1
|
Zhao X, Li D, Zhu Z, Li S, Qin Y, Yang Y. Spermidine attenuates microglial activation, neuroinflammation, and neuronal injury in rat model of vascular dementia. Neuroscience 2025; 573:355-363. [PMID: 40169099 DOI: 10.1016/j.neuroscience.2025.03.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 03/15/2025] [Accepted: 03/23/2025] [Indexed: 04/03/2025]
Abstract
Spermidine has been implicated to provide beneficial effects on cognitive function in several model organisms as well as older adults with mild and moderate dementia. Nevertheless, the potential impact of spermidine on learning and memory deficits in vascular dementia (VaD) remains largely unknown. Here, bilateral common carotid artery occlusion (BCCAo) was applied to induce chronic cerebral hypoperfusion in rats. We demonstrated that spermidine therapy improved the spatial learning performance in model animals, accompanied with decreased cerebral histopathologic injury and increased restored myelin basic protein (MBP) expression. Moreover, spermidine suppressed abnormal microglia activation, inhibited the excessive generation of proinflammatory mediators, such as tumor necrosis factor (TNF)α and inducible nitric oxide synthase (iNOS), and increased the anti-inflammatory cytokine transforming growth factor (TGF)β expression in rodent brain following hypoperfusion. Our findings indicated that spermidine alleviated cognitive impairments of rats after VaD-like injury possibly via suppressing microglia-modulated neuroinflammation and neuronal injury. These data may shed light on understanding the pathogenesis of VaD and point to the promising value of spermidine supplementation for cognition improvement.
Collapse
Affiliation(s)
- Xinyuan Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China
| | - Dan Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China
| | - Zirui Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China
| | - Suzhen Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China
| | - Yaze Qin
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China
| | - Yi Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China.
| |
Collapse
|
2
|
Shu Y, Zhou W, Zhang W, Lu L, Gao Y, Yu Y, Shan C, Tong D, Zhang X, Shi W, Liu G. Exposure to malathion impairs learning and memory of zebrafish by disrupting cholinergic signal transmission, undermining synaptic plasticity, and aggravating neuronal apoptosis. JOURNAL OF HAZARDOUS MATERIALS 2025; 488:137391. [PMID: 39892146 DOI: 10.1016/j.jhazmat.2025.137391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/09/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
The prevalence of organophosphorus pesticides, such as malathion, in water environments poses a severe threat to aquatic organisms. Although the brain is a potential target for malathion, little is known about its effect on cognitive functions in fish. In this study, we evaluated the effect of 4-week malathion exposure on the learning and memory of zebrafish using T-maze tasks. In addition to verifying the accumulation of malathion in the brain and its deleterious effects on blood-brain barrier integrity, the impacts of malathion on cholinergic signal transmission, synaptic plasticity, apoptosis, and oxidative stress were determined. Our results demonstrated that a 4-week malathion exposure resulted in typical learning and memory-deficit-like behaviors. Apart from inhibiting cholinergic signal transmission, synaptic plasticity was severely undermined by malathion (as evidenced by the disruption of BDNF/PI3K/AKT/CREB pathway, suppression of synaptophysins, and activation of microglia). Moreover, significantly higher levels of TUNEL fluorescence signals as well as apoptotic enzymes and genes probably induced by oxidative stress were detected in the brains of malathion-exposed zebrafish. Collectively, our results suggested that malathion at environmentally realistic levels can significantly undermine learning and memory of zebrafish by disrupting cholinergic signal transmission, impairing synaptic plasticity, and aggravating neuronal apoptosis via inducing oxidative stress.
Collapse
Affiliation(s)
- Yang Shu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weishang Zhou
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weixia Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lingzheng Lu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yan Gao
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yingying Yu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Conghui Shan
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Difei Tong
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xunyi Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wei Shi
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Guangxu Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
3
|
Li W, Zhang L, Xu Y, Li H, Li B, Sun S, Zhang X, Duan G, Chen Y, Zhang J, Cao Y, Li X, Liu Q, Wu Y, Zhang S, Leavenworth JW, Wang X, Zhu C. Altered monocyte subpopulations and their association with autism spectrum disorder risk in children. Brain Behav Immun 2025; 126:315-326. [PMID: 40010548 DOI: 10.1016/j.bbi.2025.02.028] [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: 07/12/2024] [Revised: 02/06/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025] Open
Abstract
OBJECTIVE Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficits in social communication, restricted interests, and repetitive behaviors. Emerging evidence suggests a link between immune dysregulation and ASD. This study investigates alterations in monocyte subpopulations and cytokine production in children with ASD and their potential associations with ASD risk and severity. METHODS Initially, the immune status of peripheral blood mononuclear cells was assessed in cohort-I of 96 typically developing (TD) children and 92 children diagnosed with ASD using flow cytometry. Subsequently, the secretion of cytokines IL-6 and IL-10 by monocytes was evaluated following stimulation with a leukocyte activation mixture and intracellular protein staining technique in cohort-II. RESULTS Children with ASD exhibited significantly higher levels of total monocytes, classical monocytes (CD14hi/CD16-), and non-classical monocytes (CD14low/CD16+) compared to TD children (p < 0.001). Elevated levels of classical monocytes (β: 0.395; 95 %CI: 0.260-0.530; p < 0.001) and non-classical monocytes (β: 0.629; 95 %CI: 0.516-0.742; p < 0.001) were significantly associated with ASD after adjusting for age, sex and body mass index. Furthermore, increased production of IL-6 by monocytes was observed in children with ASD (p = 0.001). Logistic regression analysis revealed that classical monocytes (OR: 1.104; 95 %CI: 1.062-1.147; p < 0.001), non-classical monocytes (OR: 2.913; 95 %CI: 2.130-3.986; p < 0.001) and IL-6 production by monocytes (OR: 1.306; 95 %CI: 1.096-1.557; p = 0.003) are risk factors for ASD. Spearman correlation analysis revealed a negative correlation between classical monocyte levels and adaptive behavior developmental quotient (DQ) (r = - 0.377; p = 0.001), fine motor DQ (r = - 0.329; p = 0.003) and personal-social DQ (r = - 0.247; p = 0.029) in children with ASD. CONCLUSION Elevated classical and non-classical monocytes are potential risk factors for ASD and may influence neurodevelopmental outcomes. Further research is needed to elucidate the precise mechanisms and therapeutic implications.
Collapse
Affiliation(s)
- Wenhua Li
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg 40530, Sweden
| | - Lingling Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hongwei Li
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Bingbing Li
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Shuang Sun
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xiaoli Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Guiqin Duan
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yiwen Chen
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jie Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yangyang Cao
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xiaoping Li
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qianqian Liu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yanan Wu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Shan Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg 40530, Sweden
| | - Jianmei W Leavenworth
- Department of Neurosurgery and Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham AL 35233, USA
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Centre of Perinatal Medicine and Health, Institute of Clinical Science, University of Gothenburg 40530 Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg 40530, Sweden; Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
4
|
Ju J, Pan Y, Yang X, Li X, Chen J, Wu S, Hou ST. The "don't eat me" signal CD47 is associated with microglial phagocytosis defects and autism-like behaviors in 16p11.2 deletion mice. Proc Natl Acad Sci U S A 2025; 122:e2411080122. [PMID: 40238451 DOI: 10.1073/pnas.2411080122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 03/13/2025] [Indexed: 04/18/2025] Open
Abstract
Various pathological characteristics of autism spectrum disorder (ASD) stem from abnormalities in brain resident immune cells, specifically microglia, to prune unnecessary synapses or neural connections during early development. Animal models of ASD exhibit an abundance of synapses in different brain regions, which is strongly linked to the appearance of ASD behaviors. Overexpression of CD47 on neurons acts as a "don't eat me" signal, safeguarding synapses from inappropriate pruning by microglia. Indeed, CD47 overexpression occurs in 16p11.2 deletion carriers, causing decreased synaptic phagocytosis and the manifestation of ASD characteristics. However, the role of CD47 in synaptic pruning impairment leading to ASD phenotypes in the 16p11.2 deletion mouse model is unclear. Moreover, whether blocking CD47 can alleviate ASD mice's behavioral deficits remains unknown. Here, we demonstrate a strong link between increased CD47 expression, decreased microglia phagocytosis capacity, and increased impairment in social novelty preference in the 16p11.2 deletion mice. The reduction in microglia phagocytosis caused a rise in excitatory synapses and transmission in the prefrontal cortex of 16p11.2 deletion mice. Importantly, blocking CD47 using a specific CD47 antibody or reducing CD47 expression using a specific short hairpin RNA (shRNA) enhanced the microglia phagocytosis and reduced excitatory transmission. Reduction in CD47 expression improved social novelty preference deficits in 16p11.2 mice. These findings demonstrate that CD47 is associated with the ASD phenotypes in the 16p11.2 deletion mice and could be a promising target for the development of treatment for ASD.
Collapse
Affiliation(s)
- Jun Ju
- Brain Research Centre, Department of Neurobiology, School of Life Sciences, Southern University of Science and Technology, Nanshan District, Shenzhen, Guangdong 1088, People's Republic of China
| | - Yifan Pan
- Brain Research Centre, Department of Neurobiology, School of Life Sciences, Southern University of Science and Technology, Nanshan District, Shenzhen, Guangdong 1088, People's Republic of China
| | - Xinyi Yang
- Brain Research Centre, Department of Neurobiology, School of Life Sciences, Southern University of Science and Technology, Nanshan District, Shenzhen, Guangdong 1088, People's Republic of China
| | - Xuanyi Li
- Brain Research Centre, Department of Neurobiology, School of Life Sciences, Southern University of Science and Technology, Nanshan District, Shenzhen, Guangdong 1088, People's Republic of China
| | - Jinghong Chen
- Brain Research Centre, Department of Neurobiology, School of Life Sciences, Southern University of Science and Technology, Nanshan District, Shenzhen, Guangdong 1088, People's Republic of China
| | - Shiyu Wu
- Brain Research Centre, Department of Neurobiology, School of Life Sciences, Southern University of Science and Technology, Nanshan District, Shenzhen, Guangdong 1088, People's Republic of China
| | - Sheng-Tao Hou
- Brain Research Centre, Department of Neurobiology, School of Life Sciences, Southern University of Science and Technology, Nanshan District, Shenzhen, Guangdong 1088, People's Republic of China
| |
Collapse
|
5
|
Malhotra S, Donneger F, Farrell JS, Dudok B, Losonczy A, Soltesz I. Integrating endocannabinoid signaling, CCK interneurons, and hippocampal circuit dynamics in behaving animals. Neuron 2025:S0896-6273(25)00188-6. [PMID: 40267911 DOI: 10.1016/j.neuron.2025.03.016] [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: 07/12/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 04/25/2025]
Abstract
The brain's endocannabinoid signaling system modulates a diverse range of physiological phenomena and is also involved in various psychiatric and neurological disorders. The basic components of the molecular machinery underlying endocannabinoid-mediated synaptic signaling have been known for decades. However, limitations associated with the short-lived nature of endocannabinoid lipid signals had made it challenging to determine the spatiotemporal specificity and dynamics of endocannabinoid signaling in vivo. Here, we discuss how novel technologies have recently enabled unprecedented insights into endocannabinoid signaling taking place at specific synapses in behaving animals. In this review, we primarily focus on cannabinoid-sensitive inhibition in the hippocampus in relation to place cell properties to illustrate the potential of these novel methodologies. In addition, we highlight implications of these approaches and insights for the unraveling of cannabinoid regulation of synapses in vivo in other brain circuits in both health and disease.
Collapse
Affiliation(s)
- Shreya Malhotra
- Department of Neurosurgery, Stanford University, Stanford, CA, USA.
| | - Florian Donneger
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Jordan S Farrell
- Department of Neurology, Harvard Medical School, Boston, MA, USA; Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA; F.M. Kirby Neurobiology Center, Harvard Medical School, Boston, MA, USA
| | - Barna Dudok
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, NY, USA; Kavli Institute for Brain Sciences, Columbia University, New York, NY, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| |
Collapse
|
6
|
Wang L, Liu R, Wang Y. The roles of extracellular vesicles in mental disorders: information carriers, biomarkers, therapeutic agents. Front Pharmacol 2025; 16:1591469. [PMID: 40271072 PMCID: PMC12014780 DOI: 10.3389/fphar.2025.1591469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025] Open
Abstract
Mental disorders are complex conditions that encompass various symptoms and types, affecting approximately 1 in 8 people globally. They place a significant burden on both families and society as a whole. So far, the etiology of mental disorders remains poorly understood, making diagnosis and treatment particularly challenging. Extracellular vesicles (EVs) are nanoscale particles produced by cells and released into the extracellular space. They contain bioactive molecules including nucleotides, proteins, lipids, and metabolites, which can mediate intercellular communication and are involved in various physiological and pathological processes. Recent studies have shown that EVs are closely linked to mental disorders like schizophrenia, major depressive disorder, and bipolar disorder, playing a key role in their development, diagnosis, prognosis, and treatment. Therefore, based on recent research findings, this paper aims to describe the roles of EVs in mental disorders and summarize their potential applications in diagnosis and treatment, providing new ideas for the future clinical transformation and application of EVs.
Collapse
Affiliation(s)
| | | | - Ying Wang
- Department of Pharmacy, Tianjin Anding Hospital, Tianjin, China
| |
Collapse
|
7
|
Chen XF, Kroke B, Ni J, Munoz C, Appleman M, Jacobs B, Tran T, Nguyen KV, Qiu C, Stonestreet BS, Marshall J. Novel peptidomimetic compounds attenuate hypoxic-ischemic brain injury in neonatal rats. Exp Neurol 2025; 386:115151. [PMID: 39832663 DOI: 10.1016/j.expneurol.2025.115151] [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: 10/21/2024] [Revised: 01/04/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Hypoxic-ischemic (HI) brain injury is a common neurological problem in neonates. The postsynaptic density protein-95 (PSD-95) is an excitatory synaptic scaffolding protein that regulates synaptic function, and represents a potential therapeutic target to attenuate HI brain injury. Syn3 and d-Syn3 are novel high affinity cyclic peptides that bind the PDZ3 domain of PSD-95. We investigated the neuroprotective efficacy of Syn3 and d-Syn3 after exposure to HI in neonatal rodents. Postnatal (P) day-7 rats were treated with Syn3 and d-Syn3 at zero, 24, and 48-h after carotid artery ligation and 90-min of 8 % oxygen. Hemispheric volume atrophy and Iba-1 positive microglia were quantified by cresyl violet and immunohistochemical staining. Treatment with Syn3 and d-Syn3 reduced tissue volume loss by 47.0 % and 41.0 % in the male plus female, and by 42.1 % and 65.0 % in the male groups, respectively. Syn3 reduced tissue loss by 52.3 % in females. D-Syn3 reduced Iba-1 positive microglia/DAPI ratios in the pooled group, males, and females. Syn3 effects were observed in the pooled group and females. Changes in Iba-1 positive microglia/DAPI cellular ratios correlated directly with reduced hemispheric volume loss, suggesting that Syn3 and d-Syn3 provide neuroprotection in part by their effects on Iba-1 positive microglia. The pathogenic cis phosphorylated Thr231 in Tau (cis P-tau) is a marker of neuronal injury. Cis P-tau was induced in cortical cells of the placebo-treated pooled group, males and females after HI, and reduced by treatment with d-Syn3. Therefore, treatment with these peptidomimetic agents exert neuroprotective effects after exposure of neonatal subjects to HI related brain injury.
Collapse
Affiliation(s)
- Xiaodi F Chen
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA.
| | - Brynn Kroke
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA; Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Jun Ni
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Christian Munoz
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA; Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Mark Appleman
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Bryce Jacobs
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Tuong Tran
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA
| | - Kevin V Nguyen
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA
| | - Chenxi Qiu
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Barbara S Stonestreet
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA; Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA; Emerita, Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA
| | - John Marshall
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA.
| |
Collapse
|
8
|
Tack RWP, Amboni C, van Nuijs D, Pekna M, Vergouwen MDI, Rinkel GJE, Hol EM. Inflammation, Anti-inflammatory Interventions, and Post-stroke Cognitive Impairment: a Systematic Review and Meta-analysis of Human and Animal Studies. Transl Stroke Res 2025; 16:535-546. [PMID: 38012509 PMCID: PMC11976800 DOI: 10.1007/s12975-023-01218-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/30/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023]
Abstract
The pathophysiology and treatment of post-stroke cognitive impairment (PSCI) are not clear. Stroke triggers an inflammatory response, which might affect synapse function and cognitive status. We performed a systematic review and meta-analysis to assess whether patients with PSCI have increased levels of inflammatory markers and whether anti-inflammatory interventions in animals decrease PSCI. We systematically searched PubMed, EMBASE, and PsychInfo for studies on stroke. For human studies, we determined the standardized mean difference (SMD) on the association between PSCI and markers of inflammation. For animal studies, we determined the SMD of post-stroke cognitive outcome after an anti-inflammatory intervention. Interventions were grouped based on proposed mechanism of action. In patients, the SMD of inflammatory markers for those with versus those without PSCI was 0.46 (95% CI 0.18; 0.76; I2 = 92%), and the correlation coefficient between level of inflammation and cognitive scores was - 0.25 (95% CI - 0.34; - 0.16; I2 = 75%). In animals, the SMD of cognition for those treated with versus those without anti-inflammatory interventions was 1.43 (95% CI 1.12; 1.74; I2 = 83%). The largest effect sizes in treated animals were for complement inhibition (SMD = 1.94 (95% CI 1.50; 2.37), I2 = 51%) and fingolimod (SMD = 2.1 (95% CI 0.75; 3.47), I2 = 81%). Inflammation is increased in stroke survivors with cognitive impairment and is negatively correlated with cognitive functioning. Anti-inflammatory interventions seem to improve cognitive functioning in animals. Complement inhibition and fingolimod are promising therapies on reducing PSCI.
Collapse
Affiliation(s)
- Reinier W P Tack
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| | - Claudia Amboni
- Department of Translational Neuroscience, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Danny van Nuijs
- Department of Translational Neuroscience, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Marcela Pekna
- Laboratory of Regenerative Neuroimmunology, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Mervyn D I Vergouwen
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Gabriel J E Rinkel
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
9
|
Xin J, Chen Y, Zhang L, Ma L. E2F2 induces microglial activation and augments depressive-like behavior in mice by repressing PTPN6 transcription. Cytotechnology 2025; 77:66. [PMID: 39991704 PMCID: PMC11845659 DOI: 10.1007/s10616-025-00730-w] [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: 12/16/2024] [Accepted: 02/13/2025] [Indexed: 02/25/2025] Open
Abstract
Depression is the leading contributor to disability and suicide ideation. Informed by the insights from bioinformatics analyses, this study investigates the roles of E2F transcription factor 2 (E2F2) and protein tyrosine phosphatase non-receptor type 6 (PTPN6) in the activation of microglia and the manifestation of depressive-like behavior in mice. Chronic unpredictable mild stress was applied to induce a mouse model of depression, while a cellular model featuring microglia was established through exposure to lipopolysaccharide and adenosine triphosphate. E2F2 was upregulated whereas PTPN6 was downregulated in these models. Notably, E2F2 was found to bind to the PTPN6 promoter, thereby repressing its transcription. Various behavioral tests demonstrated that silencing of E2F2, accomplished via shRNA transfection, led to increased locomotor activity, heightened social interaction rates, enhanced sucrose preference, and reduced immobility time in response to stress stimuli in mice. Furthermore, E2F2 silencing effectively reduced expression of Iba1, a microglial activation marker, and decreased concentrations of pro-inflammatory cytokines both in vivo and in vitro. However, these mitigating effects were countered by additional PTPN6 silencing. In conclusion, this study investigation underscores the role of E2F2 in promoting inflammatory activation of microglia and exacerbating depressive-like behavior in mice by repressing PTPN6 transcription.
Collapse
Affiliation(s)
- Jiali Xin
- Department of Psychiatry and Psychology, The Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Nangang District, Harbin, 150001 Heilongjiang P.R. China
| | - Yao Chen
- Department of Psychiatry and Psychology, The Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Nangang District, Harbin, 150001 Heilongjiang P.R. China
| | - Leijing Zhang
- Department of Psychiatry and Psychology, The Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Nangang District, Harbin, 150001 Heilongjiang P.R. China
| | - Lan Ma
- Department of Psychiatry and Psychology, The Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Nangang District, Harbin, 150001 Heilongjiang P.R. China
| |
Collapse
|
10
|
Zhang Y, Tang Y, Illes P. Modification of Neural Circuit Functions by Microglial P2Y6 Receptors in Health and Neurodegeneration. Mol Neurobiol 2025; 62:4139-4148. [PMID: 39400857 PMCID: PMC11880064 DOI: 10.1007/s12035-024-04531-8] [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: 05/13/2024] [Accepted: 10/02/2024] [Indexed: 10/15/2024]
Abstract
Neural circuits consisting of neurons and glial cells help to establish all functions of the CNS. Microglia, the resident immunocytes of the CNS, are endowed with UDP-sensitive P2Y6 receptors (P2Y6Rs) which regulate phagocytosis/pruning of excessive synapses during individual development and refine synapses in an activity-dependent manner during adulthood. In addition, this type of receptor plays a decisive role in primary (Alzheimer's disease, Parkinson's disease, neuropathic pain) and secondary (epilepsy, ischemic-, mechanical-, or irradiation-induced) neurodegeneration. A whole range of microglial cytokines controlled by P2Y6Rs, such as the interleukins IL-1β, IL-6, IL-8, and tumor necrosis factor-α (TNF-α), leads to neuroinflammation, resulting in neurodegeneration. Hence, small molecular antagonists of P2Y6Rs and genetic knockdown of this receptor provide feasible ways to alleviate inflammation-induced neurological disorders but might also interfere with the regulation of the synaptic circuitry. The present review aims at investigating this dual role of P2Y6Rs in microglia, both in shaping neural circuits by targeted phagocytosis and promoting neurodegenerative illnesses by fostering neuroinflammation through multiple transduction mechanisms.
Collapse
Affiliation(s)
- Yi Zhang
- International Joint Research Centre on Purinergic Signaling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yong Tang
- International Joint Research Centre on Purinergic Signaling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China.
- School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Peter Illes
- International Joint Research Centre on Purinergic Signaling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China.
- Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
11
|
Martinez MX, Mahler SV. Potential roles for microglia in drug addiction: Adolescent neurodevelopment and beyond. J Neuroimmunol 2025; 404:578600. [PMID: 40199197 DOI: 10.1016/j.jneuroim.2025.578600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/14/2025] [Accepted: 03/27/2025] [Indexed: 04/10/2025]
Abstract
Adolescence is a sensitive period for development of addiction-relevant brain circuits, and it is also when people typically start experimenting with drugs. Unfortunately, such substance use may cause lasting impacts on the brain, and might increase vulnerability to later-life addictions. Microglia are the brain's immune cells, but their roles in shaping neural connectivity and synaptic plasticity, especially in developmental sensitive periods like adolescence, may also contribute to addiction-related phenomena. Here, we overview how drugs of abuse impact microglia, and propose that they may play poorly-understood, but important roles in addiction vulnerability and progression.
Collapse
Affiliation(s)
- Maricela X Martinez
- Department of Neurobiology and Behavior, University of California, 2221 McGaugh Hall, Irvine, CA 92697, USA.
| | - Stephen V Mahler
- Department of Neurobiology and Behavior, University of California, 2221 McGaugh Hall, Irvine, CA 92697, USA
| |
Collapse
|
12
|
Huang Z, Ren Z, Wang S, Xiao L, Ling Y, Xie Y, Wang G, Zhou B. Urolithin A alleviates schizophrenic-like behaviors and cognitive impairment in rats through modulation of neuroinflammation, neurogenesis, and synaptic plasticity. Sci Rep 2025; 15:10477. [PMID: 40140679 PMCID: PMC11947095 DOI: 10.1038/s41598-025-93554-9] [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: 09/25/2024] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Cognitive impairment in schizophrenia occurs in the early stages of the disease and is closely associated with prognosis. Alleviation of cognitive impairment in schizophrenia faces major challenges owing to the lack of preventive and therapeutic drugs that are novel and effective. Urolithin A (UA) is a gut microbial metabolite of ellagic acid that has demonstrated neuroprotective effects in multiple neurological disease models. Nonetheless, the neuromodulatory role of UA in schizophrenia is yet to be elucidated. Wistar rat pups were separated from their mothers for 24 h on postnatal days (PNDs) 9-10 to establish an early-life stress model. The pups were pretreated with UA at different administration times (2, 4, and 6 weeks) and doses (50, 100, and 150 mg/kg) from adolescence (PND29). Behavioral tests were performed after the end of the administration. Subsequently, hippocampal samples were collected for histopathological and molecular evaluations. Male offspring rats subjected to maternal separation exhibited increased sensitivity to prepulse inhibition and cognitive impairment, accompanied by severe neuroinflammation and impaired neurogenesis. However, UA attenuated maternal separation-induced prepulse inhibition deficits and cognitive impairments and restored hippocampal neurogenesis in a dose-dependent manner. Furthermore, UA pretreatment preserved dendritic spine density, synapses, and presynaptic vesicles. In addition, it exerted anti-inflammatory effects by inhibiting microglial activation and expression of the proinflammatory cytokines tumor necrosis factor-α, interleukin-6, and interleukin-1β. Potential mechanisms included upregulation of brain-derived neurotrophic factor protein expression and activation of the extracellular signal-regulated kinase signaling pathway. This study is the first preclinical evaluation of the effects of UA on cognitive impairment in schizophrenia. The findings suggest that changes in cognitive function linked to schizophrenia are driven by the interaction among neuroinflammation, neurogenesis, and synaptic plasticity and that UA has the potential to reverse these processes. These observations provide evidence for future clinical trials of UA as a dietary supplement for preventing schizophrenia.
Collapse
Affiliation(s)
- Zhengyuan Huang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China
- Department of Pharmacy, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China
| | - Zhongyu Ren
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China
| | - Sanwang Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China
| | - Ling Xiao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China
- Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yipeng Ling
- Department of Pharmacy, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China
| | - Yinping Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China
- Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China.
- Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Donghu Road 115#, Wuhan, 430071, China.
| | - Benhong Zhou
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China.
- Department of Pharmacy, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China.
| |
Collapse
|
13
|
Marongiu R, Platholi J, Park L, Yu F, Sommer G, Woods C, Milner TA, Glass MJ. Perimenopause promotes neuroinflammation in select hippocampal regions in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643317. [PMID: 40161644 PMCID: PMC11952527 DOI: 10.1101/2025.03.14.643317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder characterized by age-dependent amyloid beta (Aβ) aggregation and accumulation, neuroinflammation, and cognitive deficits. Significantly, there are prominent sex differences in the risk, onset, progression, and severity of AD, as well as response to therapies, with disease burden disproportionally affecting women. Although menopause onset (i.e., perimenopause) may be a critical transition stage for AD susceptibility in women, the role of early ovarian decline in initial disease pathology, particularly key neuroinflammatory processes, is not well understood. To study this, we developed a unique mouse model of perimenopausal AD by combining an accelerated ovarian failure (AOF) model of menopause induced by 4-vinylcyclohexene diepoxide (VCD) with the 5xFAD transgenic AD mouse model. To target early stages of disease progression, 5xFAD females were studied at a young age (∼4 months) and at the beginning stage of ovarian failure analogous to human perimenopause (termed "peri-AOF"), and compared to age-matched males. Assessment of neuropathology was performed by immunohistochemical labeling of Aβ as well as markers of astrocyte and microglia activity in the hippocampus, a brain region involved in learning and memory that is deleteriously impacted during AD. Our results show that genotype, AOF, and sex contributed to AD-like pathology. Aggregation of Aβ was heightened in female 5xFAD mice and further increased at peri-AOF, with hippocampal subregion specificity. Further, select increases in glial activation also paralleled Aβ pathology in distinct hippocampal subregions. However, cognitive function was not affected by peri-AOF. These findings align with the hypothesis that perimenopause constitutes a period of susceptibility for AD pathogenesis in women.
Collapse
|
14
|
Belančić A, Janković T, Gkrinia EMM, Kristić I, Rajič Bumber J, Rački V, Pilipović K, Vitezić D, Mršić-Pelčić J. Glial Cells in Spinal Muscular Atrophy: Speculations on Non-Cell-Autonomous Mechanisms and Therapeutic Implications. Neurol Int 2025; 17:41. [PMID: 40137462 PMCID: PMC11944370 DOI: 10.3390/neurolint17030041] [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: 02/09/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by homozygous deletions or mutations in the SMN1 gene, leading to progressive motor neuron degeneration. While SMA has been classically viewed as a motor neuron-autonomous disease, increasing evidence indicates a significant role of glial cells-astrocytes, microglia, oligodendrocytes, and Schwann cells-in the disease pathophysiology. Astrocytic dysfunction contributes to motor neuron vulnerability through impaired calcium homeostasis, disrupted synaptic integrity, and neurotrophic factor deficits. Microglia, through reactive gliosis and complement-mediated synaptic stripping, exacerbate neurodegeneration and neuroinflammation. Oligodendrocytes exhibit impaired differentiation and metabolic support, while Schwann cells display abnormalities in myelination, extracellular matrix composition, and neuromuscular junction maintenance, further compromising motor function. Dysregulation of pathways such as NF-κB, Notch, and JAK/STAT, alongside the upregulation of complement proteins and microRNAs, reinforces the non-cell-autonomous nature of SMA. Despite the advances in SMN-restorative therapies, they do not fully mitigate glial dysfunction. Targeting glial pathology, including modulation of reactive astrogliosis, microglial polarization, and myelination deficits, represents a critical avenue for therapeutic intervention. This review comprehensively examines the multifaceted roles of glial cells in SMA and highlights emerging glia-targeted strategies to enhance treatment efficacy and improve patient outcomes.
Collapse
Affiliation(s)
- Andrej Belančić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Tamara Janković
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | | | - Iva Kristić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Jelena Rajič Bumber
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Valentino Rački
- Department of Neurology, Clinical Hospital Centre Rijeka, Krešimirova 42, 51000 Rijeka, Croatia;
| | - Kristina Pilipović
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Dinko Vitezić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Jasenka Mršić-Pelčić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| |
Collapse
|
15
|
Hermann DM, Wang C, Mohamud Yusuf A, Herz J, Doeppner TR, Giebel B. Extracellular vesicles lay the ground for neuronal plasticity by restoring mitochondrial function, cell metabolism and immune balance. J Cereb Blood Flow Metab 2025:271678X251325039. [PMID: 40072028 PMCID: PMC11904928 DOI: 10.1177/0271678x251325039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/15/2025] [Accepted: 02/14/2025] [Indexed: 03/15/2025]
Abstract
Extracellular vesicles (EVs) convey complex signals between cells that can be used to promote neuronal plasticity and neurological recovery in brain disease models. These EV signals are multimodal and context-dependent, making them unique therapeutic principles. This review analyzes how EVs released from various cell sources control neuronal metabolic function, neuronal survival and plasticity. Preferential sites of EV communication in the brain are interfaces between pre- and postsynaptic neurons at synapses, between astrocytes and neurons at plasma membranes or tripartite synapses, between oligodendrocytes and neurons at axons, between microglial cells/macrophages and neurons, and between cerebral microvascular cells and neurons. At each of these interfaces, EVs support mitochondrial function and cell metabolism under physiological conditions and orchestrate neuronal survival and plasticity in response to brain injury. In the injured brain, the promotion of neuronal survival and plasticity by EVs is tightly linked with EV actions on mitochondrial function, cell metabolism, oxidative stress and immune responses. Via the stabilization of cell metabolism and immune balance, neuronal plasticity responses are activated and functional neurological recovery is induced. As such, EV lay the ground for neuronal plasticity.
Collapse
Affiliation(s)
- Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Chen Wang
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ayan Mohamud Yusuf
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Josephine Herz
- Department of Pediatrics I, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Thorsten R Doeppner
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Department of Neurology, University Hospital Gießen and Marburg, Justus-Liebig-University Gießen, Gießen, Germany
| |
Collapse
|
16
|
Ding Y. Histone deacetylases: the critical enzymes for microglial activation involved in neuropathic pain. Front Pharmacol 2025; 16:1515787. [PMID: 40115267 PMCID: PMC11922887 DOI: 10.3389/fphar.2025.1515787] [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: 10/23/2024] [Accepted: 02/17/2025] [Indexed: 03/23/2025] Open
Abstract
Neuropathic pain is a common health problem in clinical practice that can be caused by many different factors, including infection, ischemia, trauma, diabetes mellitus, nerve compression, autoimmune disorders, cancer, trigeminal neuralgia, and abuse of certain drugs. This type of pain can persistently affect patients for a long time, even after the rehabilitation of their damaged tissues. Researchers have identified the crucial role of microglial activation in the pathogenesis of neuropathic pain. Furthermore, emerging evidence has shown that the expression and/or activities of different histone deacetylases (HDACs) can modulate microglial function and neuropathic pain. In this review, we will summarize and discuss the functions and mechanisms of HDACs in microglial activation and neuropathic pain development. Additionally, we will also list the emerging HDAC inhibitors or activators that may contribute to therapeutic advancement in alleviating neuropathic pain.
Collapse
Affiliation(s)
- Yi Ding
- Department of Anesthesiology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
17
|
Huang LY, Zhang YD, Chen J, Fan HD, Wang W, Wang B, Ma JY, Li PP, Pu HW, Guo XY, Shen JG, Qi SH. Maintaining moderate levels of hypochlorous acid promotes neural stem cell proliferation and differentiation in the recovery phase of stroke. Neural Regen Res 2025; 20:845-857. [PMID: 38886957 PMCID: PMC11433893 DOI: 10.4103/1673-5374.392889] [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: 02/08/2023] [Revised: 05/17/2023] [Accepted: 11/23/2023] [Indexed: 06/20/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202503000-00029/figure1/v/2024-06-17T092413Z/r/image-tiff It has been shown clinically that continuous removal of ischemia/reperfusion-induced reactive oxygen species is not conducive to the recovery of late stroke. Indeed, previous studies have shown that excessive increases in hypochlorous acid after stroke can cause severe damage to brain tissue. Our previous studies have found that a small amount of hypochlorous acid still exists in the later stage of stroke, but its specific role and mechanism are currently unclear. To simulate stroke in vivo, a middle cerebral artery occlusion rat model was established, with an oxygen-glucose deprivation/reoxygenation model established in vitro to mimic stroke. We found that in the early stage (within 24 hours) of ischemic stroke, neutrophils produced a large amount of hypochlorous acid, while in the recovery phase (10 days after stroke), microglia were activated and produced a small amount of hypochlorous acid. Further, in acute stroke in rats, hypochlorous acid production was prevented using a hypochlorous acid scavenger, taurine, or myeloperoxidase inhibitor, 4-aminobenzoic acid hydrazide. Our results showed that high levels of hypochlorous acid (200 μM) induced neuronal apoptosis after oxygen/glucose deprivation/reoxygenation. However, in the recovery phase of the middle cerebral artery occlusion model, a moderate level of hypochlorous acid promoted the proliferation and differentiation of neural stem cells into neurons and astrocytes. This suggests that hypochlorous acid plays different roles at different phases of cerebral ischemia/reperfusion injury. Lower levels of hypochlorous acid (5 and 100 μM) promoted nuclear translocation of β-catenin. By transfection of single-site mutation plasmids, we found that hypochlorous acid induced chlorination of the β-catenin tyrosine 30 residue, which promoted nuclear translocation. Altogether, our study indicates that maintaining low levels of hypochlorous acid plays a key role in the recovery of neurological function.
Collapse
Affiliation(s)
- Lin-Yan Huang
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yi-De Zhang
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Jie Chen
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Hai-Di Fan
- School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- Department of Laboratory Medicine, Branch Hospital of Huai’an First People’s Hospital, Huai’an, Jiangsu Province, China
| | - Wan Wang
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Bin Wang
- Department of Laboratory Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Ju-Yun Ma
- School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Peng-Peng Li
- Department of Laboratory Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Hai-Wei Pu
- Department of Laboratory Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Xin-Yian Guo
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Jian-Gang Shen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Su-Hua Qi
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| |
Collapse
|
18
|
Kuhn BN, Cannella N, Chitre AS, Nguyen KMH, Cohen K, Chen D, Peng B, Ziegler KS, Lin B, Johnson BB, Missfeldt Sanches T, Crow AD, Lunerti V, Gupta A, Dereschewitz E, Soverchia L, Hopkins JL, Roberts AT, Ubaldi M, Abdulmalek S, Kinen A, Hardiman G, Chung D, Polesskaya O, Solberg Woods LC, Ciccocioppo R, Kalivas PW, Palmer AA. Genome-wide association study reveals multiple loci for nociception and opioid consumption behaviors associated with heroin vulnerability in outbred rats. Mol Psychiatry 2025:10.1038/s41380-025-02922-4. [PMID: 40000848 DOI: 10.1038/s41380-025-02922-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/20/2024] [Accepted: 02/07/2025] [Indexed: 02/27/2025]
Abstract
The increased prevalence of opioid use disorder (OUD) makes it imperative to disentangle the biological mechanisms contributing to individual differences in OUD vulnerability. OUD shows strong heritability, however genetic variants contributing to vulnerability remain poorly defined. We performed a genome-wide association study using over 850 male and female heterogeneous stock (HS) rats to identify genes underlying behaviors associated with OUD such as nociception, as well as heroin-taking, extinction and seeking behaviors. By using an animal model of OUD, we were able to identify genetic variants associated with distinct OUD behaviors while maintaining a uniform environment, an experimental design not easily achieved in humans. Furthermore, we used a novel non-linear network-based clustering approach to characterize rats based on OUD vulnerability to assess genetic variants associated with OUD susceptibility. Our findings confirm the heritability of several OUD-like behaviors, including OUD susceptibility. Additionally, several genetic variants associated with nociceptive threshold prior to heroin experience, heroin consumption, escalation of intake, and motivation to obtain heroin were identified. Tom1, a microglial component, was implicated for nociception. Several genes involved in dopaminergic signaling, neuroplasticity and substance use disorders, including Brwd1, Pcp4, Phb1l2 and Mmp15 were implicated for the heroin traits. Additionally, an OUD vulnerable phenotype was associated with genetic variants for consumption and break point, suggesting a specific genetic contribution for OUD-like traits contributing to vulnerability. Together, these findings identify novel genetic markers related to the susceptibility to OUD-relevant behaviors in HS rats.
Collapse
Affiliation(s)
- Brittany N Kuhn
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| | - Nazzareno Cannella
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Apurva S Chitre
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Khai-Minh H Nguyen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Katarina Cohen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Denghui Chen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Beverly Peng
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Kendra S Ziegler
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Bonnie Lin
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Benjamin B Johnson
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | | | - Ayteria D Crow
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Veronica Lunerti
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Arkobrato Gupta
- The Interdisciplinary Ph.D. Program in Biostatistics, The Ohio State University, Columbus, OH, USA
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Eric Dereschewitz
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Laura Soverchia
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Jordan L Hopkins
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Analyse T Roberts
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Massimo Ubaldi
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Sarah Abdulmalek
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Analia Kinen
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Camerino, Italy
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Gary Hardiman
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
- Departments of Medicine and Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Dongjun Chung
- The Interdisciplinary Ph.D. Program in Biostatistics, The Ohio State University, Columbus, OH, USA
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Oksana Polesskaya
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | | | - Roberto Ciccocioppo
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
19
|
Nelson N, Miller V, Broadie K. Neuron-to-glia and glia-to-glia signaling directs critical period experience-dependent synapse pruning. Front Cell Dev Biol 2025; 13:1540052. [PMID: 40040788 PMCID: PMC11876149 DOI: 10.3389/fcell.2025.1540052] [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: 12/05/2024] [Accepted: 01/31/2025] [Indexed: 03/06/2025] Open
Abstract
Experience-dependent glial synapse pruning plays a pivotal role in sculpting brain circuit connectivity during early-life critical periods of development. Recent advances suggest a layered cascade of intercellular communication between neurons and glial phagocytes orchestrates this precise, targeted synapse elimination. We focus here on studies from the powerful Drosophila forward genetic model, with reference to complementary findings from mouse work. We present both neuron-to-glia and glia-to-glia intercellular signaling pathways directing experience-dependent glial synapse pruning. We discuss a putative hierarchy of secreted long-distance cues and cell surface short-distance cues that act to sequentially orchestrate glia activation, infiltration, target recognition, engulfment, and then phagocytosis for synapse pruning. Ligand-receptor partners mediating these stages in different contexts are discussed from recent Drosophila and mouse studies. Signaling cues include phospholipids, small neurotransmitters, insulin-like peptides, and proteins. Conserved receptors for these ligands are discussed, together with mechanisms where the receptor identity remains unknown. Potential mechanisms are proposed for the tight temporal-restriction of heightened experience-dependent glial synapse elimination during early-life critical periods, as well as potential means to re-open such plasticity at maturity.
Collapse
Affiliation(s)
- Nichalas Nelson
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, United States
| | - Vanessa Miller
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, United States
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, United States
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University and Medical Center, Nashville, TN, United States
- Kennedy Center for Research on Human Development, Vanderbilt University and Medical Center, Nashville, TN, United States
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN, United States
| |
Collapse
|
20
|
Memudu AE, Olukade BA, Nwanama KE, Alex GS. Models developed to explain the effects of stress on brain and behavior. PROGRESS IN BRAIN RESEARCH 2025; 291:339-361. [PMID: 40222786 DOI: 10.1016/bs.pbr.2025.01.018] [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: 04/15/2025]
Abstract
There is an integral relationship between stress, brain function and behavior. Over the year's extensive research has led to the development of various models to explain the intricate intersection between brain and stress. This chapter delves into some of the theoretical frameworks that explains the neurobiological and behavioral responses to stress using key models of stress such as the allostatic load model, which is the most common model that describes how chronic stress affect brain structure and function resulting in long-term changes in regions such as the hippocampus, amygdala, and prefrontal cortex which phenotypically express as cognitive impairments, emotional dysfunction seen in various forms of neurological disorder. The neuro-endocrine model, follows the glucocorticoid cascade hypothesis, that associates prolonged stress exposure to hippocampal damage and cognitive decline via alteration in the hypothalamic-pituitary-adrenal (HPA) axis and the overproduction of stress hormones like cortisol which can induce hippocampal atrophy, impair learning and memory, and promote depressive-like behaviors. The neurobiological stress model addresses the role of the hypothalamic-pituitary-adrenal (HPA) axis and stress-related neurotransmitters in shaping behavioral responses, emphasizing alterations in neuroplasticity and synaptic function. These models demonstrate how chronic stress can alter neural plasticity, neurotransmitter systems, and synaptic connectivity, affecting behavior and cognitive function. Hence by integrating molecular, neurobiological, and behavioral perspectives, these models offer a comprehensive understanding of how stress alters brain activity and behavior. The chapter further showcase how these models direct the development of medical interventions, shedding light on potential therapies that target the underlying molecular mechanisms of stress-induced brain changes.
Collapse
Affiliation(s)
- Adejoke Elizabeth Memudu
- Anatomy Department, Neuroscience Unit, Faculty of Basic Medical Sciences Edo State University Uzairue, Iyamho-Uzairue, Edo State, Nigeria.
| | - Baliqis Adejoke Olukade
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | | | - Gideon S Alex
- University of Port Harcourt, Port Harcourt, Rivers State, Nigeria
| |
Collapse
|
21
|
Chinnathambi S, Rangappa N, Chandrashekar M. Internalization of extracellular Tau oligomers in Alzheimer's disease. Adv Clin Chem 2025; 126:1-29. [PMID: 40185532 DOI: 10.1016/bs.acc.2025.01.005] [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] [Indexed: 04/07/2025]
Abstract
A key factor in the progression of Alzheimer's disease (AD) is internalization of extracellular Tau oligomers (ecTauOs) by neuroglial cells. Aberrant hyperphosphorylation of Tau results in their dissociation from microtubules and formation of toxic intracellular Tau oligomers (icTauOs). These are subsequently released to the extracellular space following neuronal dysfunction and death. Although receptor mediated internalization of these ecTauOs by other neurons, microglia and astrocytes can facilitate elimination, incomplete degradation thereof promotes inflammation, exacerbates pathologic spread and accelerates neurodegeneration. Targeting Tau oligomer degradation pathways, blocking internalization receptors, and mitigating neuroinflammation are proposed as therapeutic strategies to control Tau propagation and toxicity. This review highlights the urgent need for innovative approaches to prevent the spread of Tau pathology, emphasizing its implications for AD and related neurodegenerative diseases.
Collapse
Affiliation(s)
- Subashchandrabose Chinnathambi
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India.
| | - Nagaraj Rangappa
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India
| | - Madhura Chandrashekar
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India
| |
Collapse
|
22
|
Cao ZL, Zhu LX, Wang HM, Zhu LJ. Microglial Regulation of Neural Networks in Neuropsychiatric Disorders. Neuroscientist 2025:10738584251316558. [PMID: 39932233 DOI: 10.1177/10738584251316558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2025]
Abstract
Microglia serve as vital innate immune cells in the central nervous system, playing crucial roles in the generation and development of brain neurons, as well as mediating a series of immune and inflammatory responses. The morphologic transitions of microglia are closely linked to their function. With the advent of single-cell sequencing technology, the diversity of microglial subtypes is increasingly recognized. The intricate interactions between microglia and neuronal networks have significant implications for psychiatric disorders and neurodegenerative diseases. A deeper investigation of microglia in neurologic diseases such as Alzheimer disease, depression, and epilepsy can provide valuable insights in understanding the pathogenesis of diseases and exploring novel therapeutic strategies, thereby addressing issues related to central nervous system disorders.
Collapse
Affiliation(s)
- Zi-Lin Cao
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| | - Li-Xia Zhu
- Patent Examination Cooperation (JIANGSU) Center of the Patent Office, China National Intellectual Property Administration (CNIPA), Suzhou, China
| | - Hong-Mei Wang
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| | - Li-Juan Zhu
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
23
|
Hemati H, Blanton MB, True HE, Koura J, Khadka R, Grant KA, Messaoudi I. Phenotypic and Functional Alterations in Peripheral Blood Mononuclear Cell-Derived Microglia in a Primate Model of Chronic Alcohol Consumption. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636708. [PMID: 39975066 PMCID: PMC11839131 DOI: 10.1101/2025.02.05.636708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Alcohol-induced dysregulation of microglial activity is associated with neuroinflammation, cognitive decline, heightened risk for neurodegenerative diseases, alcohol dependence, and escalation of alcohol drinking. Given the challenge of longitudinally sampling primary microglia, we optimized an in vitro method to differentiate peripheral blood mononuclear cells (PBMC) from non-human primates (NHP) into microglia-like cells (induced-microglia; iMGL). The iMGLs displayed transcriptional profiles distinct from those of monocyte progenitors and closely resembling those of primary microglia. Notably, morphological features showed that differentiated iMGLs derived from NHPs with chronic alcohol consumption (CAC) possessed a more mature-like microglial morphology. Additionally, dysregulation in key inflammatory and regulatory markers alongside increased baseline phagocytic activity was observed in CAC-derived IMGLs in the resting state. Phenotypic and functional assessments following LPS stimulation indicated the presence of an immune-tolerant phenotype and enrichment of a CD86+ hyper-inflammatory subpopulation in iMGLs derived from ethanol-exposed animals. Collectively, these findings demonstrate that in vitro differentiation of PBMC offers a minimally invasive approach to studying the impact of CAC on microglial function revealing that CAC reshapes both functional and transcriptional profiles of microglia.
Collapse
Affiliation(s)
- Hami Hemati
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Madison B Blanton
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Heather E True
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Jude Koura
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Rupak Khadka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, OR, United States
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, OR, United States
| | - Ilhem Messaoudi
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
24
|
Zhao R, Wang J, Chung SK, Xu B. New insights into anti-depression effects of bioactive phytochemicals. Pharmacol Res 2025; 212:107566. [PMID: 39746497 DOI: 10.1016/j.phrs.2024.107566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
Depression is one of the most common psychological disorders, and due to its high prevalence and mortality rates, it imposes a significant disease burden. Contemporary treatments for depression involve various synthetic drugs, which have limitations such as side effects, single targets, and slow onset of action. Unlike synthetic medications, phytochemicals offer the benefits of a multi-target and multi-pathway mode of treatment for depression. In this literature review, we describe the pharmacological actions, experimental models, and clinical trials of the antidepressant effects of various phytochemicals. Additionally, we summarize the potential mechanisms by which these phytochemicals prevent depression, including regulating neurotransmitters and their receptors, the HPA axis, inflammatory responses, managing oxidative stress, neuroplasticity, and the gut microbiome. Phytochemicals exert therapeutic effects through multiple pathways and targets, making traditional Chinese medicine (TCM) a promising adjunctive antidepressant for the prevention, alleviation, and treatment of depression. Therefore, this review aims to provide robust evidence for subsequent research into developing phytochemical resources as effective antidepressant agents.
Collapse
Affiliation(s)
- Ruohan Zhao
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, Guangdong 519087, China
| | - Jingwen Wang
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, Guangdong 519087, China
| | - Sookja Kim Chung
- Faculty of Medicine, Macau University of Science and Technology, Macau, China.
| | - Baojun Xu
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, Guangdong 519087, China.
| |
Collapse
|
25
|
Strohm AO, Oldfield S, Hernady E, Johnston CJ, Marples B, O'Banion MK, Majewska AK. Biological sex, microglial signaling pathways, and radiation exposure shape cortical proteomic profiles and behavior in mice. Brain Behav Immun Health 2025; 43:100911. [PMID: 39677060 PMCID: PMC11634995 DOI: 10.1016/j.bbih.2024.100911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 12/17/2024] Open
Abstract
Patients receiving cranial radiation therapy experience tissue damage and cognitive deficits that severely decrease their quality of life. Experiments in rodent models show that these adverse neurological effects are in part due to functional changes in microglia, the resident immune cells of the central nervous system. Increasing evidence suggests that experimental manipulation of microglial signaling can regulate radiation-induced changes in the brain and behavior. Furthermore, many studies show sex-dependent neurological effects of radiation exposure. Despite this, few studies have used both males and females to explore how sex and microglial function interact to influence radiation effects on the brain. Here, we used a system levels approach to examine how deficiencies in purinergic and fractalkine signaling, two important microglial signaling pathways, impact brain proteomic and behavioral profiles in irradiated and control male and female mice. We performed a comprehensive analysis of the cortical proteomes from irradiated and control C57BL/6J, P2Y12-/-, and CX3CR1-/- mice of both sexes using multiple bioinformatics methods. We identified distinct proteins and biological processes, as well as behavioral profiles, regulated by sex, genotype, radiation exposure, and their interactions. Disrupting microglial signaling, had the greatest impact on proteomic expression, with CX3CR1-/- mice showing the most distinct proteomic profile characterized by upregulation of CX3CL1. Surprisingly, radiation exposure caused relatively smaller proteomic changes in glial and synaptic proteins, including Rgs10, Crybb1, C1qa, and Hexb. While we observed some radiation effects on locomotor behavior, biological sex as well as loss of P2Y12 and CX3CR1 signaling had a stronger influence on locomotor outcomes in our model. Lastly, loss of P2Y12 and CX3CR1 strongly regulated exploratory behaviors. Overall, our findings provide novel insights into the molecular pathways and proteins that are linked to P2Y12 and CX3CR1 signaling, biological sex, radiation exposure, and their interactions.
Collapse
Affiliation(s)
- Alexandra O. Strohm
- Departments of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Sadie Oldfield
- Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Eric Hernady
- Radiation Oncology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Carl J. Johnston
- Pediatrics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Brian Marples
- Radiation Oncology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - M. Kerry O'Banion
- Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Ania K. Majewska
- Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Center for Visual Science, University of Rochester Medical Center, Rochester, NY, 14642, USA
| |
Collapse
|
26
|
Tripathi A, Bartosh A, Mata J, Jacks C, Madeshiya AK, Hussein U, Hong LE, Zhao Z, Pillai A. Microglial type I interferon signaling mediates chronic stress-induced synapse loss and social behavior deficits. Mol Psychiatry 2025; 30:423-434. [PMID: 39095477 DOI: 10.1038/s41380-024-02675-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
Inflammation and synapse loss have been associated with deficits in social behavior and are involved in pathophysiology of many neuropsychiatric disorders. Synapse loss, characterized by reduction in dendritic spines can significantly disrupt synaptic connectivity and neural circuitry underlying social behavior. Chronic stress is known to induce loss of spines and dendrites in the prefrontal cortex (PFC), a brain region implicated in social behavior. However, the underlying mechanisms are not well understood. In the present study, we investigated the role of type I Interferon (IFN-I) signaling in chronic unpredictable stress (CUS)-induced synapse loss and behavior deficits in mice. We found increased expression of type I IFN receptor (IFNAR) in microglia following CUS. Conditional knockout of microglial IFNAR in adult mice rescued CUS-induced social behavior deficits and synapse loss. Bulk RNA sequencing data show that microglial IFNAR deletion attenuated CUS-mediated changes in the expression of genes such as Keratin 20 (Krt20), Claudin-5 (Cldn5) and Nuclear Receptor Subfamily 4 Group A Member 1 (Nr4a1) in the PFC. Cldn5 and Nr4a1 are known for their roles in synaptic plasticity. Krt20 is an intermediate filament protein responsible for the structural integrity of epithelial cells. The reduction in Krt20 following CUS presents a novel insight into the potential contribution of cytokeratin in stress-induced alterations in neuroplasticity. Overall, these results suggest that microglial IFNAR plays a critical role in regulating synaptic plasticity and social behavior deficits associated with chronic stress conditions.
Collapse
Affiliation(s)
- Ashutosh Tripathi
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alona Bartosh
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jocelyn Mata
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chale Jacks
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Amit Kumar Madeshiya
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Usama Hussein
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Lymphoma & Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L Elliot Hong
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhongming Zhao
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Anilkumar Pillai
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
27
|
Li X, Liu H, Xing P, Li T, Fang Y, Chen S, Dong S. Exosomal circRNAs: Deciphering the novel drug resistance roles in cancer therapy. J Pharm Anal 2025; 15:101067. [PMID: 39957900 PMCID: PMC11830318 DOI: 10.1016/j.jpha.2024.101067] [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: 04/11/2024] [Revised: 07/13/2024] [Accepted: 08/03/2024] [Indexed: 02/18/2025] Open
Abstract
Exosomal circular RNA (circRNAs) are pivotal in cancer biology, and tumor pathophysiology. These stable, non-coding RNAs encapsulated in exosomes participated in cancer progression, tumor growth, metastasis, drug sensitivity and the tumor microenvironment (TME). Their presence in bodily fluids positions them as potential non-invasive biomarkers, revealing the molecular dynamics of cancers. Research in exosomal circRNAs is reshaping our understanding of neoplastic intercellular communication. Exploiting the natural properties of exosomes for targeted drug delivery and disrupting circRNA-mediated pro-tumorigenic signaling can develop new treatment modalities. Therefore, ongoing exploration of exosomal circRNAs in cancer research is poised to revolutionize clinical management of cancer. This emerging field offers hope for significant breakthroughs in cancer care. This review underscores the critical role of exosomal circRNAs in cancer biology and drug resistance, highlighting their potential as non-invasive biomarkers and therapeutic targets that could transform the clinical management of cancer.
Collapse
Affiliation(s)
- Xi Li
- Department of Vascular and Thyroid Surgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Hanzhe Liu
- Department of Critical Care Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
| | - Peiyu Xing
- Department of Ophthalmology, China Medical University the Fourth People's Hospital of Shenyang, Shenyang, 110031, China
| | - Tian Li
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yi Fang
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Shuang Chen
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Siyuan Dong
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, 110001, China
| |
Collapse
|
28
|
García-Juárez M, García-Rodríguez A, Cruz-Carrillo G, Flores-Maldonado O, Becerril-Garcia M, Garza-Ocañas L, Torre-Villalvazo I, Camacho-Morales A. Intermittent Fasting Improves Social Interaction and Decreases Inflammatory Markers in Cortex and Hippocampus. Mol Neurobiol 2025; 62:1511-1535. [PMID: 39002056 DOI: 10.1007/s12035-024-04340-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 06/28/2024] [Indexed: 07/15/2024]
Abstract
Autism spectrum disorder (ASD) is a psychiatric condition characterized by reduced social interaction, anxiety, and stereotypic behaviors related to neuroinflammation and microglia activation. We demonstrated that maternal exposure to Western diet (cafeteria diet or CAF) induced microglia activation, systemic proinflammatory profile, and ASD-like behavior in the offspring. Here, we aimed to identify the effect of alternate day fasting (ADF) as a non-pharmacologic strategy to modulate neuroinflammation and ASD-like behavior in the offspring prenatally exposed to CAF diet. We found that ADF increased plasma beta-hydroxybutyrate (BHB) levels in the offspring exposed to control and CAF diets but not in the cortex (Cx) and hippocampus (Hpp). We observed that ADF increased the CD45 + cells in Cx of both groups; In control individuals, ADF promoted accumulation of CD206 + microglia cells in choroid plexus (CP) and increased in CD45 + macrophages cells and lymphocytes in the Cx. Gestational exposure to CAF diet promoted defective sociability in the offspring; ADF improved social interaction and increased microglia CD206 + in the Hpp and microglia complexity in the dentate gyrus. Additionally, ADF led to attenuation of the ER stress markers (Bip/ATF6/p-JNK) in the Cx and Hpp. Finally, biological modeling showed that fasting promotes higher microglia complexity in Cx, which is related to improvement in social interaction, whereas in dentate gyrus sociability is correlated with less microglia complexity. These data suggest a contribution of intermittent fasting as a physiological stimulus capable of modulating microglia phenotype and complexity in the brain, and social interaction in male mice.
Collapse
Affiliation(s)
- Martín García-Juárez
- Facultad de Medicina, Departamento de Bioquímica, Universidad Autónoma de Nuevo León, Madero y Dr. Aguirre Pequeño. Col. Mitras Centro, C.P. 64460, Monterrey, Nuevo León, Mexico
- Centro de Investigación y Desarrollo en Ciencias de La Salud, Universidad Autónoma de Nuevo León, Unidad de Neurometabolismo, Monterrey, Nuevo León, Mexico
| | - Adamary García-Rodríguez
- Facultad de Medicina, Departamento de Bioquímica, Universidad Autónoma de Nuevo León, Madero y Dr. Aguirre Pequeño. Col. Mitras Centro, C.P. 64460, Monterrey, Nuevo León, Mexico
- Centro de Investigación y Desarrollo en Ciencias de La Salud, Universidad Autónoma de Nuevo León, Unidad de Neurometabolismo, Monterrey, Nuevo León, Mexico
| | - Gabriela Cruz-Carrillo
- Facultad de Medicina, Departamento de Bioquímica, Universidad Autónoma de Nuevo León, Madero y Dr. Aguirre Pequeño. Col. Mitras Centro, C.P. 64460, Monterrey, Nuevo León, Mexico
- Centro de Investigación y Desarrollo en Ciencias de La Salud, Universidad Autónoma de Nuevo León, Unidad de Neurometabolismo, Monterrey, Nuevo León, Mexico
| | - Orlando Flores-Maldonado
- Facultad de Medicina, Departamento de Microbiología, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Miguel Becerril-Garcia
- Facultad de Medicina, Departamento de Microbiología, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Lourdes Garza-Ocañas
- Department of Pharmacology and Toxicology, College of Medicine, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, México
| | - Ivan Torre-Villalvazo
- Departamento de Fisiología de La Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), 14080, Mexico City, Mexico
| | - Alberto Camacho-Morales
- Facultad de Medicina, Departamento de Bioquímica, Universidad Autónoma de Nuevo León, Madero y Dr. Aguirre Pequeño. Col. Mitras Centro, C.P. 64460, Monterrey, Nuevo León, Mexico.
- Centro de Investigación y Desarrollo en Ciencias de La Salud, Universidad Autónoma de Nuevo León, Unidad de Neurometabolismo, Monterrey, Nuevo León, Mexico.
- College of Medicine, Universidad Autónoma de Nuevo Leon, San Nicolás de los Garza, NL, Mexico.
| |
Collapse
|
29
|
O'Keeffe M, Booker SA, Walsh D, Li M, Henley C, Simões de Oliveira L, Liu M, Wang X, Banqueri M, Ridley K, Dissanayake KN, Martinez-Gonzalez C, Craigie KJ, Vasoya D, Leah T, He X, Hume DA, Duguid I, Nolan MF, Qiu J, Wyllie DJA, Dando OR, Gonzalez-Sulser A, Gan J, Pridans C, Kind PC, Hardingham GE. Typical development of synaptic and neuronal properties can proceed without microglia in the cortex and thalamus. Nat Neurosci 2025; 28:268-279. [PMID: 39762658 PMCID: PMC11802452 DOI: 10.1038/s41593-024-01833-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/23/2024] [Indexed: 02/08/2025]
Abstract
Brain-resident macrophages, microglia, have been proposed to have an active role in synaptic refinement and maturation, influencing plasticity and circuit-level connectivity. Here we show that several neurodevelopmental processes previously attributed to microglia can proceed without them. Using a genetically modified mouse that lacks microglia (Csf1r∆FIRE/∆FIRE), we find that intrinsic properties, synapse number and synaptic maturation are largely normal in the hippocampal CA1 region and somatosensory cortex at stages where microglia have been implicated. Seizure susceptibility and hippocampal-prefrontal cortex coherence in awake behaving animals, processes that are disrupted in mice deficient in microglia-enriched genes, are also normal. Similarly, eye-specific segregation of inputs into the lateral geniculate nucleus proceeds normally in the absence of microglia. Single-cell and single-nucleus transcriptomic analyses of neurons and astrocytes did not uncover any substantial perturbation caused by microglial absence. Thus, the brain possesses remarkable adaptability to execute developmental synaptic refinement, maturation and connectivity in the absence of microglia.
Collapse
Affiliation(s)
- Mary O'Keeffe
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Sam A Booker
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Darren Walsh
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Mosi Li
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - Chloe Henley
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Laura Simões de Oliveira
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Mingshan Liu
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - Xingran Wang
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - Maria Banqueri
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Katherine Ridley
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - Kosala N Dissanayake
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Cristina Martinez-Gonzalez
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Kirsty J Craigie
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Deepali Vasoya
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - Tom Leah
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - Xin He
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - David A Hume
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Ian Duguid
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Matthew F Nolan
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Jing Qiu
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - David J A Wyllie
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Owen R Dando
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - Alfredo Gonzalez-Sulser
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Jian Gan
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - Clare Pridans
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
- Centre for Inflammation Research, The Queen's Medical Research Institute, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Peter C Kind
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK.
| | - Giles E Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK.
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
30
|
Rong X, Wei F, Jiang Y, Ma Q, Wang D, Shen J. Microglial activation and hypothalamic structural plasticity in HFD obesity: insights from semaglutide and minocycline. J Lipid Res 2025; 66:100736. [PMID: 39724960 PMCID: PMC11788729 DOI: 10.1016/j.jlr.2024.100736] [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: 06/20/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
High-fat diet (HFD)-induced microglial activation contributes to hypothalamic inflammation and obesity, but the mechanisms linking microglia to structural changes remain unclear. This study explored the role of microglia in impairing hypothalamic synaptic plasticity in diet-induced obesity mice and evaluated the therapeutic potential of semaglutide (Sema) and minocycline (MI). Six-week-old C57BL/6J mice were divided into low-fat diet and HFD groups. At week 30, the HFD-fed mice were treated daily with Sema or MI for six weeks. Confocal microscopy assessed hypothalamic dendritic spines, synaptic organization, and microglia-synapse interactions. We also analyzed microglial morphology, CD68/CD11b colocalization with Iba-1, synaptic marker expression, and phagocytosis-related pathways (C1q, C3, CD11b). BV2 microglia were used to examine the direct effects of Sema or MI on microglia and validate the in vivo findings. HFD feeding induced microglial activation, as indicated by increased colocalization of CD68 or synaptophysin and CD11b with Iba-1, along with elevated C1q, C3, and CD11b expression, signaling enhanced synaptic phagocytosis. This was accompanied by reduced hypothalamic dendritic spines, decreased synaptic marker expression, and disrupted excitatory/inhibitory synaptic organization in the melanocortin system, as well as impaired glucose metabolism, disrupted leptin-ghrelin balance, and increased food intake and body weight. Sema and MI treatments reversed the pathological changes of microglial activation and restored hypothalamic synaptic structure, although their effects on synaptic organization and metabolic outcomes differed. Our findings highlight the key role of microglial activation in hypothalamic synaptic impairment in diet-induced obesity models, with Sema and MI possibly offering distinct therapeutic pathways to mitigate these impairments.
Collapse
Affiliation(s)
- Xi Rong
- Department of Endocrinology and Metabolism, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong Province, China; Department of Geriatric Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Fang Wei
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yuqi Jiang
- Department of Endocrinology and Metabolism, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong Province, China
| | - Qintao Ma
- Department of Endocrinology and Metabolism, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong Province, China
| | - Dongmei Wang
- Department of Endocrinology and Metabolism, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong Province, China
| | - Jie Shen
- Department of Endocrinology and Metabolism, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong Province, China.
| |
Collapse
|
31
|
Ji Y, Yang C, Pang X, Yan Y, Wu Y, Geng Z, Hu W, Hu P, Wu X, Wang K. Repetitive transcranial magnetic stimulation in Alzheimer's disease: effects on neural and synaptic rehabilitation. Neural Regen Res 2025; 20:326-342. [PMID: 38819037 PMCID: PMC11317939 DOI: 10.4103/nrr.nrr-d-23-01201] [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: 07/19/2023] [Revised: 10/23/2023] [Accepted: 12/13/2023] [Indexed: 06/01/2024] Open
Abstract
Alzheimer's disease is a neurodegenerative disease resulting from deficits in synaptic transmission and homeostasis. The Alzheimer's disease brain tends to be hyperexcitable and hypersynchronized, thereby causing neurodegeneration and ultimately disrupting the operational abilities in daily life, leaving patients incapacitated. Repetitive transcranial magnetic stimulation is a cost-effective, neuro-modulatory technique used for multiple neurological conditions. Over the past two decades, it has been widely used to predict cognitive decline; identify pathophysiological markers; promote neuroplasticity; and assess brain excitability, plasticity, and connectivity. It has also been applied to patients with dementia, because it can yield facilitatory effects on cognition and promote brain recovery after a neurological insult. However, its therapeutic effectiveness at the molecular and synaptic levels has not been elucidated because of a limited number of studies. This study aimed to characterize the neurobiological changes following repetitive transcranial magnetic stimulation treatment, evaluate its effects on synaptic plasticity, and identify the associated mechanisms. This review essentially focuses on changes in the pathology, amyloidogenesis, and clearance pathways, given that amyloid deposition is a major hypothesis in the pathogenesis of Alzheimer's disease. Apoptotic mechanisms associated with repetitive transcranial magnetic stimulation procedures and different pathways mediating gene transcription, which are closely related to the neural regeneration process, are also highlighted. Finally, we discuss the outcomes of animal studies in which neuroplasticity is modulated and assessed at the structural and functional levels by using repetitive transcranial magnetic stimulation, with the aim to highlight future directions for better clinical translations.
Collapse
Affiliation(s)
- Yi Ji
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Chaoyi Yang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Xuerui Pang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Yibing Yan
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Yue Wu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Zhi Geng
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Wenjie Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Panpan Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui Province, China
| | - Xingqi Wu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui Province, China
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui Province, China
- Department of Psychology and Sleep Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
32
|
Chen J, He Y, Zhong J, Fu Y, Yuan S, Hou L, Zhang X, Meng F, Lin WJ, Ji F, Wang Z. Transcranial near-infrared light promotes remyelination through AKT1/mTOR pathway to ameliorate postoperative neurocognitive disorder in aged mice. Neuroscience 2025; 565:358-368. [PMID: 39653248 DOI: 10.1016/j.neuroscience.2024.12.007] [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: 07/04/2024] [Revised: 11/10/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024]
Abstract
Postoperative neurocognitive disorder (PND) is a prevalent complication following surgery and anesthesia, characterized by progressive cognitive decline. The precise etiology of PND remains unknown, and effective targeted therapeutic strategies are lacking. Transcranial near-infrared light (tNIRL) has shown potential benefits for cognitive dysfunction diseases, but its effect on PND remains unclear. Our previous research indicated a close association between demyelination and PND. In other central nervous system (CNS) disorders, tNIRL has been demonstrated to facilitate remyelination in response to demyelination. In this study, we established the PND model in 18-month-old male C57BL/6 mice using isoflurane anesthesia combined with left common carotid artery exposure. Following surgery, PND-aged mice were subjected to daily 2.5-minute tNIRL treatment at 810 nm for three consecutive days. Subsequently, we observed that tNIRL significantly improved cognitive performance and reduced inflammatory cytokine levels in the hippocampus of PND mice. Furthermore, tNIRL increased the expression of oligodendrocyte transcription factor 2 (OLIG2) and myelin basic protein (MBP), promoting remyelination while enhancing synaptic function-associated proteins such as synaptophysin (SYP) and postsynaptic density protein 95 (PSD95). Further investigation revealed that tNIRL may activate the AKT1/mTOR pathway to facilitate remyelination in PND mice. These findings indicate that tNIRL is a novel non-invasive therapeutic approach for treating PND.
Collapse
Affiliation(s)
- Jiawei Chen
- Department of Anesthesiology, Meishan City People's Hospital, Meishan, Sichuan, China
| | - Yuqing He
- Department of Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Junying Zhong
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yanni Fu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shangyan Yuan
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Longjie Hou
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaojun Zhang
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Fanqing Meng
- Department of Anesthesiology, Jinan Maternity and Child Care Hospital, Jinan, Shandong, China
| | - Wei-Jye Lin
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fengtao Ji
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Zhi Wang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
33
|
Wu C, Li M, Chen Z, Feng S, Deng Q, Duan R, Liu TCY, Yang L. Remote photobiomodulation ameliorates behavioral and neuropathological outcomes in a rat model of repeated closed head injury. Transl Psychiatry 2025; 15:8. [PMID: 39799140 PMCID: PMC11724958 DOI: 10.1038/s41398-025-03228-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 12/13/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025] Open
Abstract
Repeated closed-head injuries (rCHI) from activities like contact sports, falls, military combat, and traffic accidents pose a serious risk due to their cumulative impact on the brain. Often, rCHI is not diagnosed until symptoms of irreversible brain damage appear, highlighting the need for preventive measures. This study assessed the prophylactic efficacy of remote photobiomodulation (PBM) targeted at the lungs against rCHI-induced brain injury and associated behavioral deficits. Utilizing the "Marmarou" weight-drop model, rCHI was induced in rats on days 0, 5, and 10. Remote PBM, employing an 808 nm continuous wave laser, was administered daily in 2-min sessions per lung side over 20 days. Behavioral deficits were assessed through three-chamber social interaction, forced swim, grip strength, open field, elevated plus maze, and Barnes maze tests. Immunofluorescence staining and 3D reconstruction evaluated neuronal damage, apoptosis, degeneration, and the morphology of microglia and astrocytes, as well as astrocyte and microglia-mediated excessive synapse elimination. Additionally, 16S rDNA amplicon sequencing analyzed changes in the lung microbiome following remote PBM treatment. Results demonstrated that remote PBM significantly improved depressive-like behaviors, motor dysfunction, and social interaction impairment while enhancing grip strength and reducing neuronal damage, apoptosis, and degeneration induced by rCHI. Analysis of lung microbiome changes revealed an enrichment of lipopolysaccharide (LPS) biosynthesis pathways, suggesting a potential link to neuroprotection. Furthermore, remote PBM mitigated hyperactivation of cortical microglia and astrocytes and significantly reduced excessive synaptic phagocytosis by these cells, highlighting its potential as a preventive strategy for rCHI with neuroprotective effects.
Collapse
Affiliation(s)
- Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China
| | - Meng Li
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China
| | - Zhe Chen
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China
| | - Shu Feng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China
| | - Qianting Deng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China
| | - Rui Duan
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Timon Cheng-Yi Liu
- Laboratory of Laser Sports Medicine, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
34
|
Godeanu S, Mușat MI, Scheller A, Osiac E, Cătălin B. Minimal differences observed when comparing the morphological profiling of microglia obtained by confocal laser scanning and optical sectioning microscopy. Front Neuroanat 2025; 18:1507140. [PMID: 39829733 PMCID: PMC11739110 DOI: 10.3389/fnana.2024.1507140] [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: 10/07/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025] Open
Abstract
Background While widefield microscopy has long been constrained by out-of-focus scattering, advancements have generated a solution in the form of confocal laser scanning microscopy (cLSM) and optical sectioning microscopy using structured illumination (OSM). In this study, we aim to investigate, using microglia branching, if cLSM and OSM can produce images with comparable morphological characteristics. Results By imaging the somatosensory microglia from a tissue slice of a 3-week-old mouse and establishing morphological parameters that characterizes the microglial branching pattern, we were able to show that there is no difference in total length of the branch tree, number of branches, mean branch length and number of primary to terminal branches. We did find that area-based parameters such as mean occupied area and mean surveillance area were bigger in cLSM isolated microglia compared to OSM ones. Additionally, by investigating the difference in acquisition time between techniques and personal costs we were able to establish that the amortization could be made in 6.11 ± 2.93 years in the case of countries with a Human Development Index (HDI) = 7-9 and 7.06 ± 3.13 years, respectably, for countries with HDI < 7. As such, OSM systems seem a valid option if one just wants basic histological evaluation, and cLSM should be considered for groups that demand higher resolution or volumetric images.
Collapse
Affiliation(s)
- Sânziana Godeanu
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Craiova, Romania
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Saarbrücken, Germany
| | - Mădălina Iuliana Mușat
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Anja Scheller
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Saarbrücken, Germany
- Center for Gender-Specific Biology and Medicine (CGBM), University of Saarland, Saarbrücken, Germany
| | - Eugen Osiac
- Department of Biophysics, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Bogdan Cătălin
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Craiova, Romania
- Department of Physiology, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| |
Collapse
|
35
|
Salminen A. The role of inhibitory immune checkpoint receptors in the pathogenesis of Alzheimer's disease. J Mol Med (Berl) 2025; 103:1-19. [PMID: 39601807 PMCID: PMC11739239 DOI: 10.1007/s00109-024-02504-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: 06/24/2024] [Revised: 10/16/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
There is mounting evidence that microglial cells have a key role in the pathogenesis of Alzheimer's disease (AD). In AD pathology, microglial cells not only are unable to remove β-amyloid (Aβ) plaques and invading pathogens but also are involved in synaptic pruning, chronic neuroinflammation, and neuronal degeneration. Microglial cells possess many different inhibitory immune checkpoint receptors, such as PD-1, LILRB2-4, Siglecs, and SIRPα receptors, which can be targeted by diverse cell membrane-bound and soluble ligand proteins to suppress the functions of microglia. Interestingly, in the brains of AD patients there are elevated levels of many of the inhibitory ligands acting via these inhibitory checkpoint receptors. For instance, Aβ oligomers, ApoE4, and fibronectin are able to stimulate the LILRB2-4 receptors. Increased deposition of sialoglycans, e.g., gangliosides, inhibits microglial function via Siglec receptors. AD pathology augments the accumulation of senescent cells, which are known to possess a high level of PD-L1 proteins, and thus, they can evade immune surveillance. A decrease in the expression of SIRPα receptor in microglia and its ligand CD47 in neurons enhances the phagocytic pruning of synapses in AD brains. Moreover, cerebral neurons contain inhibitory checkpoint receptors which can inhibit axonal growth, reduce synaptic plasticity, and impair learning and memory. It seems that inappropriate inhibitory immune checkpoint signaling impairs the functions of microglia and neurons thus promoting AD pathogenesis. KEY MESSAGES: Microglial cells have a major role in the pathogenesis of AD. A decline in immune activity of microglia promotes AD pathology. Microglial cells and neurons contain diverse inhibitory immune checkpoint receptors. The level of ligands for inhibitory checkpoint receptors is increased in AD pathology. Impaired signaling of inhibitory immune checkpoint receptors promotes AD pathology.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
36
|
Bonomi RE, Pietrzak R, Cosgrove KP. Neuroglia in anxiety disorders. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:335-346. [PMID: 40148054 DOI: 10.1016/b978-0-443-19102-2.00008-9] [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: 03/29/2025]
Abstract
Anxiety disorders are some of the most prevalent in the world and are extraordinarily debilitating to many individuals, costing millions in disability. One of the most disabling is posttraumatic stress disorder (Snijders et al., 2020). Understanding the pathophysiology of these illnesses further and the cell types involved will allow better targeting of treatments. Glial cells, encompassing microglia, astrocytes, and oligodendrocytes, play critical roles in the pathophysiology of PTSD and other anxiety illnesses. Each of these cell types interacts with aspects of neuro-epigenetics, neuroimmune, and neuronal signaling and may contribute to the pathophysiology of anxiety illnesses. This chapter covers the literature on the role of glial cells in the neurobiology and pathology of anxiety disorders, more specifically PTSD. PTSD is one of the most debilitating anxiety disorders and one of the most complicated from a neurobiologic perspective. This chapter also features a discussion surrounding the current state of treatment and some of the hypothesized mechanisms for novel treatments including tetrahydrocannabidiol and 3,4-methylenedioxymethamphetamine. Finally, thoughts on the future directions for precision treatment and pharmacologic development with a focus on neuroglia are undertaken.
Collapse
Affiliation(s)
- Robin E Bonomi
- Department of Psychiatry, Yale University, New Haven, CT, United States
| | - Robert Pietrzak
- Department of Psychiatry, Yale University, New Haven, CT, United States
| | - Kelly P Cosgrove
- Department of Psychiatry, Yale University, New Haven, CT, United States.
| |
Collapse
|
37
|
Lendemeijer B, de Vrij FMS. In vitro models for human neuroglia. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:213-227. [PMID: 40122626 DOI: 10.1016/b978-0-443-19104-6.00015-2] [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: 03/25/2025]
Abstract
Neuroglia are a heterogenous population of cells in the nervous system. In the central nervous system, this group is classified into astrocytes, oligodendrocytes, and microglia. Neuroglia in the peripheral nervous system are divided into Schwann cells and enteric glia. These groups of cells display considerable differences in their developmental origin, morphology, function, and regional abundance. Compared to animal models, human neuroglia differ in their transcriptomic profile, morphology, and function. Investigating the physiology of healthy or diseased human neuroglia in vivo is challenging due to the inaccessibility of the tissue. Therefore, researchers have developed numerous in vitro models attempting to replicate the natural tissue environment. Earlier models made use of postmortem, postsurgical, or fetal tissue to establish human neuroglial cells in vitro, either as a pure population of the desired cell type or as organotypic slice cultures. Advancements in human stem cell differentiation techniques have greatly enhanced the possibilities for creating in vitro models of human neuroglia. This chapter provides an overview of the current models used to study the functioning and development of human neuroglia in vitro, both in health and disease.
Collapse
Affiliation(s)
- Bas Lendemeijer
- Department of Psychiatry, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Psychiatry, Columbia University Medical Center, New York, NY, United States
| | - Femke M S de Vrij
- Department of Psychiatry, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
38
|
Henry RJ, Loane DJ. Unraveling the complexity of microglial responses in traumatic brain and spinal cord injury. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:113-132. [PMID: 40148040 DOI: 10.1016/b978-0-443-19102-2.00015-6] [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: 03/29/2025]
Abstract
Microglia, the resident innate immune cells of the central nervous system (CNS), play an important role in neuroimmune signaling, neuroprotection, and neuroinflammation. In the healthy CNS, microglia adopt a surveillant and antiinflammatory phenotype characterized by a ramified scanning morphology that maintains CNS homeostasis. In response to acquired insults, such as traumatic brain injury (TBI) or spinal cord injury (SCI), microglia undergo a dramatic morphologic and functional switch to that of a reactive state. This microglial switch is initially protective and supports the return of the injured tissue to a physiologic homeostatic state. However, there is now a significant body of evidence that both TBI and SCI can result in a chronic state of microglial activation, which contributes to neurodegeneration and impairments in long-term neurologic outcomes in humans and animal models. In this review, we discuss the complex role of microglia in the pathophysiology of TBI and SCI, and recent advancements in knowledge of microglial phenotypic states in the injured CNS. Furthermore, we highlight novel therapeutic strategies targeting chronic microglial responses in experimental models and discuss how they may ultimately be translated to the clinic for human brain and SCI.
Collapse
Affiliation(s)
- Rebecca J Henry
- Department of Pharmacology, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.
| | - David J Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
39
|
Hong C, Wang L, Zhou X, Zou L, Xiang X, Deng H, Li Q, Wu Y, Liu L, Li T. Protective Effects of Mdivi-1 on Cognition Disturbance Following Sepsis in Mice via Alleviating Microglia Activation and Polarization. CNS Neurosci Ther 2025; 31:e70149. [PMID: 39791542 PMCID: PMC11719124 DOI: 10.1111/cns.70149] [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: 05/20/2024] [Revised: 10/09/2024] [Accepted: 10/20/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Neuroinflammation is one of the essential pathogeneses of cognitive damage suffering from sepsis-associated encephalopathy (SAE). Lots of evidences showed the microglia presented mitochondrial fragmentation during SAE. This study investigated the protective effects and novel mechanisms of inhibiting microglia mitochondrial fragmentation via mitochondrial division inhibitor 1 (Mdivi-1) on cognitive damage in SAE. METHODS The SAE model was performed by cecal ligation and puncture (CLP), and Mdivi-1 was administrated via intraperitoneal injection. Morris water maze was performed to assess cognitive function. Mitochondrial morphology was observed by electron microscope or MitoTracker staining. The qRT-PCR, immunofluorescence staining, and western blots were used to detect the inflammatory factors and protein content, respectively. Flow cytometry was used to detect the polarization of hippocampal microglia. Bioinformatics analysis was used to verify hypotheses. RESULTS Mdivi-1 administration alleviated sepsis-induced mitochondrial fragmentation, microglia activation, polarization, and cognitive damage. The mechanisms study showed neuroinflammation and oxidative stress were suppressed via NF-κB and Keap1/Nrf2/HO-1 pathways following Mdivi-1 administration; meanwhile, pyroptosis in microglia was reduced, which was associated with enhanced autophagosome formation via p62 elevation following Mdivi-1 administration. CONCLUSION Inhibition of microglia mitochondrial fragmentation is beneficial to SAE cognitive disturbance, the mechanisms are related to alleviating neuroinflammation, oxidative stress, and pyroptosis.
Collapse
Affiliation(s)
- Chen Hong
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Li Wang
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Xiaowei Zhou
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Liyong Zou
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Xinming Xiang
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Haoyue Deng
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Qinghui Li
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Yue Wu
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Liangming Liu
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Tao Li
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| |
Collapse
|
40
|
Yang F, Gao W, Wang J, Li X, Li H. Progress of Chinese Medicine in Regulating Microglial Polarization against Alzheimer's Disease. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2255-2275. [PMID: 39721955 DOI: 10.1142/s0192415x24500873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Alzheimer's disease (AD), the predominant form of dementia, is a neurodegenerative disorder of the central nervous system (CNS) characterized by a subtle onset and a spectrum of cognitive and functional declines. The clinical manifestation of AD encompasses memory deficits, cognitive deterioration, and behavioral disturbances, culminating in a severe impairment of daily living skills. Despite its high prevalence, accounting for 60-70% of all dementia cases, there remains an absence of curative therapeutics. Microglia (MG), the resident immune cells of the CNS, exhibit a bifurcated role in AD pathogenesis. Functioning in a neuroprotective capacity, MGs express scavenger receptors, facilitating the clearance of [Formula: see text]-amyloid protein (A[Formula: see text]) and cellular debris. Conversely, aberrant activation of MGs can lead to the secretion of pro-inflammatory cytokines, thereby propagating neuroinflammatory responses that are detrimental to neuronal integrity. The dynamics of MG activation and the ensuing neuroinflammation are pivotal in the evolution of AD. Chinese medicine (CM), a treasure trove of traditional Chinese cultural practices, has demonstrated significant potential in the therapeutic management of AD. Over the past triennium, CM has garnered considerable research attention for its multifaceted approaches to AD, including the regulation of MG polarization. This review synthesizes current knowledge on the origins, polarization dynamics, and mechanistic interplay of MG with AD pathology. It further explores the nexus between MG polarization and cardinal pathological hallmarks of AD, such as A[Formula: see text] plaque deposition, hyperphosphorylation of tau, synaptic plasticity impairments, neuroinflammation, and brain-gut-axis dysregulation. The review also encapsulates the therapeutic strategies of CM, which encompass monomers, formulae, and acupuncture. These strategies modulate MG polarization in the context of AD treatment, thereby providing a robust theoretical framework in which to conduct future investigative endeavors in both the clinical and preclinical realms.
Collapse
Affiliation(s)
- Fengge Yang
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
| | - Wei Gao
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
- Jiangsu College of Nursing Jiangsu, Huaian, Huaiyin 223001, P. R. China
| | - Junting Wang
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
- The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P. R. China
| | - Xue Li
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
| | - Honglin Li
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
- The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P. R. China
| |
Collapse
|
41
|
Li J, Li C, Subedi U, Subedi P, Panchatcharam M, Sun H. The Role of Endothelial L-PGDS in the Pro-Angiogenic and Anti-Inflammatory Effects of Low-Dose Alcohol Consumption. Cells 2024; 13:2007. [PMID: 39682755 PMCID: PMC11640003 DOI: 10.3390/cells13232007] [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: 11/06/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Light alcohol consumption (LAC) may reduce the incidence and improve the prognosis of ischemic stroke. Recently, we found that LAC promotes cerebral angiogenesis and inhibits early inflammation following ischemic stroke. In addition, LAC upregulates lipocalin-type prostaglandin D2 synthase (L-PGDS) in the brain. Thus, we determined the role of endothelial L-PGDS in the protective effect of LAC. In in vitro studies, chronic exposure to low-concentration ethanol upregulated L-PGDS and significantly increased the proliferation in cultured C57BL/6J mouse brain microvascular endothelial cells (MBMVECs). AT-56, a selective L-PGDS inhibitor, abolished low-concentration ethanol exposure-induced proliferation. In in vivo studies, 8-week gavage feeding with 0.7 g/kg/day ethanol, defined as LAC, promoted cerebral angiogenesis under physiological conditions and following ischemic stroke in male C57BL/6J mice. In addition, LAC inhibited the post-ischemic expression of adhesion molecules, neutrophil infiltration, and microglial activation. AT-56 and endothelial cell (EC)-specific L-PGDS conditional knockout did not significantly alter cerebral angiogenesis and post-ischemic inflammation in the control mice but eliminated the pro-angiogenic and anti-inflammatory effects of LAC. Furthermore, EC-specific L-PGDS conditional knockout alleviated the neuroprotective effect of LAC against cerebral ischemia/reperfusion injury. These findings suggest that endothelial L-PGDS may be crucial in the pro-angiogenic and anti-inflammatory effects of LAC against ischemic stroke.
Collapse
Affiliation(s)
| | | | | | | | | | - Hong Sun
- Department of Cellular Biology and Anatomy, LSU Health Shreveport, Shreveport, LA 71103, USA; (J.L.); (C.L.); (U.S.); (P.S.); (M.P.)
| |
Collapse
|
42
|
Wehrli JM, Xia Y, Meister L, Tursunova S, Kleim B, Bach DR, Quednow BB. Forget me not: The effect of doxycycline on human declarative memory. Eur Neuropsychopharmacol 2024; 89:1-9. [PMID: 39217739 DOI: 10.1016/j.euroneuro.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
Investigations into neuroprotective drugs are in high demand for the treatment of neurodegenerative diseases, such as multiple sclerosis or Alzheimer's disease, but also psychiatric disorders, such as depression, trauma, and substance use. One potential drug class being investigated are tetracyclines impacting on a variety of neuroprotective mechanisms. At the same time, tetracyclines like doxycycline have been suggested to affect human fear and spatial memory as well as reducing declarative memory retention. Based on the assumed necessity for synaptic consolidation in hippocampus-dependent learning, we hypothesised declarative memory may be similarly impaired by doxycycline as fear and spatial memory. Therefore, in this study we investigate the potential diminishing effects of doxycycline on consolidation of declarative memory in healthy humans. Additionally, to test for effect specificity we assessed motor memory, sustained attention, and processing speed. We administered a neuropsychological test battery in three independent randomized placebo-controlled double-blind trials (RCTs), in which healthy young volunteers (total N = 252) either received a single oral dose doxycycline (200 mg, n = 126) or placebo (n = 126) in a between-subject design. We found no evidence for a detrimental effect of doxycycline on declarative memory; instead, doxycycline improved declarative learning (p-value=0.022, Cohen's d=0.15) and memory consolidation (p=0.040, d=0.26). Contrarily, doxycycline slightly reduced motor learning (p=0.001, d=0.10) but subtly strengthened long-term motor memory (p=0.001, d=0.10). These results suggest that doxycycline can improve declarative learning and memory without having long term negative effects on other cognitive domains in healthy humans. Our results give hope to further investigate doxycycline in neuroprotective treatment applications.
Collapse
Affiliation(s)
- Jelena M Wehrli
- Department of Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Lenggstrasse 31, Zurich 8032, Switzerland.
| | - Yanfang Xia
- Department of Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Lenggstrasse 31, Zurich 8032, Switzerland
| | - Laura Meister
- Department of Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Lenggstrasse 31, Zurich 8032, Switzerland
| | - Sarrina Tursunova
- Department of Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Lenggstrasse 31, Zurich 8032, Switzerland
| | - Birgit Kleim
- Department of Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Lenggstrasse 31, Zurich 8032, Switzerland
| | - Dominik R Bach
- Department of Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Lenggstrasse 31, Zurich 8032, Switzerland; University of Bonn, Transdisciplinary Research Area "Life and Health", Hertz Chair for Artificial Intelligence and Neuroscience, Bonn, Germany
| | - Boris B Quednow
- Experimental Pharmacopsychology and Psychological Addiction Research, Department of Adult Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, Joint Center of University of Zurich and Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| |
Collapse
|
43
|
Gruol DL. The Neuroimmune System and the Cerebellum. CEREBELLUM (LONDON, ENGLAND) 2024; 23:2511-2537. [PMID: 37950146 PMCID: PMC11585519 DOI: 10.1007/s12311-023-01624-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/20/2023] [Indexed: 11/12/2023]
Abstract
The recognition that there is an innate immune system of the brain, referred to as the neuroimmune system, that preforms many functions comparable to that of the peripheral immune system is a relatively new concept and much is yet to be learned. The main cellular components of the neuroimmune system are the glial cells of the brain, primarily microglia and astrocytes. These cell types preform many functions through secretion of signaling factors initially known as immune factors but referred to as neuroimmune factors when produced by cells of the brain. The immune functions of glial cells play critical roles in the healthy brain to maintain homeostasis that is essential for normal brain function, to establish cytoarchitecture of the brain during development, and, in pathological conditions, to minimize the detrimental effects of disease and injury and promote repair of brain structure and function. However, dysregulation of this system can occur resulting in actions that exacerbate or perpetuate the detrimental effects of disease or injury. The neuroimmune system extends throughout all brain regions, but attention to the cerebellar system has lagged that of other brain regions and information is limited on this topic. This article is meant to provide a brief introduction to the cellular and molecular components of the brain immune system, its functions, and what is known about its role in the cerebellum. The majority of this information comes from studies of animal models and pathological conditions, where upregulation of the system facilitates investigation of its actions.
Collapse
Affiliation(s)
- Donna L Gruol
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
44
|
Ji Y, Ma Y, Ma Y, Wang Y, Zhao X, Xu L, Ge S. An Amino Acids and Dipeptide Injection Inhibits the TNF-α/HMGB1 Inflammatory Signaling Pathway to Reduce Pyroptosis and M1 Microglial Polarization in POCD Mice: the Gut to the Brain. Mol Neurobiol 2024; 61:10097-10114. [PMID: 38700653 DOI: 10.1007/s12035-024-04209-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 04/27/2024] [Indexed: 11/24/2024]
Abstract
Peripheral surgery-induced neural inflammation is a key pathogenic mechanism of postoperative cognitive dysfunction (POCD). However, the mechanism underlying neuroinflammation and associated neural injury remains elusive. Surgery itself can lead to gut damage, and the occurrence of POCD is accompanied by high levels of TNF-α in the serum and blood‒brain barrier (BBB) damage. Reductions in stress, inflammation and protein loss have been emphasized as strategies for enhanced recovery after surgery (ERAS). We designed an amino acids and dipeptide (AAD) formula for injection that could provide intestinal protection during surgery. Through the intraoperative infusion of AAD based on the ERAS concept, we aimed to explore the effect of AAD injection on POCD and its underlying mechanism from the gut to the brain. Here, we observed that AAD injection ameliorated neural injury in POCD, in addition to restoring the function of the intestinal barrier and BBB. We also found that TNF-α levels decreased in the ileum, blood and hippocampus. Intestinal barrier protectors and TNF-α inhibitors also alleviated neural damage. AAD injection treatment decreased HMGB1 production, pyroptosis, and M1 microglial polarization and increased M2 polarization. In vitro, AAD injection protected the impaired gut barrier and decreased TNF-α production, alleviating damage to the BBB by stimulating cytokine transport in the body. HMGB1 and Caspase-1 inhibitors decreased pyroptosis and M1 microglial polarization and increased M2 polarization to protect TNF-α-stimulated microglia in vitro. Collectively, these findings suggest that the gut barrier-TNF-α-BBB-HMGB1-Caspase-1 inflammasome-pyroptosis-M1 microglia pathway is a novel mechanism of POCD related to the gut-brain axis and that intraoperative AAD infusion is a potential treatment for POCD.
Collapse
Affiliation(s)
- Yelong Ji
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Yuanyuan Ma
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Yimei Ma
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Ying Wang
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Xining Zhao
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Li Xu
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Shengjin Ge
- Department of Anaesthesia, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
45
|
Aktar S, Kato A, Toda K, Takahashi S, Maeda-Yamamoto M, Ferdousi F, Isoda H. Transcriptomic evidence of black soybean ethanolic extract and 2-aminobutyric acid in suppressing neuroinflammation and enhancing synaptic transmission. Biomed Pharmacother 2024; 181:117633. [PMID: 39488055 DOI: 10.1016/j.biopha.2024.117633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
INTRODUCTION Recently, the awareness of the beneficial utilization of natural bioactive compounds in treating neuroinflammation has gained particular attention. We aimed to understand the anti-neuroinflammatory effect of black soybeans (Glycine max (L.) Merr) ethanolic extract (BBEE) and its bioactive compound, 2-aminobutyric acid (2-AB), against LPS-induced SH-SY5Y cells. METHOD Cell viability and the optimum therapeutic dose were confirmed by MTT assay. We conducted a whole-transcriptomic analysis of BBEE and 2-AB in LPS-induced SH-SY5Y cells using microarray normalized with SST-RMA. DEGs were selected based on p-value < 0.05 and fold change > 2, and validated by RT-qPCR and immunocytochemical analyses. RESULTS We found that both BBEE and 2-AB down-regulated the expression of inflammatory cytokines IL6 and TNFA under LPS-induced conditions. This was also observed in the microarray data, showing downregulation of several inflammatory pathways, such as NF-kB, and IL6-JAK/STAT3-signaling pathways. In contrast, it upregulated the expression of CALML3, GRIN2, and GRIA2 gene expressions, which influence the AMPK and CAMK2 signaling pathways, indicating the potential of BBEE in neurotransmission and synaptic function. Also, immunofluorescence analysis revealed that 2-AB treatment significantly increased PSD-95 and Ca2+ levels, suggesting its effect on synaptic transmission essential for brain function. CONCLUSION Our findings suggest the potential anti-neuroinflammatory effects of BBEE and 2-AB, which may offer therapeutic and preventive benefits in mitigating neurological disorders. Given that BB is widely consumed in many Asian countries, our study may encourage its incorporation into the daily diet to slow inflammation-induced neurodegenerative disorders, reduce age-related cognitive decline, and enhance overall brain function.
Collapse
Affiliation(s)
- Sharmin Aktar
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba 305-8572, Japan
| | - Ayumi Kato
- Tsukuba Life Science Innovation (T-LSI) Program, University of Tsukuba, Japan
| | - Kyoko Toda
- Research Center of Genetic Resources, National Agriculture and Food Research Organization (NARO), Japan
| | - Shinya Takahashi
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba 305-8572, Japan; Tsukuba Life Science Innovation (T-LSI) Program, University of Tsukuba, Japan; Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8572, Japan
| | - Mari Maeda-Yamamoto
- Institute of Food Research, National Agriculture and Food Research Organization (NARO), Japan
| | - Farhana Ferdousi
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba 305-8572, Japan; Tsukuba Life Science Innovation (T-LSI) Program, University of Tsukuba, Japan; Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8572, Japan
| | - Hiroko Isoda
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba 305-8572, Japan; Tsukuba Life Science Innovation (T-LSI) Program, University of Tsukuba, Japan; Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8572, Japan.
| |
Collapse
|
46
|
Yang Y, Wu J, Jia L, Feng S, Qi Z, Yu H, Wu Y, Wang S. Berberine modulates microglial polarization by activating TYROBP in Alzheimer's disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156237. [PMID: 39566407 DOI: 10.1016/j.phymed.2024.156237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/29/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Characterized by β-amyloid (Aβ) plaques, neurofibrillary tangles, and aberrant neuroinflammation in the brain, Alzheimer's disease (AD) is the most common neurodegenerative disease. Microglial polarization is a subtle mechanism which maintains immunological homeostasis and has emerged as a putative therapeutic to combat AD. Berberine (BBR) is a natural alkaloid compound with multiple pharmacological effects, and has shown considerable therapeutic potential against inflammatory disorders. However, BBR functions and underlying mechanisms in neuroinflammation remain unclear. PURPOSE To examine BBR pharmacological effects and mechanisms in neuroinflammation with a view to treating AD. METHODS BBR effects on cognitive performance in 5 × FAD mice were assessed using open field, Y-maze, and Morris Water Maze (MWM) tests. Neuroinflammation-related markers and Aβ pathology were examined in brain sections from mice. Transcriptomic analyses of hippocampus tissues were also conducted. Microglial BV2 cells were also used to verify potential BBR mechanisms in neuroinflammation and microglial polarization. RESULTS BBR improved cognitive performance, reduced amyloid pathology, and alleviated aberrant neuroinflammation in an AD mouse model. BBR induced microglial polarization to an M2-like phenotype, which was manifested by lowered and elevated proinflammatory and anti-inflammatory cytokine production, respectively, improved microglial uptake and Aβ clearance. Mechanistically, BBR directly interacted with TYROBP and promoted its activation by stabilizing TYROBP oligomerization. TYROBP knockdown aggravated M1-like polarization and pro-inflammatory gene expression in microglial cells in the presence of lipopolysaccharide (LPS)+Aβ, while blocked microglial M2-like polarization benefited from BBR administration. CONCLUSIONS BBR modulated neuroinflammation by regulating microglial polarization via TYROBP activation. Our study provided new insight into BBR pharmacological actions in regulating microglial homeostasis and combating AD.
Collapse
Affiliation(s)
- Yu Yang
- Shandong Key Laboratory of Psychiatric and Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, 272013, Shandong, China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, 272013, Shandong, China
| | - Jiwen Wu
- Shandong Key Laboratory of Psychiatric and Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, 272013, Shandong, China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, 272013, Shandong, China
| | - Luping Jia
- Shandong Key Laboratory of Psychiatric and Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, 272013, Shandong, China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, 272013, Shandong, China
| | - Shicheng Feng
- Shandong Key Laboratory of Psychiatric and Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, 272013, Shandong, China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, 272013, Shandong, China
| | - Zihan Qi
- Shandong Key Laboratory of Psychiatric and Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, 272013, Shandong, China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, 272013, Shandong, China
| | - Hao Yu
- Shandong Key Laboratory of Psychiatric and Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, 272013, Shandong, China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, 272013, Shandong, China
| | - Yili Wu
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, Zhejiang Provincial Clinical Research Center for Mental Health, School of Mental Health, Institute of Aging, Wenzhou Medical University, Wenzhou, 325000, China; Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, 325000, China.
| | - Shuai Wang
- Shandong Key Laboratory of Psychiatric and Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, 272013, Shandong, China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, 272013, Shandong, China.
| |
Collapse
|
47
|
Tu H, Yuan L, Ni B, Lin Y, Wang K. Siglecs-mediated immune regulation in neurological disorders. Pharmacol Res 2024; 210:107531. [PMID: 39615617 DOI: 10.1016/j.phrs.2024.107531] [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: 07/25/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024]
Abstract
The surfaces of various immune cells are rich in glycan chains, including the sialic-acid-binding immunoglobulin-like lectins (Siglecs) family. As an emerging glyco-immune checkpoint, Siglecs have the ability to bind and interact with various glycoproteins, thereby eliciting a series of downstream reactions to modulate the immune response. The impact of Siglecs has been extensively studied in tumor immunotherapy. However, research in neurological disorders and neurological diseases is very limited, and therapeutic options involving Siglecs need further exploration. Siglecs play a crucial role in the development, homeostasis, and repair processes of the nervous system, especially in degenerative diseases. This review summarizes studies on the immunomodulatory role mediated by Siglecs expressed on different immune cells in various neurological disorders, elucidates how dysregulated sialic acid contributes to several psychiatric disorders, and discusses the progress and limitations of research on the treatment of neurological disorders.
Collapse
Affiliation(s)
- Huifang Tu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Limei Yuan
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Bo Ni
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yufeng Lin
- Department of Neurology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300190, China.
| | - Kaiyuan Wang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| |
Collapse
|
48
|
Tan Z, Xia R, Zhao X, Yang Z, Liu H, Wang W. Potential key pathophysiological participant and treatment target in autism spectrum disorder: Microglia. Mol Cell Neurosci 2024; 131:103980. [PMID: 39580060 DOI: 10.1016/j.mcn.2024.103980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/28/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024] Open
Abstract
Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders characterized by social and communication deficits, as well as restricted or repetitive behaviors or interests. Although the etiology of ASD remains unclear, there is abundant evidence suggesting that microglial dysfunction is likely to be a significant factor in the pathophysiology of ASD. Microglia, the primary innate immune cells in the central nervous system (CNS), play a crucial role in brain development and homeostasis. Recently, numerous studies have shown that microglia in ASD models display various abnormalities including morphology, function, cellular interactions, genetic and epigenetic factors, as well as the expression of receptors, transcription factors, and cytokines. They impact normal neural development through various mechanisms contributing to ASD, such as neuroinflammation, and alterations in synaptic formation and pruning. The focus of this review is on recent studies regarding microglial abnormalities in ASD and their effects on the onset and progression of ASD at both cellular and molecular levels. It can provide insight into the specific contribution of microglia to ASD pathogenesis and help in designing potential therapeutic and preventative strategies targeting microglia.
Collapse
Affiliation(s)
- Zehua Tan
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ruixin Xia
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xin Zhao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zile Yang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Haiying Liu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
49
|
Zong T, Li N, Han F, Liu J, Deng M, Li V, Zhang M, Zhou Y, Yu M. Microglial depletion rescues spatial memory impairment caused by LPS administration in adult mice. PeerJ 2024; 12:e18552. [PMID: 39559328 PMCID: PMC11572354 DOI: 10.7717/peerj.18552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/29/2024] [Indexed: 11/20/2024] Open
Abstract
Recent studies have highlighted the importance of microglia, the resident macrophages in the brain, in regulating cognitive functions such as learning and memory in both healthy and diseased states. However, there are conflicting results and the underlying mechanisms are not fully understood. In this study, we examined the effect of depleting adult microglia on spatial learning and memory under both physiological conditions and lipopolysaccharide (LPS)-induced neuroinflammation. Our results revealed that microglial depletion by PLX5622 caused mild spatial memory impairment in mice under physiological conditions; however, it prevented memory deficits induced by systemic LPS insult. Inactivating microglia through minocycline administration replicated the protective effect of microglial depletion on LPS-induced memory impairment. Furthermore, our study showed that PLX5622 treatment suppressed LPS-induced neuroinflammation, microglial activation, and synaptic dysfunction. These results strengthen the evidence for the involvement of microglial immunoactivation in LPS-induced synaptic and cognitive malfunctions. They also suggest that targeting microglia may be a potential approach to treating neuroinflammation-associated cognitive dysfunction seen in neurodegenerative diseases.
Collapse
Affiliation(s)
- Tao Zong
- Affiliated Qingdao Third People’s Hospital, Department of Otorhinolaryngology Head and Neck, Qingdao University, Qingdao, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
| | - Na Li
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
- Qingdao Binhai University, Qingdao, Shandong, China
| | - Fubing Han
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, China, China
| | - Junru Liu
- Department of Neurology, Affiliated Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, China, China
| | - Mingru Deng
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
- Department of Neurology, Affiliated Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, China, China
| | - Vincent Li
- Beverly Hills High School, Unaffiliated, Beverly Hills, California, United States
| | - Meng Zhang
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
| | - Yu Zhou
- Affiliated Qingdao Third People’s Hospital, Department of Otorhinolaryngology Head and Neck, Qingdao University, Qingdao, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
- Department of Neurology, Affiliated Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, China, China
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Ming Yu
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
50
|
Oue K, Yamawaki Y, Ouhara K, Imado E, Tamura T, Doi M, Shimizu Y, Yoshida M, Mizuno N, Morioka N, Kanematsu T, Irifune M, Ago Y. Oral administration of Porphyromonas gingivalis to mice with diet-induced obesity impairs cognitive function associated with microglial activation in the brain. J Oral Microbiol 2024; 16:2419155. [PMID: 39553478 PMCID: PMC11565673 DOI: 10.1080/20002297.2024.2419155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 01/04/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024] Open
Abstract
Objective Both periodontal disease and obesity are risk factors for dementia, but their links to 1brain function remain unclear. In this study, we examined the effects of oral infection with a periodontal pathogen on cognitive function in a mouse model of obesity, focusing on the roles of microglia. Methods To create a mouse model of diet-induced obesity and periodontitis, male C57BL/6 J mice were first fed a high-fat diet containing 60% lipid calories for 18 weeks, beginning at 12 weeks of age, to achieve diet-induced obesity. Then, Porphyromonas gingivalis administration in the oral cavity twice weekly for 6 weeks was performed to induce periodontitis in obese mice. Results Obese mice orally exposed to P. gingivalis showed cognitive impairment in the novel object recognition test. Increased expression levels of inflammatory cytokines (e.g. interleukin-1β and tumor necrosis factor-α) were observed in the hippocampus of P. gingivalis-treated obese mice. Immunohistochemical analysis revealed that microglia cell body size was increased in the hippocampus and prefrontal cortex of P. gingivalis-treated obese mice, indicating microglial activation. Furthermore, depletion of microglia by PLX3397, a colony-stimulating factor 1 receptor inhibitor, ameliorated cognitive dysfunction. Conclusion These results suggest that microglia mediate periodontal infection-induced cognitive dysfunction in obesity.
Collapse
Affiliation(s)
- Kana Oue
- Department of Dental Anesthesiology, Division of Oral and Maxillofacial Surgery and Oral Medicine, Hiroshima University Hospital, Hiroshima, Japan
| | - Yosuke Yamawaki
- Department of Advanced Pharmacology, Daiichi University of Pharmacy, Fukuoka, Japan
| | - Kazuhisa Ouhara
- Department of Periodontal Medicine, Division of Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Eiji Imado
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tetsuya Tamura
- Department of Periodontal Medicine, Division of Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mitsuru Doi
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yoshitaka Shimizu
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mitsuhiro Yoshida
- Department of Dental Anesthesiology, Division of Oral and Maxillofacial Surgery and Oral Medicine, Hiroshima University Hospital, Hiroshima, Japan
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Noriyoshi Mizuno
- Department of Periodontal Medicine, Division of Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Norimitsu Morioka
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takashi Kanematsu
- Department of Cell Biology, Aging Science, and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Masahiro Irifune
- Department of Dental Anesthesiology, Division of Oral and Maxillofacial Surgery and Oral Medicine, Hiroshima University Hospital, Hiroshima, Japan
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|