the original studies investigating the effects of adrenal gland secretion to modern high-throughput multidimensional analyses, stress research has been a topic of scientific interest spanning just over a century. SUMMARY The objective of this review was to provide historical context for influential discoveries, surprising findings, and preclinical models in stress-related neuroimmune research. Furthermore, we summarize this work and present a current understanding of the stress pathways and their effects on the immune system and behavior. We focus on recent work demonstrating stress-induced immune changes within the brain and highlight studies investigating stress effects on microglia. Lastly, we conclude with potential areas for future investigation concerning microglia heterogeneity, bone marrow niches, and sex differences. KEY MESSAGES Stress is a phenomenon that ties together not only the central and peripheral nervous system, but the immune system as well. The cumulative effects of stress can enhance or suppress immune function, based on the intensity and duration of the stressor. These stress-induced immune alterations are associated with neurobiological changes, including structural remodeling of neurons and decreased neurogenesis, and these contribute to the development of behavioral and cognitive deficits. As such, research in this field has revealed important insights into neuroimmune communication as well as molecular and cellular mediators of complex behaviors relevant to psychiatric disorders.
Collapse
Affiliation(s)
- Alexander M Kuhn
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kelly E Bosis
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Eric S Wohleb
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
11
|
Takahashi A. Associations of the immune system in aggression traits and the role of microglia as mediators. Neuropharmacology 2024; 256:110021. [PMID: 38825308 DOI: 10.1016/j.neuropharm.2024.110021] [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: 11/26/2023] [Revised: 05/23/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
There is an important relationship between the immune system and aggressive behavior. Aggressive encounters acutely increase the levels of proinflammatory cytokines, and there are positive correlations between aggressive traits and peripheral proinflammatory cytokines. Endotoxin lipopolysaccharide (LPS) treatment, which results in peripheral immune activation, decreases aggressive behavior as one of the sickness behavioral symptoms. In contrast, certain brain infections and chronic interferon treatment are associated with increased aggression. Indeed, the effects of proinflammatory cytokines on the brain in aggressive behavior are bidirectional, depending on the type and dose of cytokine, target brain region, and type of aggression. Some studies have suggested that microglial activation and neuroinflammation influence intermale aggression in rodent models. In addition, pathological conditions as well as physiological levels of cytokines produced by microglia play an important role in social and aggressive behavior in adult animals. Furthermore, microglial function in early development is necessary for the establishment of the social brain and the expression of juvenile social behaviors, including play fighting. Overall, this review discusses the important link between the immune system and aggressive traits and the role of microglia as mediators of this link.
Collapse
Affiliation(s)
- Aki Takahashi
- Laboratory of Behavioral Neurobiology, Institute of Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
| |
Collapse
|
12
|
Farinha-Ferreira M, Magalhães DM, Neuparth-Sottomayor M, Rafael H, Miranda-Lourenço C, Sebastião AM. Unmoving and uninflamed: Characterizing neuroinflammatory dysfunction in the Wistar-Kyoto rat model of depression. J Neurochem 2024; 168:2443-2460. [PMID: 38430009 DOI: 10.1111/jnc.16083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/30/2024] [Accepted: 02/05/2024] [Indexed: 03/03/2024]
Abstract
Reductionistic research on depressive disorders has been hampered by the limitations of animal models. Recently, it has been hypothesized that neuroinflammation is a key player in depressive disorders. The Wistar-Kyoto (WKY) rat is an often-used animal model of depression, but no information so far exists on its neuroinflammatory profile. As such, we compared male young adult WKY rats to Wistar (WS) controls, with regard to both behavioral performance and brain levels of key neuroinflammatory markers. We first assessed anxiety- and depression-like behaviors in a battery consisting of the Elevated Plus Maze (EPM), the Novelty Suppressed Feeding (NSFT), Open Field (OFT), Social Interaction (SIT), Forced Swim (FST), Sucrose Preference (SPT), and Splash tests (ST). We found that WKY rats displayed increased NSFT feeding latency, decreased OFT center zone permanence, decreased EPM open arm permanence, decreased SIT interaction time, and increased immobility in the FST. However, WKY rats also evidenced marked hypolocomotion, which is likely to confound performance in such tests. Interestingly, WKY rats performed similarly, or even above, to WS levels in the SPT and ST, in which altered locomotion is not a significant confound. In a separate cohort, we assessed prefrontal cortex (PFC), hippocampus and amygdala levels of markers of astrocytic (GFAP, S100A10) and microglial (Iba1, CD86, Ym1) activation status, as well as of three key proinflammatory cytokines (IL-1β, IL-6, TNF-α). There were no significant differences between strains in any of these markers, in any of the regions assessed. Overall, results highlight that behavioral data obtained with WKY rats as a model of depression must be carefully interpreted, considering the marked locomotor activity deficits displayed. Furthermore, our data suggest that, despite WKY rats replicating many depression-associated neurobiological alterations, as shown by others, this is not the case for neuroinflammation-related alterations, thus representing a novel limitation of this model.
Collapse
Affiliation(s)
- Miguel Farinha-Ferreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Daniela M Magalhães
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Mariana Neuparth-Sottomayor
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Hugo Rafael
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Catarina Miranda-Lourenço
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
13
|
Myers T, Birmingham EA, Rhoads BT, McGrath AG, Miles NA, Schuldt CB, Briand LA. Post-weaning social isolation alters sociability in a sex-specific manner. Front Behav Neurosci 2024; 18:1444596. [PMID: 39267986 PMCID: PMC11390411 DOI: 10.3389/fnbeh.2024.1444596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
Adolescence is a critical period for brain development in humans and stress exposure during this time can have lasting effects on behavior and brain development. Social isolation and loneliness are particularly salient stressors that lead to detrimental mental health outcomes particularly in females, although most of the preclinical work on social isolation has been done in male animals. Our lab has developed a model of post-weaning adolescent social isolation that leads to increased drug reward sensitivity and altered neuronal structure in limbic brain regions. The current study utilized this model to determine the impact of adolescent social isolation on a three-chamber social interaction task both during adolescence and adulthood. We found that while post-weaning isolation does not alter social interaction during adolescence (PND45), it has sex-specific effects on social interaction in young adulthood (PND60), potentiating social interaction in male mice and decreasing it in female mice. As early life stress can activate microglia leading to alterations in neuronal pruning, we next examined the impact of inhibiting microglial activation with daily minocycline administration during the first 3 weeks of social isolation on these changes in social interaction. During adolescence, minocycline dampened social interaction in male mice, while having no effect in females. In contrast, during young adulthood, minocycline did not alter the impact of adolescent social isolation in males, with socially isolated males exhibiting higher levels of social interaction compared to their group housed counterparts. In females, adolescent minocycline treatment reversed the effect of social isolation leading to increased social interaction in the social isolation group, mimicking what is seen in naïve males. Taken together, adolescent social isolation leads to sex-specific effects on social interaction in young adulthood and adolescent minocycline treatment alters the effects of social isolation in females, but not males.
Collapse
Affiliation(s)
- Teneisha Myers
- Neuroscience Program, Temple University, Philadelphia, PA, United States
| | - Elizabeth A. Birmingham
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, United States
| | - Brigham T. Rhoads
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, United States
| | - Anna G. McGrath
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, United States
| | - Nylah A. Miles
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, United States
| | - Carmen B. Schuldt
- Neuroscience Program, Temple University, Philadelphia, PA, United States
| | - Lisa A. Briand
- Neuroscience Program, Temple University, Philadelphia, PA, United States
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, United States
| |
Collapse
|
14
|
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
|
15
|
Myers T, Birmingham EA, Rhoads BT, McGrath AG, Miles NA, Schuldt CB, Briand LA. Post-weaning social isolation alters sociability in a sex-specific manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.603129. [PMID: 39026733 PMCID: PMC11257562 DOI: 10.1101/2024.07.11.603129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Adolescence is a critical period for brain development in humans and stress exposure during this time can have lasting effects on behavior and brain development. Social isolation and loneliness are particularly salient stressors that lead to detrimental mental health outcomes particularly in females, although most of the preclinical work on social isolation has been done in male animals. Our lab has developed a model of post-weaning adolescent social isolation that leads to increased drug reward sensitivity and altered neuronal structure in limbic brain regions. The current study utilized this model to determine the impact of adolescent social isolation on a three-chamber social interaction task both during adolescence and adulthood. We found that while post-weaning isolation does not alter social interaction during adolescence (PND45), it has sex-specific effects on social interaction in adulthood (PND60), potentiating social interaction in male mice and decreasing it in female mice. As early life stress can activate microglia leading to alterations in neuronal pruning, we next examined the impact of inhibiting microglial activation with daily minocycline administration during the first three weeks of social isolation on these changes in social interaction. During adolescence, minocycline dampened social interaction in male mice, while having no effect in females. In contrast, during adulthood, minocycline did not alter the impact of adolescent social isolation in males, with socially isolated males exhibiting higher levels of social interaction compared to their group housed counterparts. In females, adolescent minocycline treatment reversed the effect of social isolation leading to increased social interaction in the social isolation group, mimicking what is seen in naïve males. Taken together, adolescent social isolation leads to sex-specific effects on social interaction in adulthood and adolescent minocycline treatment alters the effects of social isolation in females, but not males.
Collapse
|
16
|
Rawls A, Nyugen D, Dziabis J, Anbarci D, Clark M, Dzirasa K, Bilbo SD. Microglial MyD88-dependent pathways are regulated in a sex-specific manner in the context of HMGB1-induced anxiety. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590482. [PMID: 38712142 PMCID: PMC11071353 DOI: 10.1101/2024.04.22.590482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Chronic stress is a significant risk factor for the development and recurrence of anxiety disorders. Chronic stress impacts the immune system, causing microglial functional alterations in the medial prefrontal cortex (mPFC), a brain region involved in the pathogenesis of anxiety. High mobility group box 1 protein (HMGB1) is an established modulator of neuronal firing and a potent pro-inflammatory stimulus released from neuronal and non-neuronal cells following stress. HMGB1, in the context of stress, acts as a danger-associated molecular pattern (DAMP), instigating robust proinflammatory responses throughout the brain, so much so that localized drug delivery of HMGB1 alters behavior in the absence of any other forms of stress, i.e., social isolation, or behavioral stress models. Few studies have investigated the molecular mechanisms that underlie HMGB1-associated behavioral effects in a cell-specific manner. The aim of this study is to investigate cellular and molecular mechanisms underlying HMGB1-induced behavioral dysfunction with regard to cell-type specificity and potential sex differences. Here, we report that both male and female mice exhibited anxiety-like behavior following increased HMGB1 in the mPFC as well as changes in microglial morphology. Interestingly, our results demonstrate that HMGB1-induced anxiety may be mediated by distinct microglial MyD88-dependent mechanisms in females compared to males. This study supports the hypothesis that MyD88 signaling in microglia may be a crucial mediator of the stress response in adult female mice.
Collapse
Affiliation(s)
- Ashleigh Rawls
- Department of Pharmacology, Duke University, Durham, North Carolina, United States of America
| | - Dang Nyugen
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, United States of America
| | - Julia Dziabis
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, United States of America
| | - Dilara Anbarci
- Department of Cell Biology, Duke University, Durham, North Carolina, United States of America
| | - Madeline Clark
- Department of Neurobiology, Duke University, Durham, North Carolina, United States of America
| | - Kafui Dzirasa
- Department of Neurobiology, Duke University, Durham, North Carolina, United States of America
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, United States of America
- Department of Neurobiology, Duke University, Durham, North Carolina, United States of America
| |
Collapse
|
17
|
Xie G, Gao X, Guo Q, Liang H, Yao L, Li W, Ma B, Wu N, Han X, Li J. Cannabidiol ameliorates PTSD-like symptoms by inhibiting neuroinflammation through its action on CB2 receptors in the brain of male mice. Brain Behav Immun 2024; 119:945-964. [PMID: 38759736 DOI: 10.1016/j.bbi.2024.05.016] [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: 11/14/2023] [Revised: 05/05/2024] [Accepted: 05/12/2024] [Indexed: 05/19/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is a debilitating mental health disease related to traumatic experience, and its treatment outcomes are unsatisfactory. Accumulating research has indicated that cannabidiol (CBD) exhibits anti-PTSD effects, however, the underlying mechanism of CBD remains inadequately investigated. Although many studies pertaining to PTSD have primarily focused on aberrations in neuronal functioning, the present study aimed to elucidate the involvement and functionality of microglia/macrophages in PTSD while also investigated the modulatory effects of CBD on neuroinflammation associated with this condition. We constructed a modified single-prolonged stress (SPS) mice PTSD model and verified the PTSD-related behaviors by various behavioral tests (contextual freezing test, elevated plus maze test, tail suspension test and novel object recognition test). We observed a significant upregulation of Iba-1 and alteration of microglial/macrophage morphology within the prefrontal cortex and hippocampus, but not the amygdala, two weeks after the PTSD-related stress, suggesting a persistent neuroinflammatory phenotype in the PTSD-modeled group. CBD (10 mg/kg, i.p.) inhibited all PTSD-related behaviors and reversed the alterations in both microglial/macrophage quantity and morphology when administered prior to behavioral assessments. We further found increased pro-inflammatory factors, decreased PSD95 expression, and impaired synaptic density in the hippocampus of the modeled group, all of which were also restored by CBD treatment. CBD dramatically increased the level of anandamide, one of the endocannabinoids, and cannabinoid type 2 receptors (CB2Rs) transcripts in the hippocampus compared with PTSD-modeled group. Importantly, we discovered the expression of CB2Rs mRNA in Arg-1-positive cells in vivo and found that the behavioral effects of CBD were diminished by CB2Rs antagonist AM630 (1 mg/kg, i.p.) and both the behavioral and molecular effects of CBD were abolished in CB2Rs knockout mice. These findings suggest that CBD would alleviate PTSD-like behaviors in mice by suppressing PTSD-related neuroinflammation and upregulation and activation of CB2Rs may serve as one of the underlying mechanisms for this therapeutic effect. The present study offers innovative experimental evidence supporting the utilization of CBD in PTSD treatment from the perspective of its regulation of neuroinflammation, and paves the way for leveraging the endocannabinoid system to regulate neuroinflammation as a potential therapeutic approach for psychiatric disorders.
Collapse
Affiliation(s)
- Guanbo Xie
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Xinwei Gao
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Qingchun Guo
- Chinese Institute for Brain Research, Beijing 102206, China; School of Biomedical Engineering, Capital Medical University, Beijing 100069, China
| | - Haizhen Liang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Lan Yao
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Wenjuan Li
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Baiping Ma
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ning Wu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Xiao Han
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Jin Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| |
Collapse
|
18
|
Antunes GF, Gouveia FV, Kuroki MA, Oliveira Martins D, Pagano RDL, Pinheiro Campos AC, Martinez RCR. Neuroinflammation in the prefrontal-amygdala-hippocampus network is associated with maladaptive avoidance behaviour. Heliyon 2024; 10:e30427. [PMID: 38694029 PMCID: PMC11061725 DOI: 10.1016/j.heliyon.2024.e30427] [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: 10/16/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/03/2024] Open
Abstract
Maladaptive avoidance behaviour is often observed in patients suffering from anxiety and trauma- and stressor-related disorders. The prefrontal-amygdala-hippocampus network is implicated in learning and memory consolidation. Neuroinflammation in this circuitry alters network dynamics, resulting in maladaptive avoidance behaviour. The two-way active avoidance test is a well-established translational model for assessing avoidance responses to stressful situations. While some animals learn the task and show adaptive avoidance (AA), others show strong fear responses to the test environment and maladaptive avoidance (MA). Here, we investigated if a distinct neuroinflammation pattern in the prefrontal-amygdala-hippocampus network underlies the behavioural difference observed in these animals. Wistar rats were tested 8 times and categorized as AA or MA based on behaviour. Brain recovery followed for the analysis of neuroinflammatory markers in this network. AA and MA presented distinct patterns of neuroinflammation, with MA showing increased astrocyte, EAAT-2, IL-1β, IL-17 and TNF-ɑ in the amygdala. This neuroinflammatory pattern may underlie these animals' fear response and maladaptive avoidance. Further studies are warranted to determine the specific contributions of each inflammatory factor, as well as the possibility of treating maladaptive avoidance behaviour in patients with psychiatric disorders with anti-inflammatory drugs targeting the amygdala.
Collapse
Affiliation(s)
| | - Flavia Venetucci Gouveia
- Division of Neuroscience, Hospital Sirio-Libanes, Sao Paulo, Brazil
- Neuroscience and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | | | | | | | | | - Raquel Chacon Ruiz Martinez
- Division of Neuroscience, Hospital Sirio-Libanes, Sao Paulo, Brazil
- LIM/23, Institute of Psychiatry, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| |
Collapse
|
19
|
McCallum RT, Thériault RK, Manduca JD, Russell ISB, Culmer AM, Doost JS, Martino TA, Perreault ML. Nrf2 activation rescues stress-induced depression-like behaviour and inflammatory responses in male but not female rats. Biol Sex Differ 2024; 15:16. [PMID: 38350966 PMCID: PMC10863247 DOI: 10.1186/s13293-024-00589-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/31/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is a recurring affective disorder that is two times more prevalent in females than males. Evidence supports immune system dysfunction as a major contributing factor to MDD, notably in a sexually dimorphic manner. Nuclear factor erythroid 2-related factor 2 (Nrf2), a regulator of antioxidant signalling during inflammation, is dysregulated in many chronic inflammatory disorders; however, its role in depression and the associated sex differences have yet to be explored. Here, we investigated the sex-specific antidepressant and immunomodulatory effects of the potent Nrf2 activator dimethyl fumarate (DMF), as well as the associated gene expression profiles. METHODS Male and female rats were treated with vehicle or DMF (25 mg/kg) whilst subjected to 8 weeks of chronic unpredictable stress. The effect of DMF treatment on stress-induced depression- and anxiety-like behaviours, as well as deficits in recognition and spatial learning and memory were then assessed. Sex differences in hippocampal (HIP) microglial activation and gene expression response were also evaluated. RESULTS DMF treatment during stress exposure had antidepressant effects in male but not female rats, with no anxiolytic effects in either sex. Recognition learning and memory and spatial learning and memory were impaired in chronically stressed males and females, respectively, and DMF treatment rescued these deficits. DMF treatment also prevented stress-induced HIP microglial activation in males. Conversely, females displayed no HIP microglial activation associated with stress exposure. Last, chronic stress elicited sex-specific alterations in HIP gene expression, many of which were normalized in animals treated with DMF. Of note, most of the differentially expressed genes in males normalized by DMF were related to antioxidant, inflammatory or immune responses. CONCLUSIONS Collectively, these findings support a greater role of immune processes in males than females in a rodent model of depression. This suggests that pharmacotherapies that target Nrf2 have the potential to be an effective sex-specific treatment for depression.
Collapse
Affiliation(s)
- Ryan T McCallum
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Rachel-Karson Thériault
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Joshua D Manduca
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Isaac S B Russell
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Angel M Culmer
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Janan Shoja Doost
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Tami A Martino
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Melissa L Perreault
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
20
|
Zhao C, Chen Z, Lu X, Hu W, Yang R, Lu Q, Chen B, Huang C. Microglia-Dependent Reversal of Depression-Like Behaviors in Chronically Stressed Mice by Administration of a Specific Immuno-stimulant β-Glucan. Neurochem Res 2024; 49:519-531. [PMID: 37962706 DOI: 10.1007/s11064-023-04056-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: 07/21/2023] [Revised: 10/19/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023]
Abstract
In recent years, the decline of microglia in the hippocampus has been shown to play a role in the development of depression, and its reversal shows marked antidepressant-like effects. β-glucan is a polysaccharide from Saccharomyces cerevisiae and has numerous beneficial effects on the nervous system, including improving axon regeneration and cognition. Considering its immuno-stimulatory activities in cultured microglia and brain tissues, we hypothesize that β-glucan may be a potential candidate to correct the functional deficiency of microglia and thereby alleviate depression-like behaviors in chronically stressed animals. An expected, our results showed that a single injection of β-glucan 5 h before behavioral tests at a dose of 10 or 20 mg/kg, but not at a dose of 5 mg/kg, reversed the depression-like behavior induced by chronic stress in mice in the tail suspension test, forced swimming test, and sucrose preference test. The effect of β-glucan (20 mg/kg) also showed time-dependent properties that were statistically significant 5 and 8, but not 3, hours after drug injection and persisted for at least 7 days. Fourteen days after β-glucan injection, no antidepressant-like effect was observed anymore. However, this effect was overcome by a second β-glucan injection (20 mg/kg) 14 days after the first β-glucan injection. Stimulation of microglia appeared to mediate the antidepressant-like effect of β-glucan, because both inhibition of microglia and their depletion prevented the antidepressant-like effect of β-glucan. Based on these effects of β-glucan, β-glucan administration could be developed as a new strategy for the treatment of depression.
Collapse
Affiliation(s)
- Cheng Zhao
- Department of Pharmacy, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong, 226001, Jiangsu, China.
| | - Zhuo Chen
- Invasive Technology Department, First People's Hospital of Nantong City, the Second Affiliated Hospital of Nantong University, #666 Shengli Road, Nantong, 226006, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu Province, China
| | - Wenfeng Hu
- Department of Pharmacy, Affiliated Maternal and Child Health Hospital of Nantong University, #399 Shijidadao, Nantong, 226007, China
| | - Rongrong Yang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Qun Lu
- Department of Pharmacy, Nantong Third Hospital Affiliated to Nantong University, #60 Middle Qingnian Road, Nantong, 226006, Jiangsu, China
| | - Bingran Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu Province, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu Province, China.
| |
Collapse
|
21
|
Dadwal S, Heneka MT. Microglia heterogeneity in health and disease. FEBS Open Bio 2024; 14:217-229. [PMID: 37945346 PMCID: PMC10839410 DOI: 10.1002/2211-5463.13735] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/12/2023] [Accepted: 11/08/2023] [Indexed: 11/12/2023] Open
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), have received significant attention due to their critical roles in maintaining brain homeostasis and mediating cerebral immune responses. Understanding the origin of microglia has been a subject of great interest, and emerging evidence suggests that microglia consist of multiple subpopulations with unique molecular and functional characteristics. These subpopulations of microglia may exhibit specialized roles in response to different environmental cues as in disease conditions. The newfound understanding of microglial heterogeneity has significant implications for elucidating their roles in both physiological and pathological conditions. In the context of disease, microglia have been studied rigorously as they play a very important role in neuroinflammation. Dysregulated microglial activation and function contribute to chronic inflammation. Further exploration of microglial heterogeneity and their interactions with other cell types in the CNS will undoubtedly pave the way to novel therapeutic strategies targeting microglia-mediated pathologies. In this review, we discuss the latest advances in the field of microglia research, focusing specifically on the origin and subpopulations of microglia, the populations of microglia types in the brains of patients with neurodegenerative diseases, and how microglia are regulated in the healthy CNS.
Collapse
Affiliation(s)
- Shilauni Dadwal
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgBelvalLuxembourg
| | - Michael T. Heneka
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgBelvalLuxembourg
- Division of Infectious Diseases and ImmunologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| |
Collapse
|
22
|
Melbourne JK, Wooden JI, Carlson ER, Anasooya Shaji C, Nixon K. Neuroimmune Activation and Microglia Reactivity in Female Rats Following Alcohol Dependence. Int J Mol Sci 2024; 25:1603. [PMID: 38338883 PMCID: PMC10855949 DOI: 10.3390/ijms25031603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/12/2024] [Accepted: 01/13/2024] [Indexed: 02/12/2024] Open
Abstract
The rates of alcohol use disorder among women are growing, yet little is known about how the female brain is affected by alcohol. The neuroimmune system, and specifically microglia, have been implicated in mediating alcohol neurotoxicity, but most preclinical studies have focused on males. Further, few studies have considered changes to the microglial phenotype when examining the effects of ethanol on brain structure and function. Therefore, we quantified microglial reactivity in female rats using a binge model of alcohol dependence, assessed through morphological and phenotypic marker expression, coupled with regional cytokine levels. In a time- and region-dependent manner, alcohol altered the microglial number and morphology, including the soma and process area, and the overall complexity within the corticolimbic regions examined, but no significant increases in the proinflammatory markers MHCII or CD68 were observed. The majority of cytokine and growth factor levels examined were similarly unchanged. However, the expression of the proinflammatory cytokine TNFα was increased, and the anti-inflammatory IL-10, decreased. Thus, female rats showed subtle differences in neuroimmune reactivity compared to past work in males, consistent with reports of enhanced neuroimmune responses in females across the literature. These data suggest that specific neuroimmune reactions in females may impact their susceptibility to alcohol neurotoxicity and other neurodegenerative events with microglial contributions.
Collapse
Affiliation(s)
| | | | | | | | - Kimberly Nixon
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (J.K.M.)
| |
Collapse
|
23
|
Bollinger JL, Horchar MJ, Wohleb ES. Repeated Activation of Pyramidal Neurons in the Prefrontal Cortex Alters Microglial Phenotype in Male Mice. J Pharmacol Exp Ther 2024; 388:715-723. [PMID: 38129124 PMCID: PMC10801771 DOI: 10.1124/jpet.123.001759] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 11/01/2023] [Accepted: 11/06/2023] [Indexed: 12/23/2023] Open
Abstract
Aberrant neuronal activity in the cortex alters microglia phenotype and function in several contexts, including chronic psychologic stress and neurodegenerative disease. Recent findings even suggest that heightened levels of neuronal activity spur microglia to phagocytose synapses, with potential impacts on cognition and behavior. Thus, the present studies were designed to determine if activation of neurons alone-independent of disease or dysfunction-is sufficient to alter microglial phenotype in the medial prefrontal cortex (mPFC), a brain region critical in emotion regulation and cognition. In these studies, we used both an adeno-associated virus-mediated and Cre-dependent chemogenetic [designer receptors exclusively activated by designer drugs (DREADD)] approach to repeatedly activate excitatory pyramidal neurons (CaMKIIa+) neurons in the mPFC. Various molecular, cytometric, and behavioral endpoints were examined. Recurrent DREADD-induced neuronal activation led to pronounced changes in microglial density, clustering, and morphology in the mPFC and increased microglia-specific transcripts implicated in synaptic pruning (e.g., Csf1r, Cd11b). Further analyses revealed that the magnitude of DREADD-induced neuronal activation was significantly correlated with measures of microglial morphology in the mPFC. These alterations in microglial phenotype coincided with an increase in microglial lysosome volume in the mPFC and selective deficits in working memory function. Altogether, these findings indicate that repeated neuronal activation alone is sufficient to drive changes in microglia phenotype and function in the mPFC. Future studies using optogenetic and chemogenetic approaches to manipulate neural circuits need to consider microglial and other nonneuronal contributions to physiologic and behavioral outcomes. SIGNIFICANCE STATEMENT: Microglia are highly attuned to fluctuations in neuronal activity. Here we show that repeated activation of pyramidal neurons in the prefrontal cortex induces broad changes in microglia phenotype; this includes upregulation of pathways associated with microglial proliferation, microglia-neuron interactions, and lysosome induction. Our findings suggest that studies using chemogenetic or optogenetic approaches to manipulate neural circuits should be mindful of indirect effects on nonneuronal cells and their potential contribution to measured outcomes.
Collapse
Affiliation(s)
- Justin L Bollinger
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Matthew J Horchar
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Eric S Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
24
|
Delpech JC, Valdearcos M, Nadjar A. Stress and Microglia: A Double-edged Relationship. ADVANCES IN NEUROBIOLOGY 2024; 37:333-342. [PMID: 39207700 DOI: 10.1007/978-3-031-55529-9_18] [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
Microglia are highly dynamic cells and acquire different activation states to modulate their multiple functions, which are tightly regulated by the central nervous system microenvironment in which they reside. In response to stress, that is to the appearance of non-physiological signals in their vicinity, microglia will adapt their function in order to promote a return to brain homeostasis. However, when these stress signals are chronically present, microglial response may not be adapted and lead to the establishment of a pathological state. The aim of this book chapter is to examine the substantial literature around the ability of acute and chronic stressors to affect microglial structure and function, with a special focus on psychosocial and nutritional stresses. We also discuss the molecular mechanisms known to date that explain the link between exposure to stressors and microglial activation.
Collapse
Affiliation(s)
| | - Martin Valdearcos
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Agnès Nadjar
- Neurocentre Magendie, U1215, INSERM-Université de Bordeaux, Bordeaux, France.
- Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
25
|
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
|
26
|
Maciejska A, Pomierny B, Krzyżanowska W, Starek-Świechowicz B, Skórkowska A, Budziszewska B. Mechanism of Microglial Cell Activation in the Benzophenone-3 Exposure Model. Neuroscience 2023; 533:63-76. [PMID: 37827357 DOI: 10.1016/j.neuroscience.2023.10.002] [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: 06/07/2023] [Revised: 09/19/2023] [Accepted: 10/05/2023] [Indexed: 10/14/2023]
Abstract
Benzophenone-3 (BP-3) is the most commonly used UV filter in cosmetics, which is absorbed through the skin and crosses the blood-brain barrier. This compound increases extracellular glutamate concentrations, lipid peroxidation, the number of microglia cells and induces process of apoptosis. The aim of this study was to determine the effect of BP-3 on the activation and polarization of microglial cells in the frontal cortex and hippocampus of adult male rats exposed to BP-3 prenatally and then for two weeks in adulthood. It has been found, that exposure to BP-3 reduced the expression of the marker of the M2 phenotype of glial cells in both examined brain structures. An increase in the CD86/CD206 microglial phenotype ratio, expression of transcription factor NFκB and activity of caspase-1 were observed only in the frontal cortex, whereas BP-3 increased the level of glucocorticoid receptors in the hippocampus. The in vitro study conducted in the primary culture of rat frontal cortical microglia cells showed that BP-3 increased the LPS-stimulated release of pro-inflammatory cytokines IL-1α, IL-1β, TNFα, but in cultures without LPS there was decreased IL-1α, IL-6 and TNFα production, while the IL-18 and IP-10 was elevated. The obtained results indicate that differences in the level of immunoactivation between the frontal cortex and the hippocampus may result from the action of this compound on glucocorticoid receptors. In turn, changes in cytokine production in microglial cells indicate that BP-3 aggravates the LPS-induced immunoactivation.
Collapse
Affiliation(s)
- Alicja Maciejska
- Department of Toxicological Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland.
| | - Bartosz Pomierny
- Department of Toxicological Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Weronika Krzyżanowska
- Department of Toxicological Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Beata Starek-Świechowicz
- Department of Toxicological Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Alicja Skórkowska
- Department of Toxicological Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Bogusława Budziszewska
- Department of Toxicological Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| |
Collapse
|
27
|
Abdulla ZI, Mineur YS, Crouse RB, Etherington IM, Yousuf H, Na JJ, Picciotto MR. Acetylcholine signaling in the medial prefrontal cortex mediates the ability to learn an active avoidance response following learned helplessness training. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.23.559126. [PMID: 37790481 PMCID: PMC10542494 DOI: 10.1101/2023.09.23.559126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Increased brain levels of acetylcholine (ACh) are observed in subsets of patients with depression and increasing ACh levels chronically can precipitate stress-related behaviors in humans and animals. Conversely, optimal ACh levels are required for cognition and memory. We hypothesize that ACh signaling is important for encoding both appetitive and stress-relevant memories, but that excessive increases in ACh result in a negative encoding bias in which memory formation of a stressful event is aberrantly strengthened, potentially contributing to the excessive focus on negative experience that could lead to depressive symptoms. The medial prefrontal cortex (mPFC) is critical to control the limbic system to filter exteroceptive cues and stress-related circuits. We therefore evaluated the role of ACh signaling in the mPFC in a learned helplessness task in which mice were exposed to repeated inescapable stressors followed by an active avoidance task. Using fiber photometry with a genetically-encoded ACh sensor, we found that ACh levels in the mPFC during exposure to inescapable stressors were positively correlated with later escape deficits in an active avoidance test in males, but not females. Consistent with these measurements, we found that both pharmacologically- and chemogenetically-induced increases in mPFC ACh levels resulted in escape deficits in both male and female mice, whereas chemogenetic inhibition of ACh neurons projecting to the mPFC improved escape performance in males, but impaired escape performance in females. These results highlight the adaptive role of ACh release in stress response, but also support the idea that sustained elevated ACh levels contribute to maladaptive behaviors. Furthermore, mPFC ACh signaling may contribute to depressive symptomology differentially in males and females.
Collapse
Affiliation(s)
- Zuhair I. Abdulla
- Department of Psychiatry, Yale University, 34 Park Street, New Haven, CT 06508, USA
| | - Yann S. Mineur
- Department of Psychiatry, Yale University, 34 Park Street, New Haven, CT 06508, USA
| | | | | | - Hanna Yousuf
- Department of Psychiatry, Yale University, 34 Park Street, New Haven, CT 06508, USA
| | | | - Marina R. Picciotto
- Department of Psychiatry, Yale University, 34 Park Street, New Haven, CT 06508, USA
- Yale University Interdepartmental Neuroscience Program
- Kavli Institute for Neuroscience at Yale
| |
Collapse
|
28
|
Matthews DB, Scaletty S, Trapp S, Schreiber A, Rossmann G, Imhoff B, Petersilka Q, Kastner A, Pauly J, Nixon K. Chronic intermittent ethanol exposure during adolescence produces sex- and age-dependent changes in anxiety and cognition without changes in microglia reactivity late in life. Front Behav Neurosci 2023; 17:1223883. [PMID: 37589035 PMCID: PMC10427154 DOI: 10.3389/fnbeh.2023.1223883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023] Open
Abstract
Binge-like ethanol exposure during adolescence has been shown to produce long lasting effects in animal models including anxiety-like behavior that can last into young adulthood and impairments in cognition that can last throughout most of the lifespan. However, little research has investigated if binge-like ethanol exposure during adolescence produces persistent anxiety-like behavior and concomitantly impairs cognition late in life. Furthermore, few studies have investigated such behavioral effects in both female and male rats over the lifespan. Finally, it is yet to be determined if binge-like ethanol exposure during adolescence alters microglia activation in relevant brain regions late in life. In the present study female and male adolescent rats were exposed to either 3.0 or 5.0 g/kg ethanol, or water control, in a chronic intermittent pattern before being tested in the elevated plus maze and open field task over the next ∼18 months. Animals were then trained in a spatial reference task via the Morris water maze before having their behavioral flexibility tested. Finally, brains were removed, sectioned and presumptive microglia activation determined using autoradiography for [3H]PK11195 binding. Males, but not females, displayed an anxiety-like phenotype initially following the chronic intermittent ethanol exposure paradigm which resolved in adulthood. Further, males but not females had altered spatial reference learning and impaired behavioral flexibility late in life. Conversely, [3H]PK11195 binding was significantly elevated in females compared to males late in life and the level of microglia activation interacted as a function of sex and brain regions, but there was no long-term outcome related to adolescent alcohol exposure. These data further confirm that binge-like ethanol exposure during adolescence produces alterations in behavior that can last throughout the lifespan. In addition, the data suggest that microglia activation late in life is not exacerbated by prior binge-like ethanol exposure during adolescence but the expression is sex- and brain region-dependent across the lifespan.
Collapse
Affiliation(s)
- Douglas B. Matthews
- Department of Psychology, University of Wisconsin–Eau Claire, Eau Claire, WI, United States
| | - Samantha Scaletty
- Department of Psychology, University of Wisconsin–Eau Claire, Eau Claire, WI, United States
| | - Sarah Trapp
- Department of Psychology, University of Wisconsin–Eau Claire, Eau Claire, WI, United States
| | - Areonna Schreiber
- Department of Psychology, University of Wisconsin–Eau Claire, Eau Claire, WI, United States
| | - Gillian Rossmann
- Department of Psychology, University of Wisconsin–Eau Claire, Eau Claire, WI, United States
| | - Bailey Imhoff
- Department of Psychology, University of Wisconsin–Eau Claire, Eau Claire, WI, United States
| | - Quinn Petersilka
- Department of Psychology, University of Wisconsin–Eau Claire, Eau Claire, WI, United States
| | - Abigail Kastner
- Department of Psychology, University of Wisconsin–Eau Claire, Eau Claire, WI, United States
| | - Jim Pauly
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Kimberly Nixon
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
29
|
Bollinger JL, Dadosky DT, Flurer JK, Rainer IL, Woodburn SC, Wohleb ES. Microglial P2Y12 mediates chronic stress-induced synapse loss in the prefrontal cortex and associated behavioral consequences. Neuropsychopharmacology 2023; 48:1347-1357. [PMID: 36517583 PMCID: PMC10354016 DOI: 10.1038/s41386-022-01519-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022]
Abstract
Chronic unpredictable stress (CUS) drives microglia-mediated neuronal remodeling and synapse loss in the prefrontal cortex (PFC), contributing to deficits in cognition and behavior. However, it remains unclear what mechanisms guide microglia-neuron interactions in stress. Evidence indicates that neuronal activity-dependent purinergic signaling directs microglial processes and synaptic engagement via P2Y12, a purinergic receptor exclusively expressed by microglia in the brain. Stress alters excitatory neurotransmission in the PFC, thus we aimed to determine if P2Y12 signaling promotes functional changes in microglia in chronic stress. Here we used genetic ablation of P2Y12 (P2ry12-/-) or pharmacological blockade (clopidogrel, ticagrelor) to examine the role of purinergic signaling in stress-induced microglia-neuron interaction. Multiple behavioral, physiological, and cytometric endpoints were analyzed. Deletion of P2Y12 led to a number of fundamental alterations in the PFC, including the heightened microglial number and increased dendritic spine density. Flow cytometry revealed that microglia in P2ry12-/- mice had shifts in surface levels of CX3CR1, CSF1R, and CD11b, suggesting changes in synaptic engagement and phagocytosis in the PFC. In line with this, pharmacological blockade of P2Y12 prevented CUS-induced increases in the proportion of microglia with neuronal inclusions, limited dendritic spine loss in the PFC, and attenuated alterations in stress coping behavior and working memory function. Overall, these findings indicate that microglial P2Y12 is a critical mediator of stress-induced synapse loss in the PFC and subsequent behavioral deficits.
Collapse
Affiliation(s)
- Justin L Bollinger
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - David T Dadosky
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - James K Flurer
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ivanka L Rainer
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Samuel C Woodburn
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
30
|
Wu J, Li Y, Huang Y, Liu L, Zhang H, Nagy C, Tan X, Cheng K, Liu Y, Pu J, Wang H, Wu Q, Perry SW, Turecki G, Wong ML, Licinio J, Zheng P, Xie P. Integrating spatial and single-nucleus transcriptomic data elucidates microglial-specific responses in female cynomolgus macaques with depressive-like behaviors. Nat Neurosci 2023; 26:1352-1364. [PMID: 37443281 DOI: 10.1038/s41593-023-01379-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/12/2023] [Indexed: 07/15/2023]
Abstract
Major depressive disorder represents a serious public health challenge worldwide; however, the underlying cellular and molecular mechanisms are mostly unknown. Here, we profile the dorsolateral prefrontal cortex of female cynomolgus macaques with social stress-associated depressive-like behaviors using single-nucleus RNA-sequencing and spatial transcriptomics. We find gene expression changes associated with depressive-like behaviors mostly in microglia, and we report a pro-inflammatory microglia subpopulation enriched in the depressive-like condition. Single-nucleus RNA-sequencing data result in the identification of six enriched gene modules associated with depressive-like behaviors, and these modules are further resolved by spatial transcriptomics. Gene modules associated with huddle and sit alone behaviors are expressed in neurons and oligodendrocytes of the superficial cortical layer, while gene modules associated with locomotion and amicable behaviors are enriched in microglia and astrocytes in mid-to-deep cortical layers. The depressive-like behavior associated microglia subpopulation is enriched in deep cortical layers. In summary, our findings show cell-type and cortical layer-specific gene expression changes and identify one microglia subpopulation associated with depressive-like behaviors in female non-human primates.
Collapse
Affiliation(s)
- Jing Wu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Yifan Li
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Yu Huang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Lanxiang Liu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Hanping Zhang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Xunmin Tan
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Ke Cheng
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiyun Liu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Juncai Pu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haiyang Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Qingyuan Wu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Seth W Perry
- Department of Psychiatry, College of Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Ma-Li Wong
- Department of Psychiatry, College of Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Julio Licinio
- Department of Psychiatry, College of Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Peng Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Jinfeng Laboratory, Chongqing, China.
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Jinfeng Laboratory, Chongqing, China.
| |
Collapse
|
31
|
Williams ZA, Szyszkowicz JK, Osborne N, Allehyany B, Nadon C, Udechukwu MC, Santos A, Audet MC. Sex-specific effects of voluntary wheel running on behavior and the gut microbiota-immune-brain axis in mice. Brain Behav Immun Health 2023; 30:100628. [PMID: 37396336 PMCID: PMC10308214 DOI: 10.1016/j.bbih.2023.100628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 04/21/2023] [Indexed: 07/04/2023] Open
Abstract
Physical exercise has been positioned as a promising strategy to prevent and/or alleviate anxiety and depression, but the biological processes associated with its effects on mental health have yet to be entirely determined. Although the prevalence of depression and anxiety in women is about twice that of men, very few studies have examined whether physical exercise could affect mental health differently according to sex. This study examined, in singly-housed mice, the sex-specific effects of voluntary exercise on depressive- and anxiety-like behaviors as well as on different markers along the gut microbiota-immune-brain axis. Male and female C57BL/6N mice had voluntary access to running wheels in their home-cages for 24 days or were left undisturbed in identical home-cages without running wheels. Behaviors were then examined in the open field, splash, elevated plus maze, and tail suspension tests. Gene expression of pro-inflammatory cytokines, microglia activation-related genes, and tight junction proteins was determined in the jejunum and the hippocampus, while microbiota composition and predicted function were verified in cecum contents. Voluntary exercise reduced anxiety-like behaviors and altered grooming patterns in males exclusively. Although the exercise intervention resulted in changes to brain inflammatory activity and to cecal microbiota composition and inferred function in both sexes, reductions in the jejunal expression of pro-inflammatory markers were observed in females only. These findings support the view that voluntary exercise, even when performed during a short period, is beneficial for mental and intestinal health and that its sex-specific effects on behavior could be, at least in part, related to some components of the gut microbiota-immune-brain axis.
Collapse
Affiliation(s)
| | - Joanna Kasia Szyszkowicz
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, Canada
| | - Natasha Osborne
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | | | - Christophe Nadon
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | | | - Ana Santos
- Department of Neuroscience, Carleton University, Ottawa, Canada
| | - Marie-Claude Audet
- Department of Neuroscience, Carleton University, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- School of Nutrition Sciences, University of Ottawa, Ottawa, Canada
| |
Collapse
|
32
|
Sequeira MK, Bolton JL. Stressed Microglia: Neuroendocrine-Neuroimmune Interactions in the Stress Response. Endocrinology 2023; 164:bqad088. [PMID: 37279575 PMCID: PMC11491833 DOI: 10.1210/endocr/bqad088] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/22/2023] [Accepted: 06/02/2023] [Indexed: 06/08/2023]
Abstract
Stressful life experiences are associated with the development of neuropsychiatric disorders like depression. Emerging evidence indicates that microglia, the specialized resident macrophages of the brain, may be a key mediator of the relationship between psychosocial stressor exposure and adaptive or maladaptive responses at the level of synaptic, circuit, and neuroimmune alterations. Here, we review current literature regarding how psychosocial stressor exposure changes microglial structure and function, thereby altering behavioral and brain outcomes, with a particular focus on age- and sex-dependent effects. We argue that additional emphasis should be placed in future research on investigating sex differences and the impacts of stressor exposure during sensitive periods of development, as well as going beyond traditional morphological measurements to interrogate microglial function. The bidirectional relationship between microglia and the stress response, particularly the role of microglia in the neuroendocrine control of stress-related circuits, is also an important area for future investigation. Finally, we discuss emerging themes and future directions that point to the possibility of the development of novel therapeutics for stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
| | - Jessica L Bolton
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
33
|
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
|
34
|
Maitra M, Mitsuhashi H, Rahimian R, Chawla A, Yang J, Fiori LM, Davoli MA, Perlman K, Aouabed Z, Mash DC, Suderman M, Mechawar N, Turecki G, Nagy C. Cell type specific transcriptomic differences in depression show similar patterns between males and females but implicate distinct cell types and genes. Nat Commun 2023; 14:2912. [PMID: 37217515 PMCID: PMC10203145 DOI: 10.1038/s41467-023-38530-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 05/05/2023] [Indexed: 05/24/2023] Open
Abstract
Major depressive disorder (MDD) is a common, heterogenous, and potentially serious psychiatric illness. Diverse brain cell types have been implicated in MDD etiology. Significant sexual differences exist in MDD clinical presentation and outcome, and recent evidence suggests different molecular bases for male and female MDD. We evaluated over 160,000 nuclei from 71 female and male donors, leveraging new and pre-existing single-nucleus RNA-sequencing data from the dorsolateral prefrontal cortex. Cell type specific transcriptome-wide threshold-free MDD-associated gene expression patterns were similar between the sexes, but significant differentially expressed genes (DEGs) diverged. Among 7 broad cell types and 41 clusters evaluated, microglia and parvalbumin interneurons contributed the most DEGs in females, while deep layer excitatory neurons, astrocytes, and oligodendrocyte precursors were the major contributors in males. Further, the Mic1 cluster with 38% of female DEGs and the ExN10_L46 cluster with 53% of male DEGs, stood out in the meta-analysis of both sexes.
Collapse
Affiliation(s)
- Malosree Maitra
- McGill Group for Suicide Studies, Douglas Institute, Verdun, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Haruka Mitsuhashi
- McGill Group for Suicide Studies, Douglas Institute, Verdun, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Reza Rahimian
- McGill Group for Suicide Studies, Douglas Institute, Verdun, QC, Canada
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Anjali Chawla
- McGill Group for Suicide Studies, Douglas Institute, Verdun, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Jennie Yang
- McGill Group for Suicide Studies, Douglas Institute, Verdun, QC, Canada
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Laura M Fiori
- McGill Group for Suicide Studies, Douglas Institute, Verdun, QC, Canada
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Maria Antonietta Davoli
- McGill Group for Suicide Studies, Douglas Institute, Verdun, QC, Canada
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Kelly Perlman
- McGill Group for Suicide Studies, Douglas Institute, Verdun, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Zahia Aouabed
- McGill Group for Suicide Studies, Douglas Institute, Verdun, QC, Canada
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Deborah C Mash
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Matthew Suderman
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Institute, Verdun, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Institute, Verdun, QC, Canada.
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada.
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Institute, Verdun, QC, Canada.
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
35
|
Gilbert EAB, Livingston J, Garcia-Flores E, Kehtari T, Morshead CM. Metformin Improves Functional Outcomes, Activates Neural Precursor Cells, and Modulates Microglia in a Sex-Dependent Manner After Spinal Cord Injury. Stem Cells Transl Med 2023:7174953. [PMID: 37209417 DOI: 10.1093/stcltm/szad030] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 04/20/2023] [Indexed: 05/22/2023] Open
Abstract
Spinal cord injury (SCI) results in devastating patient outcomes with few treatment options. A promising approach to improve outcomes following SCI involves the activation of endogenous precursor populations including neural stem and progenitor cells (NSPCs) which are located in the periventricular zone (PVZ), and oligodendrocyte precursor cells (OPCs) found throughout the parenchyma. In the adult spinal cord, resident NSPCs are primarily mitotically quiescent and aneurogenic, while OPCs contribute to ongoing oligodendrogenesis into adulthood. Each of these populations is responsive to SCI, increasing their proliferation and migration to the site of injury; however, their activation is not sufficient to support functional recovery. Previous work has shown that administration of the FDA-approved drug metformin is effective at promoting endogenous brain repair following injury, and this is correlated with enhanced NSPC activation. Here, we ask whether metformin can promote functional recovery and neural repair following SCI in both males and females. Our results reveal that acute, but not delayed metformin administration improves functional outcomes following SCI in both sexes. The functional improvement is concomitant with OPC activation and oligodendrogenesis. Our data also reveal sex-dependent effects of metformin following SCI with increased activation of NSPCs in females and reduced microglia activation in males. Taken together, these findings support metformin as a viable therapeutic strategy following SCI and highlight its pleiotropic effects in the spinal cord.
Collapse
Affiliation(s)
- Emily A B Gilbert
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Jessica Livingston
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Emilio Garcia-Flores
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Tarlan Kehtari
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Cindi M Morshead
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| |
Collapse
|
36
|
Zhu W, Zhang W, Yang F, Cai M, Li X, Xiang Y, Xiang J, Yang Y, Cai D. Role of PGC-1α mediated synaptic plasticity, mitochondrial function, and neuroinflammation in the antidepressant effect of Zi-Shui-Qing-Gan-Yin. Front Neurol 2023; 14:1108494. [PMID: 37251232 PMCID: PMC10213669 DOI: 10.3389/fneur.2023.1108494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 03/15/2023] [Indexed: 05/31/2023] Open
Abstract
Depression is the most prevalent psychiatric disorder, which needs deeper mechanism research studies and effective therapy. Zi-Shui-Qing-Gan-Yin (ZSQGY) is a traditional Chinese medicine decoction that has been widely used in China in the treatment of depressive symptoms. The aim of the study was to examine the anti-depressive effects of ZSQGY and the possible mechanism of action in the monosodium glutamate (MSG)-induced depressive model and the corticosterone (CORT)-induced PC12 cell model. Liquid chromatography-mass spectrometry (LC-MS) was performed to determine the major compounds in the water extract of ZSQGY. The depressive behaviors were evaluated by the field swimming test (FST), the sucrose preference test (SPT), and the open field test (OFT). Golgi staining and transmission electron microscopy (TEM) were performed to display the alterations of synaptic ultrastructure. The mitochondrion function and inflammatory factors were also quantified. The changes in peroxisome proliferator-activated receptor-γ co-activator 1α (PGC-1α) expression were evaluated. The results of this study demonstrated that ZSQGY significantly improved depressive behaviors. ZSQGY also reversed the changes in synaptic plasticity, improved mitochondrion function, and reduced the levels of inflammatory factors. The neuroprotective effects were accompanied by the increased expression of PGC-1α. However, the beneficial changes were reversed after the inhibition of PGC-1α. These results indicated that ZSQGY effectively could improve depressive behaviors via the mechanisms that regulate synaptic structural plasticity, improve mitochondrion function, and alleviate neuroinflammation, which could, or partly, attribute to the regulation of PGC-1α.
Collapse
Affiliation(s)
- Wen Zhu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Wen Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Feng Yang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Min Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Xiangting Li
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yijin Xiang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Jun Xiang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yunke Yang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Dingfang Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Tariq MB, Lee J, McCullough LD. Sex differences in the inflammatory response to stroke. Semin Immunopathol 2023; 45:295-313. [PMID: 36355204 PMCID: PMC10924671 DOI: 10.1007/s00281-022-00969-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: 07/29/2022] [Accepted: 10/18/2022] [Indexed: 11/11/2022]
Abstract
Ischemic stroke is a leading cause of morbidity and mortality and disproportionally affects women, in part due to their higher longevity. Older women have poorer outcomes after stroke with high rates of cognitive deficits, depression, and reduced quality of life. Post-stroke inflammatory responses are also sexually dimorphic and drive differences in infarct size and recovery. Factors that influence sex-specific immune responses can be both intrinsic and extrinsic. Differences in gonadal hormone exposure, sex chromosome compliment, and environmental/social factors can drive changes in transcriptional and metabolic profiles. In addition, how these variables interact, changes across the lifespan. After the onset of ischemic injury, necrosis and apoptosis occur, which activate microglia and other glial cells within the central nervous system, promoting the release of cytokines and chemokines and neuroinflammation. Cells involved in innate and adaptive immune responses also have dual functions after stroke as they can enhance inflammation acutely, but also contribute to suppression of the inflammatory cascade and later repair. In this review, we provide an overview of the current literature on sex-specific inflammatory responses to ischemic stroke. Understanding these differences is critical to identifying therapeutic options for both men and women.
Collapse
Affiliation(s)
- Muhammad Bilal Tariq
- Memorial Hermann Hospital-Texas Medical Center, Houston, TX, 77030, USA
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St, MSB7044B, Houston, TX, 77030, USA
| | - Juneyoung Lee
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St, MSB7044B, Houston, TX, 77030, USA
| | - Louise D McCullough
- Memorial Hermann Hospital-Texas Medical Center, Houston, TX, 77030, USA.
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St, MSB7044B, Houston, TX, 77030, USA.
| |
Collapse
|
38
|
Mawson ER, Morris BJ. A consideration of the increased risk of schizophrenia due to prenatal maternal stress, and the possible role of microglia. Prog Neuropsychopharmacol Biol Psychiatry 2023; 125:110773. [PMID: 37116354 DOI: 10.1016/j.pnpbp.2023.110773] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/07/2023] [Accepted: 04/18/2023] [Indexed: 04/30/2023]
Abstract
Schizophrenia is caused by interaction of a combination of genetic and environmental factors. Of the latter, prenatal exposure to maternal stress is reportedly associated with elevated disease risk. The main orchestrators of inflammatory processes within the brain are microglia, and aberrant microglial activation/function has been proposed to contribute to the aetiology of schizophrenia. Here, we evaluate the epidemiological and preclinical evidence connecting prenatal stress to schizophrenia risk, and consider the possible mediating role of microglia in the prenatal stress-schizophrenia relationship. Epidemiological findings are rather consistent in supporting the association, albeit they are mitigated by effects of sex and gestational timing, while the evidence for microglial activation is more variable. Rodent models of prenatal stress generally report lasting effects on offspring neurobiology. However, many uncertainties remain as to the mechanisms underlying the influence of maternal stress on the developing foetal brain. Future studies should aim to characterise the exact processes mediating this aspect of schizophrenia risk, as well as focussing on how prenatal stress may interact with other risk factors.
Collapse
Affiliation(s)
- Eleanor R Mawson
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Brian J Morris
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
39
|
Doyle MA, Brown JA, Winder DG. GluN2D expression is regulated by restraint stress and supports active stress coping bouts. Neuropharmacology 2023; 225:109377. [PMID: 36528117 PMCID: PMC9839594 DOI: 10.1016/j.neuropharm.2022.109377] [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: 09/27/2022] [Revised: 11/22/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022]
Abstract
Stress coping strategies represent critical responses to environmental challenges, and active coping has been linked to stress resilience in humans. Understanding the neuroadaptations that support these strategies may provide insights into adaptive and maladaptive stress responses. NMDA receptors (NMDARs) play key roles in neuroadaptation, and NMDARs have been specifically implicated in stress responsiveness. Constitutive knockout mice have been used to implicate the GluN2D NMDAR subunit in regulation of stress-sensitive and affective behavior, but the brain regions in which GluN2D expression changes drive these effects remain unknown. Here we report that following an acute restraint stressor, GluN2D subunit expression is specifically decreased in the bed nucleus of the stria terminalis (BNST), a key region involved in stress processing, in male but not female mice, with no differences found in the thalamus or ventral hippocampus in either sex. Rodents engage in active struggling events during restraint stress that may represent active coping strategies to stress. Thus, we assessed active coping bouts during acute and chronic restraint stress sessions in GluN2D knockout mice. During the first restraint session, GluN2D knockout mice exhibited a pronounced decrease in struggling bouts during restraint stress relative to wild-type littermates, consistent with a role of GluN2D in active coping responses to stress. Repeated, daily restraint sessions revealed a sex-specific role of GluN2D expression on certain aspects of active coping behaviors, with male GluN2D KO mice exhibiting a decrease in total coping bouts measured across five sessions. However, BNST-specific knockdown of GluN2D in male mice did not alter active coping bouts, suggesting either a multi-synaptic role of GluN2D and/or a developmental role of GluN2D in this behavior. Altogether, these data are consistent with a growing literature suggesting that exploration of GluN2D control of stress circuit actions may lead to a novel therapeutic target to consider for stress-related mood disorders.
Collapse
Affiliation(s)
- Marie A Doyle
- Department of Molecular Physiology and Biophysics, Vanderbilt University, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, USA
| | - Jordan A Brown
- Vanderbilt Center for Addiction Research, Vanderbilt University, USA; Department of Pharmacology, Vanderbilt University, USA
| | - Danny G Winder
- Department of Molecular Physiology and Biophysics, Vanderbilt University, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, USA; Department of Pharmacology, Vanderbilt University, USA; Department of Psychiatry, Vanderbilt University Medical Center, USA.
| |
Collapse
|
40
|
Sanford LD, Wellman LL, Adkins AM, Guo ML, Zhang Y, Ren R, Yang L, Tang X. Modeling integrated stress, sleep, fear and neuroimmune responses: Relevance for understanding trauma and stress-related disorders. Neurobiol Stress 2023; 23:100517. [PMID: 36793998 PMCID: PMC9923229 DOI: 10.1016/j.ynstr.2023.100517] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 12/30/2022] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Sleep and stress have complex interactions that are implicated in both physical diseases and psychiatric disorders. These interactions can be modulated by learning and memory, and involve additional interactions with the neuroimmune system. In this paper, we propose that stressful challenges induce integrated responses across multiple systems that can vary depending on situational variables in which the initial stress was experienced, and with the ability of the individual to cope with stress- and fear-inducing challenges. Differences in coping may involve differences in resilience and vulnerability and/or whether the stressful context allows adaptive learning and responses. We provide data demonstrating both common (corticosterone, SIH and fear behaviors) and distinguishing (sleep and neuroimmune) responses that are associated with an individual's ability to respond and relative resilience and vulnerability. We discuss neurocircuitry regulating integrated stress, sleep, neuroimmune and fear responses, and show that responses can be modulated at the neural level. Finally, we discuss factors that need to be considered in models of integrated stress responses and their relevance for understanding stress-related disorders in humans.
Collapse
Affiliation(s)
- Larry D. Sanford
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Laurie L. Wellman
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Austin M. Adkins
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Ming-Lei Guo
- Drug Addiction Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Ye Zhang
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, Translational Neuroscience Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Rong Ren
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, Translational Neuroscience Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Linghui Yang
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, Translational Neuroscience Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangdong Tang
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, Translational Neuroscience Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
41
|
Multiparity Differentially Affects Specific Aspects of the Acute Neuroinflammatory Response to Traumatic Brain Injury in Female Mice. Neuroscience 2023; 511:86-99. [PMID: 36535576 DOI: 10.1016/j.neuroscience.2022.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 12/05/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
Pregnancy is associated with profound acute and long-term physiological changes, but the effects of such changes on brain injury outcomes are unclear. Here, we examined the effects of previous pregnancy and maternal experience (parity) on acute neuroinflammatory responses to lateral fluid percussion injury (FPI), a well-defined experimental traumatic brain injury (TBI) paradigm. Multiparous (2-3 pregnancies and motherhood experiences) and age-matched nulliparous (no previous pregnancy or motherhood experience) female mice received either FPI or sham injury and were euthanized 3 days post-injury (DPI). Increased cortical Iba1, GFAP, and CD68 immunolabeling was observed following TBI independent of parity and microglia morphology did not differ between TBI groups. However, multiparous females had fewer CD45+ cells near the site of injury compared to nulliparous females, which was associated with preserved aquaporin-4 polarization, suggesting that parity may influence leukocyte recruitment to the site of injury and maintenance of blood brain barrier permeability following TBI. Additionally, relative cortical Il6 gene expression following TBI was dependent on parity such that TBI increased Il6 expression in nulliparous, but not multiparous, mice. Together, this work suggests that reproductive history may influence acute neuroinflammatory outcomes following TBI in females.
Collapse
|
42
|
Solarz A, Majcher-Maślanka I, Kryst J, Chocyk A. Early-life stress affects peripheral, blood-brain barrier, and brain responses to immune challenge in juvenile and adult rats. Brain Behav Immun 2023; 108:1-15. [PMID: 36400335 DOI: 10.1016/j.bbi.2022.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/21/2022] [Accepted: 11/12/2022] [Indexed: 11/17/2022] Open
Abstract
Early-life stress (ELS) may affect brain maturation and neuroimmune interactions and, consequently, the inflammatory response to subsequent environmental factors later in life. Recently, the coexistence of blood-brain barrier (BBB) dysfunction and inflammation has been implicated in the etiology and progression of mental and/or neurodegenerative diseases. There are sex differences in the prevalence and outcomes of these disorders. The number of studies reporting the effects of ELS and sex on BBB functioning and neuroinflammatory processes in response to immune challenge is very limited, and the data are inconsistent. In the present study, we examined whether ELS, based on the maternal separation (MS) paradigm in rats, can condition male and female subjects to subsequent lipopolysaccharide (LPS)-induced immune challenge in juvenility or adulthood. Twenty-four hours after acute LPS injection, serum proinflammatory cytokines were measured, and BBB permeability in the medial prefrontal cortex (mPFC) and hippocampus (HP) was evaluated. Additionally, the mRNA expression of neuroinflammatory markers and BBB-related genes was also studied. We found that a single LPS challenge induced a proinflammatory response both in the periphery and in the mPFC and HP and increased BBB permeability in a sex-dependent fashion. Moreover, MS enhanced the neuroinflammatory response to LPS challenge in males (especially juveniles), whereas MS females showed no difference or a blunted central response to LPS compared with control females, mainly during adulthood. These results suggest that ELS may precondition individuals to subsequent environmental factors later in life in a sex-specific manner and potentially determine their susceptibility or resilience to mental and/or neurodegenerative diseases.
Collapse
Affiliation(s)
- Anna Solarz
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Smętna Street 12, Poland
| | - Iwona Majcher-Maślanka
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Smętna Street 12, Poland
| | - Joanna Kryst
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Smętna Street 12, Poland; Department of Chemistry and Biochemistry, Institute for Basics Sciences, Faculty of Physiotherapy, University of Physical Education, Jana Pawła II Av. 78, 31-571 Kraków, Poland
| | - Agnieszka Chocyk
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Smętna Street 12, Poland.
| |
Collapse
|
43
|
Abstract
Depression and anxiety disorders carry a tremendous worldwide burden and emerge as a significant cause of disability among western societies. Both disorders are known to disproportionally affect women, as they are twice more likely to be diagnosed and moreover, they are also prone to suffer from female-specific mood disorders. Importantly, the prevalence of these affective disorders has notably risen after the COVID pandemic, especially in women. In this chapter, we describe factors that are possibly contributing to the expression of such sex differences in depression and anxiety. For this, we overview the effect of transcriptomic and genetic factors, the immune system, neuroendocrine aspects, and cognition. Furthermore, we also provide evidence of sex differences in antidepressant response and their causes. Finally, we emphasize the importance to consider sex as a biological variable in preclinical and clinical research, which may facilitate the discovery and development of new and more efficacious antidepressant and anxiolytic pharmacotherapies for both women and men.
Collapse
Affiliation(s)
- Pavlina Pavlidi
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
44
|
De Felice E, Gonçalves de Andrade E, Golia MT, González Ibáñez F, Khakpour M, Di Castro MA, Garofalo S, Di Pietro E, Benatti C, Brunello N, Tascedda F, Kaminska B, Limatola C, Ragozzino D, Tremblay ME, Alboni S, Maggi L. Microglial diversity along the hippocampal longitudinal axis impacts synaptic plasticity in adult male mice under homeostatic conditions. J Neuroinflammation 2022; 19:292. [PMID: 36482444 PMCID: PMC9730634 DOI: 10.1186/s12974-022-02655-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
The hippocampus is a plastic brain area that shows functional segregation along its longitudinal axis, reflected by a higher level of long-term potentiation (LTP) in the CA1 region of the dorsal hippocampus (DH) compared to the ventral hippocampus (VH), but the mechanisms underlying this difference remain elusive. Numerous studies have highlighted the importance of microglia-neuronal communication in modulating synaptic transmission and hippocampal plasticity, although its role in physiological contexts is still largely unknown. We characterized in depth the features of microglia in the two hippocampal poles and investigated their contribution to CA1 plasticity under physiological conditions. We unveiled the influence of microglia in differentially modulating the amplitude of LTP in the DH and VH, showing that minocycline or PLX5622 treatment reduced LTP amplitude in the DH, while increasing it in the VH. This was recapitulated in Cx3cr1 knockout mice, indicating that microglia have a key role in setting the conditions for plasticity processes in a region-specific manner, and that the CX3CL1-CX3CR1 pathway is a key element in determining the basal level of CA1 LTP in the two regions. The observed LTP differences at the two poles were associated with transcriptional changes in the expression of genes encoding for Il-1, Tnf-α, Il-6, and Bdnf, essential players of neuronal plasticity. Furthermore, microglia in the CA1 SR region showed an increase in soma and a more extensive arborization, an increased prevalence of immature lysosomes accompanied by an elevation in mRNA expression of phagocytic markers Mertk and Cd68 and a surge in the expression of microglial outward K+ currents in the VH compared to DH, suggesting a distinct basal phenotypic state of microglia across the two hippocampal poles. Overall, we characterized the molecular, morphological, ultrastructural, and functional profile of microglia at the two poles, suggesting that modifications in hippocampal subregions related to different microglial statuses can contribute to dissect the phenotypical aspects of many diseases in which microglia are known to be involved.
Collapse
Affiliation(s)
- E. De Felice
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - E. Gonçalves de Andrade
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada
| | - M. T. Golia
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - F. González Ibáñez
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada ,grid.411081.d0000 0000 9471 1794Faculté de Médecine and Centre de Recherche, CHU de Québec-Université Laval, Quebec, Canada
| | - M. Khakpour
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada
| | - M. A. Di Castro
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - S. Garofalo
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - E. Di Pietro
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - C. Benatti
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - N. Brunello
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - F. Tascedda
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - B. Kaminska
- grid.419305.a0000 0001 1943 2944Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - C. Limatola
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy ,grid.7841.aDepartment of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur, Sapienza University, Rome, Italy
| | - D. Ragozzino
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy ,grid.417778.a0000 0001 0692 3437Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
| | - M. E. Tremblay
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada ,grid.411081.d0000 0000 9471 1794Faculté de Médecine and Centre de Recherche, CHU de Québec-Université Laval, Quebec, Canada
| | - S. Alboni
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - L. Maggi
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| |
Collapse
|
45
|
Mishra I, Pullum KB, Eads KN, Strunjas AR, Ashley NT. Peripheral Sympathectomy Alters Neuroinflammatory and Microglial Responses to Sleep Fragmentation in Female Mice. Neuroscience 2022; 505:111-124. [PMID: 36240943 PMCID: PMC9671838 DOI: 10.1016/j.neuroscience.2022.09.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/05/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022]
Abstract
Sleep loss, either induced by obstructive sleep apnea or other forms of sleep dysfunction, induces an inflammatory response, as commonly measured by increased circulating levels of pro-inflammatory cytokines. Increased catecholamines from sympathetic nervous system (SNS) activation regulates this peripheral inflammation. However, the role that catecholamines play in mediating neuroinflammation from sleep perturbations is undescribed. The aims of this study were to determine (i) the effect of peripheral SNS inhibition upon neuroinflammatory responses to sleep fragmentation (SF) and (ii) whether homeostasis can be restored after 1 week of recovery sleep. We measured gene expression levels of pro- and anti-inflammatory cytokines and microglial activity in brain (prefrontal cortex, hippocampus and hypothalamus) of female mice that were subjected to acute SF for 24 hours, chronic SF for 8 weeks, or 7 days of recovery after chronic SF. In each experiment, SF and control mice were peripherally sympathectomized with 6-hydroxydopamine (6-OHDA) or injected with vehicle. SF elevated cytokine mRNA expression in brain and increased microglial density and cell area in some regions. In addition, chronic SF promoted hyper-ramification in resting microglia upon exposure to chronic, but not acute, SF. Effects of chronic SF were more pronounced than acute SF, and 1 week of recovery was not sufficient to alleviate neuroinflammation. Importantly, 6-OHDA treatment significantly alleviated SF-induced inflammation and microglial responses. This study provides evidence of SNS regulation of neural inflammation from SF, suggesting a potential role for therapeutics that could mitigate neuroinflammatory responses to sleep dysfunction.
Collapse
Affiliation(s)
- Ila Mishra
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA; Harrington Discovery Institute, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Keelee B Pullum
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA; Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kristen N Eads
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA
| | - Anna R Strunjas
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA
| | - Noah T Ashley
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA.
| |
Collapse
|
46
|
Medina-Rodriguez EM, Rice KC, Jope RS, Beurel E. Comparison of inflammatory and behavioral responses to chronic stress in female and male mice. Brain Behav Immun 2022; 106:180-197. [PMID: 36058417 PMCID: PMC9561002 DOI: 10.1016/j.bbi.2022.08.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Major depressive disorder (MDD) is a debilitating disease with a high worldwide prevalence. Despite its greater prevalence in women, male animals are used in most preclinical studies of depression even though there are many sex differences in key components of depression, such as stress responses and immune system functions. In the present study, we found that chronic restraint stress-induced depressive-like behaviors are quite similar in male and female mice, with both sexes displaying increased immobility time in the tail suspension test and reduced social interactions, and both sexes exhibited deficits in working and spatial memories. However, in contrast to the similar depressive-like behaviors developed by male and female mice in response to stress, they displayed different patterns of pro-inflammatory cytokine increases in the periphery and the brain, different changes in microglia, and different changes in the expression of Toll-like receptor 4 in response to stress. Treatment with (+)-naloxone, a Toll-like receptor 4 antagonist that previously demonstrated anti-depressant-like effects in male mice, was more efficacious in male than female mice in reducing the deleterious effects of stress, and its effects were not microbiome-mediated. Altogether, these results suggest differential mechanisms to consider in potential sex-specific treatments of depression.
Collapse
Affiliation(s)
- Eva M Medina-Rodriguez
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL 33125, United States
| | - Kenner C Rice
- Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Richard S Jope
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL 33125, United States; Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Eléonore Beurel
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States.
| |
Collapse
|
47
|
Smiley CE, Wood SK. Stress- and drug-induced neuroimmune signaling as a therapeutic target for comorbid anxiety and substance use disorders. Pharmacol Ther 2022; 239:108212. [PMID: 35580690 DOI: 10.1016/j.pharmthera.2022.108212] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
Abstract
Stress and substance use disorders remain two of the most highly prevalent psychiatric conditions and are often comorbid. While individually these conditions have a debilitating impact on the patient and a high cost to society, the symptomology and treatment outcomes are further exacerbated when they occur together. As such, there are few effective treatment options for these patients, and recent investigation has sought to determine the neural processes underlying the co-occurrence of these disorders to identify novel treatment targets. One such mechanism that has been linked to stress- and addiction-related conditions is neuroimmune signaling. Increases in inflammatory factors across the brain have been heavily implicated in the etiology of these disorders, and this review seeks to determine the nature of this relationship. According to the "dual-hit" hypothesis, also referred to as neuroimmune priming, prior exposure to either stress or drugs of abuse can sensitize the neuroimmune system to be hyperresponsive when exposed to these insults in the future. This review completes an examination of the literature surrounding stress-induced increases in inflammation across clinical and preclinical studies along with a summarization of the evidence regarding drug-induced alterations in inflammatory factors. These changes in neuroimmune profiles are also discussed within the context of their impact on the neural circuitry responsible for stress responsiveness and addictive behaviors. Further, this review explores the connection between neuroimmune signaling and susceptibility to these conditions and highlights the anti-inflammatory pharmacotherapies that may be used for the treatment of stress and substance use disorders.
Collapse
Affiliation(s)
- Cora E Smiley
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| | - Susan K Wood
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| |
Collapse
|
48
|
Liu J, Sato Y, Falcone-Juengert J, Kurisu K, Shi J, Yenari MA. Sexual dimorphism in immune cell responses following stroke. Neurobiol Dis 2022; 172:105836. [PMID: 35932990 DOI: 10.1016/j.nbd.2022.105836] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 07/11/2022] [Accepted: 07/31/2022] [Indexed: 11/22/2022] Open
Abstract
Recent bodies of work in regard to stroke have revealed significant sex differences in terms of risk and outcome. While differences in sex hormones have been the focus of earlier research, the reasons for these differences are much more complex and require further identification. This review covers differences in sex related immune responses with a focus on differences in immune cell composition and function. While females are more susceptible to immune related diseases, they seem to have better outcomes from stroke at the experimental level with reduced pro-inflammatory responses. However, at the clinical level, the picture is much more complex with worse neurological outcomes from stroke. While the use of exogenous sex steroids can replicate some of these findings, it is apparent that many other factors are involved in the modulation of immune responses. As a result, more research is needed to better understand these differences and identify appropriate interventions and risk modification.
Collapse
Affiliation(s)
- Jialing Liu
- Dept Neurosurgery, UCSF and SF VAMC, San Francisco, CA, USA
| | - Yoshimichi Sato
- Dept Neurosurgery, UCSF and SF VAMC, San Francisco, CA, USA; Dept Neurosurgery, Tohoku University, Sendai, Japan
| | | | - Kota Kurisu
- Dept Neurosurgery, Hokkaido University, Sapporo, Japan
| | - Jian Shi
- Dept Neurology, UCSF and SF VAMC, San Francisco, CA, USA
| | | |
Collapse
|
49
|
Wang Y, Hu Z, Liu H, Gu Y, Ye M, Lu Q, Lu X, Huang C. Adolescent microglia stimulation produces long-lasting protection against chronic stress-induced behavioral abnormalities in adult male mice. Brain Behav Immun 2022; 105:44-66. [PMID: 35781008 DOI: 10.1016/j.bbi.2022.06.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/05/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022] Open
Abstract
Our previous studies had reported that microglia activation one day before stress exposure prevented the behavioral abnormalities induced by chronic stress in adult mice, and a 10-day interval between microglia stimulation and stress exposure can abolish the prophylactic effect of LPS preinjection on the behavioral abnormalities induced by chronic stress, which, however, could be rescued by repeated LPS injection. This suggests that increased stimulation of microglia results in animals developing a strong ability to prevent deleterious stress stimuli. Because microglia in the adolescent brain exhibit flexible immunological plasticity, we hypothesize that a single low-dose LPS injection during adolescence may provide long-lasting protection against behavioral abnormalities induced by chronic stress in adult mice. As expected, our results showed that a single injection of LPS (100 μg/kg) at post-natal day 28 (PND 28) prevented the development of abnormal behaviors and shifted neuroinflammatory responses toward an anti-inflammatory phenotype in adult mice treated with CSDS at their different stages of the age (PND 56, 140, and 252). Moreover, pretreatment with minocycline or PLX3397 to inhibit microglial activation abolished the prophylactic effect of LPS preinjection after PND 28 on behavioral abnormalities and neuroinflammatory responses induced by CSDS in adult mice at their different stages of the age, PND 56, 140, and 252. These results indicate that stimulation of microglia in adolescence may confer long-lasting protection against neuroinflammatory responses and behavioral abnormalities induced by chronic stress in adult mice. This may offer the potential for the development of a "vaccine-like strategy" to prevent mental disorders.
Collapse
Affiliation(s)
- Yue Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Zhichao Hu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Huijun Liu
- Department of Pharmacy, Yancheng First Hospital, the Fourth Affiliated Hospital of Nantong University, #66 Renmin South Road, Yancheng 224006, Jiangsu, China
| | - Yue Gu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Minxiu Ye
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Qun Lu
- Department of Pharmacy, Nantong Third Hospital Affiliated to Nantong University, #60 Middle Qingnian Road, Nantong 226006, Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
50
|
Gabrielli M, Raffaele S, Fumagalli M, Verderio C. The multiple faces of extracellular vesicles released by microglia: Where are we 10 years after? Front Cell Neurosci 2022; 16:984690. [PMID: 36176630 PMCID: PMC9514840 DOI: 10.3389/fncel.2022.984690] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/23/2022] [Indexed: 11/30/2022] Open
Abstract
As resident component of the innate immunity in the central nervous system (CNS), microglia are key players in pathology. However, they also exert fundamental roles in brain development and homeostasis maintenance. They are extremely sensitive and plastic, as they assiduously monitor the environment, adapting their function in response to stimuli. On consequence, microglia may be defined a heterogeneous community of cells in a dynamic equilibrium. Extracellular vesicles (EVs) released by microglia mirror the dynamic nature of their donor cells, exerting important and versatile functions in the CNS as unbounded conveyors of bioactive signals. In this review, we summarize the current knowledge on EVs released by microglia, highlighting their heterogeneous properties and multifaceted effects.
Collapse
Affiliation(s)
- Martina Gabrielli
- CNR Institute of Neuroscience, Vedano al Lambro, Italy
- *Correspondence: Martina Gabrielli,
| | - Stefano Raffaele
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Claudia Verderio
- CNR Institute of Neuroscience, Vedano al Lambro, Italy
- Claudia Verderio,
| |
Collapse
|