1
|
Conejero I, Chea M, Courtet P, Bouvier S, Pereira F. Alterations in the Blood-Brain Barrier in Mood Disorders and Neurodegenerative Diseases. Semin Thromb Hemost 2025; 51:457-464. [PMID: 40058364 DOI: 10.1055/s-0045-1804893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Depressive disorders and suicidal behaviors represent major causes of health loss. Modifications of brain microvasculature, and specifically alterations of the blood-brain barrier have been supposed to participate in the vulnerability to those disorders along with cognitive impairment, especially in the older adults. In this article, we addressed evidence linking blood-brain barrier impairments with mood disorders and suicide. Secondly, we investigated their relationship with depression in old age, and with neurodegenerative processes. Particular attention was drawn toward the potential interactions between the coagulation processes and the blood-brain barrier dysfunctions, as innovative treatment strategies may emerge from research in those fields. Overall, the studies reviewed highlight the implication of multiple dysfunctions of the blood-brain barrier in mood disorders and suicide. Impairments of the blood-brain barrier show relationships with altered expression of endothelial cell junction proteins. These modifications also implicate receptors of the extracellular matrix, the vascular endothelial growth factor, changes in perivascular astrocytes, and has links with local and systemic inflammatory processes. Dysfunctions of the blood-brain barrier underly chronic stress and participate in psychiatric diathesis in old age. In addition, we outline that coagulation processes are likely to interact with the blood-brain barrier and further contribute to neurodegenerative disorders. In conclusion, new pathophysiological models offer perspectives toward detecting new biomarkers in mood disorders and suicide. In parallel, these models open avenues for developing innovative therapeutic agents, although further considering their potential risks and eventual benefits is needed.
Collapse
Affiliation(s)
- Ismael Conejero
- Department of Psychiatry, CHU Nîmes, University of Montpellier, Nîmes, France
| | - Mathias Chea
- Department of Hematology, CHU Nîmes, Nîmes, France
- UMR 1318 INSERM-Université de Montpellier IDESP, France
- Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France
| | - Philippe Courtet
- Institute of Functional Genomics (IGF), University of Montpellier, Montpellier, France
- Department of Emergency Psychiatry & Acute Care, CHU Montpellier, University of Montpellier, Montpellier, France
| | - Sylvie Bouvier
- Department of Hematology, CHU Nîmes, Nîmes, France
- UMR 1318 INSERM-Université de Montpellier IDESP, France
- Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France
| | - Fabricio Pereira
- Department of Radiology and Medical Imaging, CHU Nîmes, Nîmes, France
- MIPA, University of Nîmes, Nîmes, France
| |
Collapse
|
2
|
Wang S, Guo L, Wang C. Potential Risk Factors of Susceptibility to Recurrent Depression. Brain Res Bull 2025:111374. [PMID: 40389099 DOI: 10.1016/j.brainresbull.2025.111374] [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: 02/01/2025] [Revised: 04/10/2025] [Accepted: 05/05/2025] [Indexed: 05/21/2025]
Abstract
BACKGROUND Major depressive disorder (MDD) is a highly prevalent and recurrent neuropsychiatric disorder associated with alterations in the BicC family RNA binding protein 1 (BICC1). However, the potential risk factors that regulate BICC1 and affect susceptibility to recurrent depression remain unclear. METHODS Herein, we firstly tested the heat shock protein 90 (HSP90), hypoxia-inducible factor 1-alpha (HIF1α), and BICC1 in the serum of the patients that were in first-episode or recurrent depression, as well as their controls. Then, through re-exposure to chronic unpredictable mild stress (CUMS) in mice, an animal model of recurrent depression was assessed. And the expression of HSP90, HIF1α, and BICC1 in the prefrontal cortex (PFC) were analyzed. RESULTS We found that HSP90, HIF1α, and BICC1 were significantly increased in the serum of depressed patients, especially in those with recurrent depression, indicating that these molecules may serve as specific pathogenetic risk factors for depression, especially depression recurrence. In addition, the recurrent depression mice model was found to be accompanied by a significant increase in expression of HSP90, HIF1α and BICC1 in the prefrontal cortex (PFC). CONCLUSIONS The current study identified HSP90, HIF1α, and BICC1 as novel potential risk factors that affect susceptibility to recurrent depression.
Collapse
Affiliation(s)
- Shuzhuo Wang
- School of Basic Medical Science, Health Center, Ningbo University, Ningbo, Zhejiang 315211, PR China; Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang 315201, China
| | - Lei Guo
- School of Basic Medical Science, Health Center, Ningbo University, Ningbo, Zhejiang 315211, PR China; Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang 315201, China.
| | - Chuang Wang
- School of Basic Medical Science, Health Center, Ningbo University, Ningbo, Zhejiang 315211, PR China; Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang 315201, China.
| |
Collapse
|
3
|
Geng Z, Peng F, Cheng Z, Su J, Song J, Han X, Li R, Li X, Cui R, Li B. Astrocytic FABP7 Alleviates Depression-Like Behaviors of Chronic Unpredictable Mild Stress Mice by Regulating Neuroinflammation and Hippocampal Spinogenesis. FASEB J 2025; 39:e70606. [PMID: 40331773 PMCID: PMC12057550 DOI: 10.1096/fj.202403417rr] [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: 12/29/2024] [Revised: 03/28/2025] [Accepted: 04/28/2025] [Indexed: 05/08/2025]
Abstract
Fatty acid binding protein 7 (FABP7) is prominently expressed in astrocytes and is a critical regulator of inflammatory responses. Accumulating evidence suggests that FABP7 is crucial in neuropsychological disease through the modulation of spinogenesis. Nonetheless, the impact of FABP7 on depressive disorders and the underlying mechanisms is not fully understood. Here, we investigated the antidepressant properties of FABP7 using the chronic unpredictable mild stress (CUMS)-induced model of depression and possible mechanisms. Our results revealed that depressive-like behavior induced by CUMS was associated with decreased levels of FABP7 protein in the hippocampus (HP). Furthermore, the overexpression of FABP7 in the HP mitigated the depressive-like behavior and increased the expression of its downstream target caveolin-1 (Cav-1). FABP7 overexpression in the HP specifically regulates the expression of the astrocyte marker protein GFAP, as well as the blood-brain barrier (BBB)-associated proteins AQP4, CLDN-5, occludin, and LRP1. Notably, the CUMS-induced upregulation of the pro-inflammatory factors IL-1β and IL-6 was also significantly reversed by FABP7 overexpression in the HP. This intervention also led to increased levels of postsynaptic proteins, including PSD95 and GluA1, as well as an increase in brain-derived neurotrophic factor (BDNF) and enhanced neuronal dendritic spine density. The findings indicate that FABP7 exerts antidepressant-like properties by inhibiting inflammation, regulating spinogenesis, and modulating BBB-related proteins.
Collapse
Affiliation(s)
- Zihui Geng
- Jilin Provinicial Key Laoratory on Molecular and Chemical GeneticSencond Hospital of Jilin UniversityChangchunPeople's Republic of China
- Engineering Lab on Screening of Antidepressant DrugsJilin Province Development and Reform CommissionChangchunPeople's Republic of China
| | - Fanzhen Peng
- Jilin Provinicial Key Laoratory on Molecular and Chemical GeneticSencond Hospital of Jilin UniversityChangchunPeople's Republic of China
- Engineering Lab on Screening of Antidepressant DrugsJilin Province Development and Reform CommissionChangchunPeople's Republic of China
| | - Ziqian Cheng
- Jilin Provinicial Key Laoratory on Molecular and Chemical GeneticSencond Hospital of Jilin UniversityChangchunPeople's Republic of China
- Engineering Lab on Screening of Antidepressant DrugsJilin Province Development and Reform CommissionChangchunPeople's Republic of China
| | - Jingyun Su
- Jilin Provinicial Key Laoratory on Molecular and Chemical GeneticSencond Hospital of Jilin UniversityChangchunPeople's Republic of China
- Engineering Lab on Screening of Antidepressant DrugsJilin Province Development and Reform CommissionChangchunPeople's Republic of China
| | - Jinfang Song
- Jilin Provinicial Key Laoratory on Molecular and Chemical GeneticSencond Hospital of Jilin UniversityChangchunPeople's Republic of China
- Engineering Lab on Screening of Antidepressant DrugsJilin Province Development and Reform CommissionChangchunPeople's Republic of China
| | - Xu Han
- Jilin Provinicial Key Laoratory on Molecular and Chemical GeneticSencond Hospital of Jilin UniversityChangchunPeople's Republic of China
- Engineering Lab on Screening of Antidepressant DrugsJilin Province Development and Reform CommissionChangchunPeople's Republic of China
| | - Runxin Li
- Jilin Provinicial Key Laoratory on Molecular and Chemical GeneticSencond Hospital of Jilin UniversityChangchunPeople's Republic of China
- Engineering Lab on Screening of Antidepressant DrugsJilin Province Development and Reform CommissionChangchunPeople's Republic of China
| | - Xin Li
- Jilin Provinicial Key Laoratory on Molecular and Chemical GeneticSencond Hospital of Jilin UniversityChangchunPeople's Republic of China
- Engineering Lab on Screening of Antidepressant DrugsJilin Province Development and Reform CommissionChangchunPeople's Republic of China
| | - Ranji Cui
- Jilin Provinicial Key Laoratory on Molecular and Chemical GeneticSencond Hospital of Jilin UniversityChangchunPeople's Republic of China
- Engineering Lab on Screening of Antidepressant DrugsJilin Province Development and Reform CommissionChangchunPeople's Republic of China
| | - Bingjin Li
- Jilin Provinicial Key Laoratory on Molecular and Chemical GeneticSencond Hospital of Jilin UniversityChangchunPeople's Republic of China
- Engineering Lab on Screening of Antidepressant DrugsJilin Province Development and Reform CommissionChangchunPeople's Republic of China
| |
Collapse
|
4
|
Dai A, Liu X, Chen Y, Wang Y, Qi H, Zeng Y, Li J. Co-exposure to ozone and polystyrene nanoplastic exacerbates cognitive impairment and anxiety-like behavior by regulating neuronal pyroptosis in mice. ENVIRONMENT INTERNATIONAL 2025; 199:109501. [PMID: 40288286 DOI: 10.1016/j.envint.2025.109501] [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: 01/14/2025] [Revised: 04/01/2025] [Accepted: 04/25/2025] [Indexed: 04/29/2025]
Abstract
Ozone (O3) and nanoplastics (NPs) are pervasive environmental pollutants that frequently co-occur in our heavily industrialized era. While it has been documented that exposure to O3 or NPs individually has neurotoxic effects, studies investigating their combined impact and the hazardous mechanisms resulting from co-exposure are limited. In this study, we established a mouse model co-exposure to polystyrene nanoparticles (PS-NPs) and O3, focusing on the prefrontal cortex (PFC), a brain region crucial for cognition and emotion. We examined the effects of O3 and PS-NPs on behavioral changes related to learning, memory, and anxiety, employing transcriptome sequencing alongside molecular and histopathological methods. Our findings indicate that combined exposure to O3 and PS-NPs disrupts the integrity of the blood-brain barrier, reducing Claudin 5 expression and leading to increased accumulation of PS-NPs in the PFC. Transcriptome sequencing demonstrated the involvement of the p38 mitogen-activated protein kinase (p38 MAPK) signaling pathway and oxidative stress in the pathological changes observed in the PFC. Through immunohistochemical and immunofluorescence analysis, we observed enhanced microglial activation, which correlates with increased production of inflammatory factors. Additionally, western blot and immunofluorescence co-labeling analyses revealed elevated expression levels of GSDMD-N, caspase-1, IL-1β, and IL-18 proteins, which are associated with neuronal pyroptosis. Finally, immunofluorescence co-labeling confirmed that the activation of the p38 MAPK pathway in neurons is involved in co-exposure-induced pyroptosis. Meanwhile, N-Acetylcysteine (NAC), a common antioxidant, can alleviate neuroinflammation and neuronal pyroptosis in the PFC, and it rescued the cognitive deficits and anxiety-like behaviors observed in the co-exposed mice. Our study illustrates that co-exposure to O3 and NPs can aggravate damage to the blood-brain barrier and elevate oxidative stress levels in the PFC, thereby increasing the occurrence of neuroinflammation and may mediate neuronal pyroptosis through activation of the p38 MAPK pathway, ultimately contributing to neurobehavioral toxicity.
Collapse
Affiliation(s)
- Anqi Dai
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xudong Liu
- Department of Food Science and Engineering, Moutai Institute, Renhuai 564507, China
| | - Yinuo Chen
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Yougang Wang
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Haomin Qi
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Yan Zeng
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan, China.
| | - Jinquan Li
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan, China; Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China.
| |
Collapse
|
5
|
Jiménez-Jiménez V, Sánchez-Cabo F, Schwartz MA, Sánchez-Álvarez M, del Pozo MÁ. A Survey for Human Tissue-Level Determinants of CAV1 Regulation and Function. Int J Mol Sci 2025; 26:3789. [PMID: 40332409 PMCID: PMC12027754 DOI: 10.3390/ijms26083789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/02/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
CAV1 is a protein-coding gene linked to several disorders, including cancer, lipodystrophy, and cardiovascular diseases. While its ability to respond to various mechanical and metabolic stimuli has been documented, a comprehensive understanding of its physiological regulation in humans is lacking. We leveraged the comprehensiveness of human post-mortem tissue data from the Genotype-Tissue Expression (GTEx) consortium, systematically exploring the sources of variability in CAV1 transcriptional levels using extensive bulk and single-nuclei RNA-seq datasets. This human-centric approach, avoiding inter-species comparisons, constitutes a unique resource to explore CAV1 regulation within the complexity of human tissues. Notably, cell type proportion was identified as a major determinant of CAV1 transcription levels across tissues. Donor physiological conditions, including disease states and end-of-life circumstances, also exhibited a tissue-specific influence. Among primary upstream regulators associated with CAV1, chromatin modifiers stood out, especially SMARCA2, which showed a positive correlation across tissues, and PRC2 complexes, which exhibited tissue-specific correlation. Upstream regulatory networks determining CAV1 levels are also enriched for annotations such as mechanobiology (e.g., TEAD4), immunity (e.g., RELA and STAT3), and metabolism (e.g., MYC and NRF1). A remarkable observation was a strong correlation between CAV1 and the relative infiltration of immune cells across tissues, supporting a potential role for CAV1 as a marker and driver of tissue immune infiltration.
Collapse
Affiliation(s)
- Víctor Jiménez-Jiménez
- Mechanoadaptation & Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain;
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain;
- Department of Quantitative Methods, CUNEF Universidad, 28040 Madrid, Spain
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain;
| | - Martin A. Schwartz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06510, USA;
- Department of Cell Biology, Yale University, New Haven, CT 06510, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| | - Miguel Sánchez-Álvarez
- Mechanoadaptation & Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain;
- Cell Compartmentalization, Homeostasis and Inflammation Lab, Department of Metabolic and Immunity Diseases, Instituto de Investigaciones Biomédicas Sols-Morreale, 28029 Madrid, Spain
| | - Miguel Ángel del Pozo
- Mechanoadaptation & Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain;
| |
Collapse
|
6
|
Gryka-Marton M, Grabowska AD, Szukiewicz D. Breaking the Barrier: The Role of Proinflammatory Cytokines in BBB Dysfunction. Int J Mol Sci 2025; 26:3532. [PMID: 40331982 PMCID: PMC12026921 DOI: 10.3390/ijms26083532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/02/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
The BBB is created by a special system of brain microvascular endothelial cells (BMECs), pericytes (PCs), the capillary basement membrane, and the terminal branches ("end-feet") of astrocytes (ACs). The key function of the BBB is to protect the central nervous system (CNS) from potentially harmful/toxic substances in the bloodstream by selectively controlling the entry of cells and molecules, including nutrients and components of the immune system. The loss of BBB integrity in response to neuroinflammation, as manifested by an increase in permeability, depends predominantly on the activity of proinflammatory cytokines. However, the pathomechanism of structural and functional changes in the BBB under the influence of individual cytokines is still poorly understood. This review summarizes the current state of knowledge on this topic, which is important from both pathophysiological and therapeutic points of view. The structures and functions of all components of the BBB are reviewed, with emphasis given to differences between this and other locations of the circulatory system. The protein composition of the interendothelial tight junctions in the context of regulating BBB permeability is presented, as is the role of pericyte-BMEC interactions in the exchange of metabolites, ions, and nucleic acids. Finally, the documented actions of proinflammatory cytokines within the BBB are discussed.
Collapse
Affiliation(s)
| | | | - Dariusz Szukiewicz
- Laboratory of the Blood–Brain Barrier, Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (M.G.-M.); (A.D.G.)
| |
Collapse
|
7
|
Bollinger JL, Johnsamuel S, Vollmer LL, Kuhn AM, Wohleb ES. Stress-induced dysfunction of neurovascular astrocytes in the prefrontal cortex contributes to sex-dependent deficits in cognition and behavior. Mol Psychiatry 2025:10.1038/s41380-025-02993-3. [PMID: 40185903 DOI: 10.1038/s41380-025-02993-3] [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: 04/26/2024] [Revised: 03/14/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
Astrocytes form an integral component of the neurovascular unit, ensheathing brain blood vessels with endfeet high in aquaporin-4 (AQP4) expression. These AQP4-rich endfeet facilitate interaction between the vascular endothelium, astrocytes, and neurons, and help stabilize vascular morphology. Studies using preclinical models of psychological stress and post-mortem tissue from patients with major depressive disorder (MDD) have reported reductions in AQP4, loss of astrocytic structures, and vascular impairment in the prefrontal cortex (PFC). Though compelling, the role of AQP4 in mediating stress-induced alterations in neurovascular function and behavior remains unclear. Here, we address this, alongside potential sex differences in chronic unpredictable stress (CUS) effects on astrocyte phenotype, blood-brain barrier integrity, and behavior. CUS led to more pronounced shifts in stress-coping behavior and working memory deficits in male- as compared to female mice. Following behavioral testing, astrocytes from the frontal cortex were isolated for gene expression analyses. We found that CUS increased transcripts associated with blood vessel maintenance in males, but either had no effect on- or decreased- these transcripts in females. Furthermore, CUS caused a reduction in vascular-localized AQP4 and elevated extravasation of a small fluorescent reporter (Dextran) in the PFC in males but not females. Studies showed that knockdown of AQP4 in the PFC is sufficient to disrupt astrocyte phenotype and increase behavioral susceptibility to a sub-chronic stressor in males yet has little effect on stress susceptibility in females. Our findings provide evidence that sex-specific alterations in astrocyte phenotype and neurovascular integrity in the PFC contribute to cognitive-behavioral consequences following stress.
Collapse
Affiliation(s)
- Justin L Bollinger
- Department of Pharmacology, Physiology, & Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Shobha Johnsamuel
- Department of Pharmacology, Physiology, & Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lauren L Vollmer
- Department of Pharmacology, Physiology, & Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Alexander M Kuhn
- Department of Pharmacology, Physiology, & Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology, Physiology, & Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
8
|
Sun ZW, Sun ZX, Zhao Y, Zhang L, Xie F, Wang X, Li JS, Zhou MY, Feng H, Qian LJ. Rutin ameliorates stress-induced blood‒brain barrier dysfunction and cognitive decline via the endothelial HDAC1‒Claudin-5 axis. Fluids Barriers CNS 2025; 22:35. [PMID: 40176114 PMCID: PMC11967129 DOI: 10.1186/s12987-025-00639-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/10/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Emerging evidence suggests that chronic stress compromises blood‒brain barrier (BBB) integrity by disrupting brain microvascular endothelial cells (BMECs), contributing to the development of cognitive impairments. Thus, targeting the BBB is expected to be a promising treatment strategy. The biological function of rutin has been investigated in neurological disorders; however, its regulatory role in stress-induced BBB damage and cognitive decline and the underlying mechanisms remain elusive. METHODS In a chronic unpredictable mild stress (CUMS) mouse model, a fluorescent dye assay and behavioral tests, including a novel object recognition test and Morris water maze, were performed to evaluate the protective effects of rutin on BBB integrity and cognition. The effects of rutin on BMEC function were also investigated in hCMEC/D3 cells (a human brain microvascular endothelial cell line) in vitro. Furthermore, the molecular mechanisms by which rutin restores BBB endothelium dysfunction were explored via RNA-seq, quantitative real-time PCR, western blotting, immunofluorescence and chromatin immunoprecipitation. Finally, biotinylated tumor necrosis factor-α (TNF-α) was employed to test the influence of rutin on the ability of circulating TNF-α to cross the BBB. RESULTS We identified that rutin attenuated BBB hyperpermeability and cognitive impairment caused by the 8-week CUMS procedure. Moreover, rutin promoted the proliferation, migration and angiogenesis ability of BMECs, and the integrity of the cellular monolayer through positively regulating the expression of genes involved. Furthermore, rutin impeded histone deacetylase 1 (HDAC1) recruitment and stabilized H3K27ac to increase Claudin-5 protein levels. Ultimately, normalization of the hippocampal HDAC1‒Claudin-5 axis by rutin blocked the infiltration of circulating TNF-α into the brain parenchyma and alleviated neuroinflammation. CONCLUSIONS This work establishes a protective role of rutin in regulating BMEC function and BBB integrity, and reveals that rutin is a potential drug candidate for curing chronic stress-induced cognitive deficits.
Collapse
Affiliation(s)
- Zhao-Wei Sun
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Zhao-Xin Sun
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise & Health, Tianjin University of Sport, Tianjin, 301617, China
| | - Yun Zhao
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Ling Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Fang Xie
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Xue Wang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Jin-Shan Li
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise & Health, Tianjin University of Sport, Tianjin, 301617, China
| | - Mao-Yang Zhou
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Hong Feng
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise & Health, Tianjin University of Sport, Tianjin, 301617, China.
| | - Ling-Jia Qian
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
| |
Collapse
|
9
|
Wang M, Yang J, Wang S, Gill H, Cheng H. Immunotherapy and the Tumor Microenvironment in Brain Metastases from Non-Small Cell Lung Cancer: Challenges and Future Directions. Curr Oncol 2025; 32:171. [PMID: 40136375 PMCID: PMC11941645 DOI: 10.3390/curroncol32030171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/09/2025] [Accepted: 03/15/2025] [Indexed: 03/27/2025] Open
Abstract
Brain metastases (BMs) are a relatively common and severe complication in advanced non-small cell lung cancer (NSCLC), significantly affecting patient prognosis. Metastatic tumor cells can alter the brain tumor microenvironment (TME) to promote an immunosuppressive state, characterized by reduced infiltration of tumor-infiltrating lymphocytes (TILs), diminished expression of programmed death-ligand 1 (PD-L1), and changes in other proinflammatory factors and immune cell populations. Microglia, the resident macrophages of the brain, play a pivotal role in modulating the central nervous system (CNS) microenvironment through interactions with metastatic cancer cells, astrocytes, and infiltrating T cells. The M2 phenotype of microglia contributes to immunosuppression in BM via the activation of signaling pathways such as STAT3 and PI3K-AKT-mTOR. Recent advances have enhanced our understanding of the immune landscape of BMs in NSCLC, particularly regarding immune evasion within the CNS. Current immunotherapeutic strategies, including immune checkpoint inhibitors, have shown promise for NSCLC patients with BM, demonstrating intracranial activity and manageable safety profiles. Future research is warranted to further explore the molecular and immune mechanisms underlying BM, aiming to develop more effective treatments.
Collapse
Affiliation(s)
- Meng Wang
- Department of Oncology (Medical Oncology), Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.W.)
| | - Jihua Yang
- Department of Oncology (Medical Oncology), Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.W.)
| | - Shuai Wang
- Department of Oncology (Medical Oncology), Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.W.)
| | - Harjot Gill
- Department of Pathology, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Haiying Cheng
- Department of Oncology (Medical Oncology), Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.W.)
| |
Collapse
|
10
|
Liu B, Dong K, Zhao Y, Wang X, Sun Z, Xie F, Qian L. Depletion of MGO or Its Derivatives Ameliorate CUMS-Induced Neuroinflammation. Cells 2025; 14:397. [PMID: 40136646 PMCID: PMC11941696 DOI: 10.3390/cells14060397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Advanced glycation end products (AGEs) are a series of structurally complex and harmful compounds formed through the reaction between the carbonyl group of reducing sugars (such as glucose and fructose) and the free amino groups of proteins, lipids, or nucleic acids. Excessive accumulation of AGEs in the body can trigger oxidative stress, induce inflammatory responses, and contribute to the development of diabetes, atherosclerosis, and neurological disorders. Within the category of dicarbonyl compounds, methylglyoxal (MGO)-a byproduct resulting from glucose degradation-serves as a pivotal precursor in the formation of AGEs and the induction of neurotoxicity. Specifically, AGEs generated from MGO display significant cytotoxicity toward cells in the central nervous system. Therefore, we aimed to investigate the role of MGO-AGEs in neuroinflammation mediated by CUMS. Interestingly, we found that the overexpression of glyoxalase 1 (GLO1) reduced the levels of MGO in corticosterone-treated microglia, thereby alleviating the inflammatory response. Furthermore, overexpression of GLO1 in the hippocampus of chronically stressed mice reduced MGO levels, mitigating CUMS-induced neuroinflammation and cognitive impairment. Additionally, when using the receptor for advanced glycation end products (RAGE) inhibitor FPS-ZM1 in primary microglia cells, we observed that despite corticosterone-induced elevation of MGO, no significant inflammatory response occurred. This suggests that RAGE clearance can reduce MGO-AGE-mediated neurotoxicity. Subsequently, we used FPS-ZM1 to treat chronically stressed mice and found that it significantly ameliorated neuroinflammation and cognitive dysfunction. These results suggest that targeting MGO metabolism could serve as a therapeutic approach to manage neuroinflammation in stress-related mental disorders.
Collapse
Affiliation(s)
- Bing Liu
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100191, China; (B.L.); (K.D.); (Y.Z.); (X.W.); (Z.S.)
| | - Ke Dong
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100191, China; (B.L.); (K.D.); (Y.Z.); (X.W.); (Z.S.)
- School of Medicine, South China University of Technology, Guangzhou 511442, China
| | - Yun Zhao
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100191, China; (B.L.); (K.D.); (Y.Z.); (X.W.); (Z.S.)
| | - Xue Wang
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100191, China; (B.L.); (K.D.); (Y.Z.); (X.W.); (Z.S.)
| | - Zhaowei Sun
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100191, China; (B.L.); (K.D.); (Y.Z.); (X.W.); (Z.S.)
| | - Fang Xie
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100191, China; (B.L.); (K.D.); (Y.Z.); (X.W.); (Z.S.)
| | - Lingjia Qian
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100191, China; (B.L.); (K.D.); (Y.Z.); (X.W.); (Z.S.)
| |
Collapse
|
11
|
Luo Y, Meng D, Tang H, Wu P, Zhang Y. Exercise alleviates CUS-induced depressive-like behaviors by modulating paracellular and transcellular permeability of the blood-brain barrier in the prefrontal cortex. Behav Brain Res 2025; 476:115286. [PMID: 39389268 DOI: 10.1016/j.bbr.2024.115286] [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: 05/22/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Increased blood-brain barrier (BBB) permeability is implicated in the pathophysiology of major depressive disorder (MDD). While aerobic exercise has shown promise in mitigating MDD symptoms by potentially preserving BBB integrity, the detailed mechanisms remain unclear. This study explores these mechanisms to assess aerobic exercise's therapeutic potential for MDD. METHODS Male C57BL/6 J mice were used in this study to investigate the effects of aerobic exercise on CUS-induced BBB permeability and depressive-like behaviors. Chronic unpredictable stress (CUS)-induced MDD mouse models were divided into three groups: Control, CUS, and CUS+Exercise. We monitored body weight, blood S100β levels, and cytokines via ELISA. Claudin-5 and Caveolin-1 (CAV-1) expressions in the medial prefrontal cortex were evaluated using Western blotting and immunofluorescence. BBB permeability was assessed using biocytin-TMR and Alb-Alexa 594 tracers. Transmission electron microscopy was used to observe ultrastructural changes in the BBB directly. Depression-related behaviors were tested through several behavioral assays. RESULTS CUS significantly increased CAV-1 expression and Alb-Alexa 594 leakage, suggesting enhanced transcellular BBB permeability. Despite unchanged Claudin-5 levels, its tight junction ultrastructure was altered, leading to increased biocytin-TMR leakage. Aerobic exercise ameliorated these disruptions, reduced inflammatory cytokines, and improved behavioral outcomes in CUS mice. CONCLUSION Disruptions in both paracellular and transcellular BBB pathways are pivotal in depression development. Aerobic exercise offers potential therapeutic benefits for MDD linked with BBB dysfunction by mitigating stress-induced structural and functional changes.
Collapse
MESH Headings
- Animals
- Blood-Brain Barrier/metabolism
- Male
- Mice, Inbred C57BL
- Prefrontal Cortex/metabolism
- Mice
- Physical Conditioning, Animal/physiology
- Stress, Psychological/metabolism
- Stress, Psychological/therapy
- Stress, Psychological/physiopathology
- Caveolin 1/metabolism
- Disease Models, Animal
- Claudin-5/metabolism
- Depressive Disorder, Major/therapy
- Depressive Disorder, Major/metabolism
- Depressive Disorder, Major/physiopathology
- Depression/therapy
- Depression/metabolism
- Behavior, Animal/physiology
- Permeability
- Exercise Therapy/methods
Collapse
Affiliation(s)
- Ye Luo
- College of fine arts, China West Normal University, Nanchong, China
| | - Dewang Meng
- College of Physical Education, China West Normal University, Nanchong, China
| | - Hui Tang
- Department of Neurosurgery, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Panwen Wu
- College of Physical Education, China West Normal University, Nanchong, China
| | - Yuan Zhang
- Department of Neurosurgery, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
12
|
Li K, Wang K, Xu SX, Xie XH, Tang Y, Zhang L, Liu Z. In vivo evidence of increased vascular endothelial growth factor in patients with major depressive disorder. J Affect Disord 2025; 368:151-159. [PMID: 39278472 DOI: 10.1016/j.jad.2024.09.073] [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: 05/21/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) is a candidate mediator of blood-brain barrier (BBB) disruption in depression. However, previous studies have mainly focused on peripheral blood VEGF levels, and the results are heterogeneous. Here we use astrocyte-derived extracellular vesicles (ADEVs) isolated from plasma to explore the in vivo changes of VEGF levels in patients with major depressive disorder (MDD). METHODS Thirty-five unmedicated patients with MDD and 35 healthy controls (HCs) were enrolled, and plasma ADEVs were isolated from each participant. VEGF levels in ADEVs and glial fibrillary acidic protein (GFAP) in plasma were measured. Additionally, Alix and CD81, two established extracellular vesicle markers, were quantified in ADEVs. RESULTS At baseline, MDD patients exhibited significantly increased levels of VEGF in ADEVs and GFAP in plasma. Following four weeks of selective serotonin reuptake inhibitor treatment, these target protein levels did not significantly change. ROC curve analysis revealed an AUC of 0.711 for VEGF in ADEVs. In exploratory analysis, VEGF levels in ADEVs were positively correlated with Alix and CD81. LIMITATIONS Multiple factors regulate BBB permeability. This study focused solely on VEGF and the sample size for longitudinal analysis was relatively small. CONCLUSION Our study is the first to confirm increased ADEV-derived VEGF levels in patients with MDD, thereby providing preliminary evidence supporting the hypothesis that the BBB is disrupted in depression.
Collapse
Affiliation(s)
- Kun Li
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Clinical Laboratory, Affiliated Hospital of West Anhui Health Vocational College, Lu'an, Anhui, China
| | - Kun Wang
- Department of Psychiatry, Affied Hospital of West Anhui Health Vocational College, Lu'an, Anhui, China
| | - Shu-Xian Xu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xin-Hui Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yan Tang
- Department of Psychiatry, Affied Hospital of West Anhui Health Vocational College, Lu'an, Anhui, China
| | - Lihong Zhang
- Clinical Laboratory, Affiliated Hospital of West Anhui Health Vocational College, Lu'an, Anhui, China
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
13
|
Feng J, Cen Q, Cui Y, Hu X, Li M, Wang L, Wei J, Sun N, Wang J, Zhang A. Lactobacillus rhamnosus: An emerging probiotic with therapeutic potential for depression. Pharmacol Res 2025; 211:107541. [PMID: 39653301 DOI: 10.1016/j.phrs.2024.107541] [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: 08/31/2024] [Revised: 11/18/2024] [Accepted: 12/06/2024] [Indexed: 12/16/2024]
Abstract
Depression, a complex psychological disorder, involves multiple biological pathways in its pathogenesis. In recent years, the gut-brain axis theory has provided novel insights into the pathogenesis of depression, particularly the crucial role of the gut microbiota in mood regulation. While there remains no universal consensus on the most efficacious strains for depression treatment, Lactobacillus rhamnosus has risen to prominence within the realm of probiotics for its potential to positively modulate depressive symptoms. This review preliminarily explores the clinical significance of Lactobacillus rhamnosus in the treatment of depression and summarizes the potential mechanisms by which Lactobacillus rhamnosus treats depression, including its regulation of gut microbiota, alterations in gene expression, improvement of intestinal barrier function, maintenance of neurotransmitter balance, suppression of inflammatory responses, modulation of the immune system, coping with oxidative stress, and optimization of metabolic processes. Future research needs to further explore these mechanisms and combine them with clinical research results to optimize treatment plans and provide more effective treatment options for patients with depression.
Collapse
Affiliation(s)
- Jing Feng
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiuyu Cen
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanru Cui
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaomin Hu
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Min Li
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Linjie Wang
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Juanfang Wei
- College of Physical Education and Health, Geely University of China, Chengdu, China
| | - Nianyi Sun
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Junyu Wang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Anren Zhang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
14
|
Liu F, Jia Y, Zhao L, Xiao LN, Cheng X, Xiao Y, Zhang Y, Zhang Y, Yu H, Deng QE, Zhang Y, Feng Y, Wang J, Gao Y, Zhang X, Geng Y. Escin ameliorates CUMS-induced depressive-like behavior via BDNF/TrkB/CREB and TLR4/MyD88/NF-κB signaling pathways in rats. Eur J Pharmacol 2024; 984:177063. [PMID: 39426465 DOI: 10.1016/j.ejphar.2024.177063] [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: 06/24/2024] [Revised: 09/25/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024]
Abstract
Major depressive disorder (MDD) is a prevalent psychiatric disorder associated with brain inflammation and neuronal damage. Derived from the Aesculus chinensis Bunge fruit, escin has shown anti-inflammatory and neuroprotective effects. However, its potential as a treatment for MDD is unclear. This study investigates the antidepressant properties of escin using in vivo experimentation. The chronic unpredictable mild stress (CUMS) model was used to analyze the potential antidepressant effects and underlying mechanisms of escin. Wistar rats were exposed to CUMS for 35 consecutive days to induce MDD. The rats were then given either escin (1, 3, and 10 mg/kg) or fluoxetine (2 mg/kg) on a daily basis. Notably, escin significantly alleviated the depressive behaviors induced by CUMS, as evaluated through a series of behavioral assessments. Moreover, escin administration reduced TNF-α, IL-1β, and IL-6 levels in the hippocampus. It also decreased serum adrenal cortical hormone (ACTH) and corticosterone (CORT) levels while increasing 5-HT and Brain-derived neurotrophic factor (BDNF) levels in the CUMS rats, as measured by the enzyme-linked immunosorbent assay (ELISA). Pathological changes in the hippocampal regions were identified through Nissl staining, and Western blotting was used to quantify the protein levels of BDNF, TrkB, CREB, TLR4, MyD88, and NF-κB. Escin mitigated neuronal injury, elevated TrkB, BDNF, and CREB, and reduced TLR4, MyD88, and NF-κB protein levels in CUMS rats. The data from this study suggest that escin holds the potential for alleviating depression-like symptoms induced by CUMS. This effect may be mediated through the modulation of two signaling pathways, BDNF/TrkB/CREB and TLR4/MyD88/NF-κB.
Collapse
Affiliation(s)
- Fengjiao Liu
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, Hebei, 050091, China
| | - Yaxin Jia
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, Hebei, 050091, China
| | - Liwei Zhao
- Science and Technology Office, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China
| | - Li-Na Xiao
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, Hebei, 050091, China
| | - Xizhen Cheng
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China
| | - Yingying Xiao
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, Hebei, 050091, China
| | - Ying Zhang
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, Hebei, 050091, China
| | - Yuling Zhang
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China
| | - Huimin Yu
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, Hebei, 050091, China
| | - Qiao-En Deng
- The Eighth Hospital of Shijiazhuang, Shijiazhuang, Hebei, 050081, China
| | - Yuanyuan Zhang
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, Hebei, 050091, China
| | - Yimeng Feng
- College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China
| | - Junfang Wang
- College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China
| | - Yonggang Gao
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, Hebei, 050091, China; Department of Preventive Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Shijiazhuang, Hebei, 050091, China.
| | - Xuan Zhang
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, Hebei, 050091, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Shijiazhuang, Hebei, 050091, China; Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050091, China.
| | - Yunyun Geng
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China; Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, Hebei, 050091, China; Heibei Key Laboratory of Chinese Medicine Research on Cardiocerebrovascular Disease, Shijiazhuang, Hebei, 050091, China.
| |
Collapse
|
15
|
Shi W, Zhang S, Yao K, Meng Q, Lu Y, Ren Y, Li L, Zhao J. Breakdown of the blood-brain barrier in depressed mice induced by chronic unpredictable mild stress. J Psychiatr Res 2024; 180:138-146. [PMID: 39418882 DOI: 10.1016/j.jpsychires.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/28/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Recent studies have suggested potential impairment of the blood-brain barrier (BBB) in depression. However, due to the limited research and variability in animal models, further investigation using diverse and stable models is necessary. METHODS A male mouse model of depression was established using the chronic unpredictable mild stress (CUMS) protocol. Following model establishment, depression-like behaviors were assessed using the sucrose preference test, tail suspension test, and forced swimming test. Morphological changes in the hippocampus were examined through hematoxylin-eosin staining. BBB permeability was evaluated using the Evans blue leakage test, fluorescein sodium (NaF) leakage test, and serum S100B content assessment. Gene and protein expression levels of BBB-related proteins in the hippocampus were determined via real-time PCR, western blotting, and immunofluorescence assays. RESULTS CUMS exposure induced depression-like behaviors, including reduced body weight gain, diminished sucrose preference, and prolonged immobility in both the tail suspension test and forced swimming test. While no significant pathological changes were observed in the hippocampus of either group, increased BBB permeability was noted in the CUMS group, as evidenced by enhanced NaF leakage into the brain parenchyma and elevated serum S100B levels. Gene expression analysis revealed downregulation of angiogenesis-related genes and tight junction proteins in the CUMS group. Additionally, protein levels of tight junction proteins Claudin-5 and ZO-1 were lower in the CUMS group compared to controls. LIMITATIONS This study is limited to a male mouse model, and the BBB in females is worth exploring in the future. CONCLUSIONS Increased BBB permeability and decreased expression of tight junction proteins Claudin5 and ZO-1 were observed in mice with CUMS-induced depression.
Collapse
Affiliation(s)
- Wei Shi
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Shan Zhang
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Kaihu Yao
- Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Qinghong Meng
- Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Yujia Lu
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yi Ren
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Li Li
- Department of International Medical Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Department of Integrated Traditional and Western Medicine, Capital Medical University, Beijing, 100050, China.
| | - Jingjie Zhao
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Department of Integrated Traditional and Western Medicine, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
16
|
Zhang J, Gao T, Chen G, Liang Y, Nie X, Gu W, Li L, Tong H, Huang W, Lu T, Bian Z, Su L. Vinegar-processed Schisandra Chinensis enhanced therapeutic effects on colitis-induced depression through tryptophan metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156057. [PMID: 39357281 DOI: 10.1016/j.phymed.2024.156057] [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: 07/04/2024] [Revised: 08/28/2024] [Accepted: 09/14/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is an inflammatory bowel disease characterized by its incurable nature and undefined etiology, which is often accompanied by a high prevalence of comorbid depression. The gut-brain axis has emerged as a promising treatment target in recent years. PURPOSE This study aimed to investigate how vinegar-processed Schisandra Chinensis (VSC) enhances therapeutic effects on depressive behavior in chronic UC mice. METHODS A chronic UC model was induced in mice using dextran sulfate sodium. The therapeutic effects of both raw and vinegar-processed Schisandra Chinensis on UC and associated depressive symptoms were assessed. Colonic mucosal damage was evaluated using hematoxylin and eosin (H&E) and Alcian blue staining. The integrity of the blood-brain barrier (BBB) and synaptic structures was visualized via transmission electron microscopy (TEM). Enzyme-linked immunosorbent assay (ELISA) was employed to quantify inflammatory cytokine levels in the colon, serum, and brain, while western blotting was performed for protein expression analysis. Additionally, metagenomic analysis was conducted to investigate gut microbiota composition. Nissl staining and immunofluorescence were used to assess hippocampal neuronal damage, and behavioral assessments including the morris water maze, open field test, forced swimming test and tail suspension test, were implemented to evaluate depressive states. Serum metabolites were analyzed using UPLC-MS/MS. RESULTS Both raw and vinegar-processed Schisandra Chinensis significantly upregulated aryl hydrocarbon receptor (AhR), inhibited NF-κB p-p65 activation, and reduced levels of pro-inflammatory cytokine. These treatments also enhanced the expression of tight junction proteins, restored colonic mucosal and BBB integrity, alleviated damage to hippocampal neurons, and improved synaptic structure. Behavioral assessments indicated that VSC was particularly effective in ameliorating depressive-like behaviors in chronic UC mice. In the gut, both treatments reshaped the gut microbial composition, restoring the relative abundance of Duncaniella, Candidatus_Amulumruptor, Alistipes, Parabacteroides, Lachnospiraceae_bacterium, uncultured_Bacteroides_sp., Candidatus_Amulumruptor_caecigallinarius, with VSC showing more pronounced effects. Serum metabolomics revealed an increase in tryptophan levels and a decrease in kynurenine and xanthurenic acid levels with VSC, indicating that tryptophan metabolism shifted from the kynurenine pathway to the 5-HT or indole pathway. However, this phenomenon did not occur with Schisandra Chinensis (SC). CONCLUSION This study demonstrated that the disruption of tryptophan metabolic balance served as a biological mechanism underlying the occurrence of depressive behaviors induced by UC. The application of SC following vinegar processing enhanced its regulatory effects on gut microbiota and tryptophan metabolism. This findings provided a new insight for the clinical management of gut-brain comorbidities.
Collapse
Affiliation(s)
- Jiuba Zhang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ting Gao
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Gang Chen
- Department of Pharmacy, Dongtai People's Hospital, Yancheng, 224200, China
| | - Yunhao Liang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xinru Nie
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wei Gu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lin Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Huangjin Tong
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Wei Huang
- Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, 213004, China
| | - Tulin Lu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Zhenhua Bian
- Department of Pharmacy, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, 214071, China.
| | - Lianlin Su
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
17
|
Dai Y, Bao L, Huang J, Zhang M, Yu J, Zhang Y, Li F, Yu B, Gong S, Kou J. Endothelial NMMHC IIA dissociation from PAR1 activates the CREB3/ARF4 signaling in thrombin-mediated intracerebral hemorrhage. J Adv Res 2024:S2090-1232(24)00500-9. [PMID: 39521432 DOI: 10.1016/j.jare.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/15/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION There is an urgent need for cerebroprotective interventions to improve the suboptimal outcomes with intracerebral hemorrhage (ICH). Despite the important role of nonmuscle myosin heavy chain IIA (NMMHC IIA) in the blood-brain barrier (BBB), its function in ICH remains unclear. OBJECTIVES The objective of this study is to explore how NMMHC IIA functions in ICH and to evaluate the effectiveness of targeting NMMHC IIA as a treatment for ICH. METHODS We firstly examined the protein expression of NMMHC IIA in clinical patients and animal models with ICH. The function of NNMMHC IIA was then corroborated by using overexpress or knockdown NMMHC IIA specifically in ECs mice and pBMECs. In addition, we explored protein interacts with NMMHC IIA and signaling pathways after ICH by LC-MS/MS and transcriptomics analysis with an emphasis on the function of PAR1 and the CREB3/ARF4 signaling pathway, and validated them in three kind of animal models. To support the clinical translation of our results, we targeted NMMHC IIA to bicalutamide selected from a library of marketed drugs and examined to validate its ameliorative effect on ICH. RESULTS We observed an upregulation of endothelial NMMHC IIA in the brain following the onset of ICH in both patients and mice, while inhibited NMMHC ⅡA improved ICH induced by thrombin, warfarin or tissue plasminogen activator (tPA) after ischemic stroke. Mechanistically, the head domain of NMMHC IIA interacted with protease-activated receptor 1 (PAR1) at the 380-430 aa region and subsequently dissociated and activated the CREB3/ARF4 signaling pathway. We found that bicalutamide and blebbistatin could bind to NMMHC IIA and effectively protect mice from thrombin-mediated ICH. CONCLUSION The findings indicated that NMMHC IIA dissociated from PAR1 and activated CREB3/ARF4 pathway, which aggravated BBB damage induced by thrombin. This suggested that NMMHC IIA was a novel potential therapeutic target for BBB-related diseases.
Collapse
Affiliation(s)
- Yujie Dai
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Liangying Bao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Juan Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Miling Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Junhe Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Yuanyuan Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Fang Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Boyang Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Shuaishuai Gong
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China.
| | - Junping Kou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China.
| |
Collapse
|
18
|
Liu B, Dong K, Chen X, Dong H, Zhao Y, Wang X, Sun Z, Xie F, Qian L. Inhibition of Glycolysis Alleviates Chronic Unpredictable Mild Stress Induced Neuroinflammation and Depression-like Behavior. Brain Sci 2024; 14:1098. [PMID: 39595861 PMCID: PMC11591872 DOI: 10.3390/brainsci14111098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Growing evidence suggests that glucose metabolism plays a crucial role in activated immune cells, significantly contributing to the occurrence and development of neuroinflammation and depression-like behaviors. Chronic stress has been reported to induce microglia activation and disturbances in glucose metabolism in the hippocampus. AIMS This study aims to investigate how chronic stress-mediated glycolysis promotes neuroinflammation and to assess the therapeutic potential of the glycolysis inhibitor, 2-deoxy-D-glucose (2-DG), in a model of chronic stress-induced neuroinflammation and depression-like behavior. METHODS In in vitro studies, we first explored the effects of 2-DG on the inflammatory response of microglia cells. The results showed that corticosterone (Cort) induced reactive oxygen species (ROS) production, increased glycolysis, and promoted the release of inflammatory mediators. However, these effects were reversed by intervention with 2-DG. Subsequently, we examined changes in depression-like behavior and hippocampal glycolysis in mice during chronic stress. The results indicated that chronic stress led to prolonged escape latency in the Morris water maze, increased platform-crossing frequency, reduced sucrose preference index, and extended immobility time in the forced swim test, all of which are indicative of depression-like behavior in mice. Additionally, we found that the expression of the key glycolytic enzyme hexokinase 2 (HK2) was upregulated in the hippocampus of stressed mice, along with an increased release of inflammatory factors. Further in vivo experiments investigated the effects of 2-DG on glycolysis and pro-inflammatory mediator production, as well as the therapeutic effects of 2-DG on chronic stress-induced depression-like behavior in mice. The results showed that 2-DG alleviated chronic stress-induced depression-like behaviors, such as improving escape latency and platform-crossing frequency in the Morris water maze, and increasing the time spent in the center of the open field. Additionally, 2-DG intervention reduced the level of glycolysis in the hippocampus and decreased the release of pro-inflammatory mediators. CONCLUSIONS These findings suggest that 2-DG can mitigate neuroinflammation and depressive behaviors by inhibiting glycolysis and inflammatory responses. Overall, our results highlight the potential of 2-DG as a therapeutic agent for alleviating chronic stress-induced neuroinflammation through the regulation of glycolysis.
Collapse
Affiliation(s)
- Bing Liu
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| | - Ke Dong
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
- School of Medicine, South China University of Technology, Guangzhou 511442, China
| | - Xiaobing Chen
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| | - Huafeng Dong
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| | - Yun Zhao
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| | - Xue Wang
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| | - Zhaowei Sun
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| | - Fang Xie
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| | - Lingjia Qian
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| |
Collapse
|
19
|
Greene C, Rebergue N, Fewell G, Janigro D, Godfrin Y, Campbell M, Lemarchant S. NX210c drug candidate peptide strengthens mouse and human blood-brain barriers. Fluids Barriers CNS 2024; 21:76. [PMID: 39334382 PMCID: PMC11438064 DOI: 10.1186/s12987-024-00577-x] [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: 06/05/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Alterations of blood-brain barrier (BBB) and blood-spinal cord barrier have been documented in various animal models of neurodegenerative diseases and in patients. Correlations of these alterations with functional deficits suggest that repairing barriers integrity may represent a disease-modifying approach to prevent neuroinflammation and neurodegeneration induced by the extravasation of blood components into the parenchyma. Here, we screened the effect of a subcommissural organ-spondin-derived peptide (NX210c), known to promote functional recovery in several models of neurological disorders, on BBB integrity in vitro and in vivo. METHODS In vitro, bEnd.3 endothelial cell (EC) monolayers and two different primary human BBB models containing EC, astrocytes and pericytes, in static and microfluidic conditions, were treated with NX210c (1-100 µM), or its vehicle, for 4 h and up to 5 days. Tight junction (TJ) protein levels, permeability to dextrans and transendothelial electrical resistance (TEER) were evaluated. In vivo, young and old mice (3- and 21-month-old, respectively) were treated daily intraperitoneally with NX210c at 10 mg/kg or its vehicle for 5 days and their brains collected at day 6 to measure TJ protein levels by immunohistochemistry. RESULTS NX210c induced an increase in claudin-5 protein expression after 24-h and 72-h treatments in mouse EC. Occludin level was also increased after a 24-h treatment. Accordingly, NX210c decreased by half the permeability of EC to a 40-kDa FITC-dextran and increased TEER. In the human static BBB model, NX210c increased by ∼ 25% the TEER from 3 to 5 days. NX210c also increased TEER in the human 3D dynamic BBB model after 4 h, which was associated with a reduced permeability to a 4-kDa FITC-dextran. In line with in vitro results, after only 5 days of daily treatments in mice, NX210c restored aging-induced reduction of claudin-5 and occludin levels in the hippocampus, and also in the cortex for occludin. CONCLUSIONS In summary, we have gathered preclinical data showing the capacity of NX210c to strengthen BBB integrity. Through this property, NX210c holds great promises of being a disease-modifying treatment for several neurological disorders with high unmet medical needs.
Collapse
Affiliation(s)
- Chris Greene
- Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | | | | | | | - Yann Godfrin
- Axoltis Pharma, 60 avenue Rockefeller, Lyon, 69008, France
- Godfrin Life-Sciences, Caluire-et-Cuire, 69300, France
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | | |
Collapse
|
20
|
Che J, Sun Y, Deng Y, Zhang J. Blood-brain barrier disruption: a culprit of cognitive decline? Fluids Barriers CNS 2024; 21:63. [PMID: 39113115 PMCID: PMC11305076 DOI: 10.1186/s12987-024-00563-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
Cognitive decline covers a broad spectrum of disorders, not only resulting from brain diseases but also from systemic diseases, which seriously influence the quality of life and life expectancy of patients. As a highly selective anatomical and functional interface between the brain and systemic circulation, the blood-brain barrier (BBB) plays a pivotal role in maintaining brain homeostasis and normal function. The pathogenesis underlying cognitive decline may vary, nevertheless, accumulating evidences support the role of BBB disruption as the most prevalent contributing factor. This may mainly be attributed to inflammation, metabolic dysfunction, cell senescence, oxidative/nitrosative stress and excitotoxicity. However, direct evidence showing that BBB disruption causes cognitive decline is scarce, and interestingly, manipulation of the BBB opening alone may exert beneficial or detrimental neurological effects. A broad overview of the present literature shows a close relationship between BBB disruption and cognitive decline, the risk factors of BBB disruption, as well as the cellular and molecular mechanisms underlying BBB disruption. Additionally, we discussed the possible causes leading to cognitive decline by BBB disruption and potential therapeutic strategies to prevent BBB disruption or enhance BBB repair. This review aims to foster more investigations on early diagnosis, effective therapeutics, and rapid restoration against BBB disruption, which would yield better cognitive outcomes in patients with dysregulated BBB function, although their causative relationship has not yet been completely established.
Collapse
Affiliation(s)
- Ji Che
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Yinying Sun
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Yixu Deng
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Jun Zhang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China.
| |
Collapse
|
21
|
Rajkumar RP. Revisiting a hypothesis: the neurovascular unit as a link between major depression and neurodegenerative disorders. Front Cell Neurosci 2024; 18:1455606. [PMID: 39157756 PMCID: PMC11327082 DOI: 10.3389/fncel.2024.1455606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024] Open
Affiliation(s)
- Ravi Philip Rajkumar
- Department of Psychiatry, Jawaharlal Institute of Postgraduate Medical Education, Pondicherry, India
| |
Collapse
|
22
|
Li T, Sun S, Li Y, Zhang Y, Wei L. Immunotherapy revolutionizing brain metastatic cancer treatment: personalized strategies for transformative outcomes. Front Immunol 2024; 15:1418580. [PMID: 39136027 PMCID: PMC11317269 DOI: 10.3389/fimmu.2024.1418580] [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: 04/16/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024] Open
Abstract
Brain metastatic cancer poses a significant clinical challenge, with limited treatment options and poor prognosis for patients. In recent years, immunotherapy has emerged as a promising strategy for addressing brain metastases, offering distinct advantages over conventional treatments. This review explores the evolving landscape of tumor immunotherapy in the context of brain metastatic cancer, focusing on the intricate interplay between the tumor microenvironment (TME) and immunotherapeutic approaches. By elucidating the complex interactions within the TME, including the role of immune cells, cytokines, and extracellular matrix components, this review highlights the potential of immunotherapy to reshape the treatment paradigm for brain metastases. Leveraging immune checkpoint inhibitors, cellular immunotherapies, and personalized treatment strategies, immunotherapy holds promise in overcoming the challenges posed by the blood-brain barrier and immunosuppressive microenvironment of brain metastases. Through a comprehensive analysis of current research findings and future directions, this review underscores the transformative impact of immunotherapy on the management of brain metastatic cancer, offering new insights and opportunities for personalized and precise therapeutic interventions.
Collapse
Affiliation(s)
- Ting Li
- Medical Oncology Department of Thoracic Cancer 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Shichen Sun
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Yubing Li
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Yanyu Zhang
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Linlin Wei
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| |
Collapse
|
23
|
Bollinger JL, Johnsamuel S, Vollmer LL, Kuhn AM, Wohleb ES. Stress-induced dysfunction of neurovascular astrocytes contributes to sex-specific behavioral deficits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594147. [PMID: 38798398 PMCID: PMC11118421 DOI: 10.1101/2024.05.14.594147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Astrocytes form an integral component of the neurovascular unit, ensheathing brain blood vessels with projections high in aquaporin-4 (AQP4) expression. These AQP4-rich projections facilitate interaction between the vascular endothelium, astrocytes, and neurons, and help stabilize vascular morphology. Studies using preclinical models of psychological stress and post-mortem tissue from patients with major depressive disorder (MDD) have reported reductions in AQP4, loss of astrocytic structures, and vascular impairment in the prefrontal cortex (PFC). Though compelling, the role of AQP4 in mediating stress-induced alterations in blood vessel function and behavior remains unclear. Here, we address this, alongside potential sex differences in chronic unpredictable stress (CUS) effects on astrocyte phenotype, blood-brain barrier integrity, and behavior. CUS led to pronounced shifts in stress-coping behavior and working memory deficits in male -but not female- mice. Following behavioral testing, astrocytes from the frontal cortex were isolated for gene expression analyses. We found that CUS increased various transcripts associated with blood vessel maintenance in astrocytes from males, but either had no effect on- or decreased- these genes in females. Furthermore, CUS caused a reduction in vascular-localized AQP4 and elevated extravasation of a small molecule fluorescent reporter (Dextran) in the PFC in males but not females. Studies showed that knockdown of AQP4 in the PFC in males is sufficient to disrupt astrocyte phenotype and increase behavioral susceptibility to a sub-chronic stressor. Collectively, these findings provide initial evidence that sex-specific alterations in astrocyte phenotype and neurovascular integrity in the PFC contribute to behavioral and cognitive consequences following chronic stress.
Collapse
Affiliation(s)
- Justin L Bollinger
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Shobha Johnsamuel
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Lauren L Vollmer
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Alexander M Kuhn
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Eric S Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
24
|
White AG, Elias E, Orozco A, Robinson SA, Manners MT. Chronic Stress-Induced Neuroinflammation: Relevance of Rodent Models to Human Disease. Int J Mol Sci 2024; 25:5085. [PMID: 38791125 PMCID: PMC11121038 DOI: 10.3390/ijms25105085] [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: 04/09/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
The brain is the central organ of adaptation to stress because it perceives and determines threats that induce behavioral, physiological, and molecular responses. In humans, chronic stress manifests as an enduring consistent feeling of pressure and being overwhelmed for an extended duration. This can result in a persistent proinflammatory response in the peripheral and central nervous system (CNS), resulting in cellular, physiological, and behavioral effects. Compounding stressors may increase the risk of chronic-stress-induced inflammation, which can yield serious health consequences, including mental health disorders. This review summarizes the current knowledge surrounding the neuroinflammatory response in rodent models of chronic stress-a relationship that is continually being defined. Many studies investigating the effects of chronic stress on neuroinflammation in rodent models have identified significant changes in inflammatory modulators, including nuclear factor-κB (NF-κB) and toll-like receptors (TLRs), and cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1β, and IL-6. This suggests that these are key inflammatory factors in the chronic stress response, which may contribute to the establishment of anxiety and depression-like symptoms. The behavioral and neurological effects of modulating inflammatory factors through gene knockdown (KD) and knockout (KO), and conventional and alternative medicine approaches, are discussed.
Collapse
Affiliation(s)
- Abigail G. White
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA
| | - Elias Elias
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA
| | - Andrea Orozco
- Department of Psychology, Williams College, Williamstown, MA 01267, USA
| | | | - Melissa T. Manners
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA
| |
Collapse
|
25
|
Wang X, Wu Y, Tian Y, Hu H, Zhao Y, Xue B, Sun Z, Wei A, Xie F, Qian LJ. GLUT1-mediated microglial proinflammatory activation contributes to the development of stress-induced spatial learning and memory dysfunction in mice. Cell Biosci 2024; 14:48. [PMID: 38627830 PMCID: PMC11020476 DOI: 10.1186/s13578-024-01229-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/05/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Stress is a recognized risk factor for cognitive decline, which triggers neuroinflammation involving microglial activation. However, the specific mechanism for microglial activation under stress and affects learning and memory remains unclear. METHODS The chronic stress mouse model was utilized to explore the relationship between microglial activation and spatial memory impairment. The effect of hippocampal hyperglycemia on microglial activation was evaluated through hippocampal glucose-infusion and the incubation of BV2 cells with high glucose. The gain-and loss-of-function experiments were conducted to investigate the role of GLUT1 in microglial proinflammatory activation. An adeno-associated virus (AAV) was employed to specifically knockdown of GLUT1 in hippocampal microglia to assess its impact on stressed-mice. RESULTS Herein, we found that chronic stress induced remarkable hippocampal microglial proinflammatory activation and neuroinflammation, which were involved in the development of stress-related spatial learning and memory impairment. Mechanistically, elevated hippocampal glucose level post-stress was revealed to be a key regulator of proinflammatory microglial activation via specifically increasing the expression of microglial GLUT1. GLUT1 overexpression promoted microglial proinflammatory phenotype while inhibiting GLUT1 function mitigated this effect under high glucose. Furthermore, specific downregulation of hippocampal microglial GLUT1 in stressed-mice relieved microglial proinflammatory activation, neuroinflammation, and spatial learning and memory injury. Finally, the NF-κB signaling pathway was demonstrated to be involved in the regulatory effect of GLUT1 on microglia. CONCLUSIONS We demonstrate that elevated glucose and GLUT1 expression induce microglia proinflammatory activation, contributing to stress-associated spatial memory dysfunction. These findings highlight significant interplay between metabolism and inflammation, presenting a possible therapeutic target for stress-related cognitive disorders.
Collapse
Affiliation(s)
- Xue Wang
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, #27 Taiping Road, Haidian, Beijing, 100850, China
| | - Yuhan Wu
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, #27 Taiping Road, Haidian, Beijing, 100850, China
| | - Yingrui Tian
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, #27 Taiping Road, Haidian, Beijing, 100850, China
- Centers for Disease Control and Prevention, Jiulongpo District, Chongqing, 400050, China
| | - Hui Hu
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, #27 Taiping Road, Haidian, Beijing, 100850, China
| | - Yun Zhao
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, #27 Taiping Road, Haidian, Beijing, 100850, China
| | - Binghua Xue
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, #27 Taiping Road, Haidian, Beijing, 100850, China
| | - Zhaowei Sun
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, #27 Taiping Road, Haidian, Beijing, 100850, China
| | - Aijun Wei
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, #27 Taiping Road, Haidian, Beijing, 100850, China
| | - Fang Xie
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, #27 Taiping Road, Haidian, Beijing, 100850, China.
| | - Ling-Jia Qian
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, #27 Taiping Road, Haidian, Beijing, 100850, China.
| |
Collapse
|
26
|
Yang J, Zhang S, Wu Q, Chen P, Dai Y, Long J, Wu Y, Lin Y. T cell-mediated skin-brain axis: Bridging the gap between psoriasis and psychiatric comorbidities. J Autoimmun 2024; 144:103176. [PMID: 38364575 DOI: 10.1016/j.jaut.2024.103176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/18/2024] [Accepted: 02/01/2024] [Indexed: 02/18/2024]
Abstract
Psoriasis, a chronic inflammatory skin condition, is often accompanied by psychiatric comorbidities such as anxiety, depression, suicidal ideation, and other mental disorders. Psychological disorders may also play a role in the development and progression of psoriasis. The intricate interplay between the skin diseases and the psychiatric comorbidities is mediated by the 'skin-brain axis'. Understanding the mechanisms underlying psoriasis and psychiatric comorbidities can help improve the efficacy of treatment by breaking the vicious cycle of diseases. T cells and related cytokines play a key role in the pathogenesis of psoriasis and psychiatric diseases, and are crucial components of the 'skin-brain axis'. Apart from damaging the blood-brain barrier (BBB) directly, T cells and secreted cytokines could interact with the hypothalamic-pituitary-adrenal axis (HPA axis) and the sympathetic nervous system (SNS) to exacerbate skin diseases or mental disorders. However, few reviews have systematically summarized the roles and mechanisms of T cells in the interaction between psoriasis and psychiatric comorbidities. In this review, we discussed several key T cells and their roles in the 'skin-brain axis', with a focus on the mechanisms underlying the interplay between psoriasis and mental commodities, to provide data that might help develop effective strategies for the treatment of both psoriasis and psychiatric comorbidities.
Collapse
Affiliation(s)
- Juexi Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China
| | - Song Zhang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qixuan Wu
- Mental Health Services, Blacktown Hospital, Blacktow, NSW, 2148, Australia
| | - Pu Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China
| | - Yan Dai
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China
| | - Junhao Long
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China
| | - Yan Wu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yun Lin
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China.
| |
Collapse
|
27
|
Wu Y, Hu H, Liu W, Zhao Y, Xie F, Sun Z, Zhang L, Dong H, Wang X, Qian L. Hippocampal Lactate-Infusion Enhances Spatial Memory Correlated with Monocarboxylate Transporter 2 and Lactylation. Brain Sci 2024; 14:327. [PMID: 38671979 PMCID: PMC11048250 DOI: 10.3390/brainsci14040327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/24/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Lactate has emerged as a key player in regulating neural functions and cognitive processes. Beyond its function as an energy substrate and signal molecule, recent research has revealed lactate to serve as an epigenetic regulator in the brain. However, the molecular mechanisms by which lactate regulates spatial memory and its role in the prevention of cognitive disorders remain unclear. Herein, we injected L-lactate (10 μmol/kg/d for 6 d) into the mouse's hippocampus, followed by the Morris water maze (MWM) test and molecular analyses. Improved spatial memory performances were observed in mice injected with lactate. Besides, lactate upregulated the expression of synaptic proteins post-synaptic density 95 (PSD95), synaptophysin (SYP), and growth associated protein 43 (GAP43) in hippocampal tissues and HT22 cells, suggesting a potential role in synaptic transmission and memory formation. The facilitative role of monocarboxylate transporter 2 (MCT2), a neuron-specific lactate transporter, in this process was confirmed, as MCT2 antagonists attenuated the lactate-induced upregulation of synaptic proteins. Moreover, lactate induced protein lactylation, a post-translational modification, which could be suppressed by MCT2 inhibition. RNA sequencing of lactated-injected hippocampal tissues revealed a comprehensive gene expression profile influenced by lactate, with significant changes in genes associated with transcriptional progress. These data demonstrate that hippocampal lactate injection enhances spatial memory in mice, potentially through the upregulation of synaptic proteins and induction of protein lactylation, with MCT2 playing a crucial role in these processes. Our findings shed light on the multi-faceted role of lactate in neural function and memory regulation, opening new avenues for therapeutic interventions targeting cognitive disorders.
Collapse
Affiliation(s)
- Yuhan Wu
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Hui Hu
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Weiwei Liu
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
- College of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Yun Zhao
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Fang Xie
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Zhaowei Sun
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Ling Zhang
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Huafeng Dong
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Xue Wang
- Institute of Military Cognition and Brain Sciences, Beijing 100850, China
| | - Lingjia Qian
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| |
Collapse
|
28
|
Murck H, Karailiev P, Karailievova L, Puhova A, Jezova D. Treatment with Glycyrrhiza glabra Extract Induces Anxiolytic Effects Associated with Reduced Salt Preference and Changes in Barrier Protein Gene Expression. Nutrients 2024; 16:515. [PMID: 38398838 PMCID: PMC10893552 DOI: 10.3390/nu16040515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
We have previously identified that low responsiveness to antidepressive therapy is associated with higher aldosterone/cortisol ratio, lower systolic blood pressure, and higher salt preference. Glycyrrhiza glabra (GG) contains glycyrrhizin, an inhibitor of 11β-hydroxysteroid-dehydrogenase type-2 and antagonist of toll-like receptor 4. The primary hypothesis of this study is that food enrichment with GG extract results in decreased anxiety behavior and reduced salt preference under stress and non-stress conditions. The secondary hypothesis is that the mentioned changes are associated with altered gene expression of barrier proteins in the prefrontal cortex. Male Sprague-Dawley rats were exposed to chronic mild stress for five weeks. Both stressed and unstressed rats were fed a diet with or without an extract of GG roots for the last two weeks. GG induced anxiolytic effects in animals independent of stress exposure, as measured in elevated plus maze test. Salt preference and intake were significantly reduced by GG under control, but not stress conditions. The gene expression of the barrier protein claudin-11 in the prefrontal cortex was increased in control rats exposed to GG, whereas stress-induced rise was prevented. Exposure to GG-enriched diet resulted in reduced ZO-1 expression irrespective of stress conditions. In conclusion, the observed effects of GG are in line with a reduction in the activity of central mineralocorticoid receptors. The treatment with GG extract or its active components may, therefore, be a useful adjunct therapy for patients with subtypes of depression and anxiety disorders with heightened renin-angiotensin-aldosterone system and/or inflammatory activity.
Collapse
Affiliation(s)
- Harald Murck
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, 35039 Marburg, Germany
| | - Peter Karailiev
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| | - Lucia Karailievova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| | - Agnesa Puhova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| | - Daniela Jezova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| |
Collapse
|
29
|
Davidson TL, Stevenson RJ. Vulnerability of the Hippocampus to Insults: Links to Blood-Brain Barrier Dysfunction. Int J Mol Sci 2024; 25:1991. [PMID: 38396670 PMCID: PMC10888241 DOI: 10.3390/ijms25041991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
The hippocampus is a critical brain substrate for learning and memory; events that harm the hippocampus can seriously impair mental and behavioral functioning. Hippocampal pathophysiologies have been identified as potential causes and effects of a remarkably diverse array of medical diseases, psychological disorders, and environmental sources of damage. It may be that the hippocampus is more vulnerable than other brain areas to insults that are related to these conditions. One purpose of this review is to assess the vulnerability of the hippocampus to the most prevalent types of insults in multiple biomedical domains (i.e., neuroactive pathogens, neurotoxins, neurological conditions, trauma, aging, neurodegenerative disease, acquired brain injury, mental health conditions, endocrine disorders, developmental disabilities, nutrition) and to evaluate whether these insults affect the hippocampus first and more prominently compared to other brain loci. A second purpose is to consider the role of hippocampal blood-brain barrier (BBB) breakdown in either causing or worsening the harmful effects of each insult. Recent research suggests that the hippocampal BBB is more fragile compared to other brain areas and may also be more prone to the disruption of the transport mechanisms that act to maintain the internal milieu. Moreover, a compromised BBB could be a factor that is common to many different types of insults. Our analysis indicates that the hippocampus is more vulnerable to insults compared to other parts of the brain, and that developing interventions that protect the hippocampal BBB may help to prevent or ameliorate the harmful effects of many insults on memory and cognition.
Collapse
Affiliation(s)
- Terry L. Davidson
- Department of Neuroscience, Center for Neuroscience and Behavior, American University, 4400 Massachusetts Avenue, NW, Washington, DC 20016, USA
| | | |
Collapse
|
30
|
Ling Y, Kang X, Yi Y, Feng S, Ma G, Qu H. CLDN5: From structure and regulation to roles in tumors and other diseases beyond CNS disorders. Pharmacol Res 2024; 200:107075. [PMID: 38228255 DOI: 10.1016/j.phrs.2024.107075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/18/2024]
Abstract
Claudin-5 (CLDN5) is an essential component of tight junctions (TJs) and is critical for the integrity of the blood-brain barrier (BBB), ensuring homeostasis and protection from damage to the central nervous system (CNS). Currently, many researchers have summarized the role and mechanisms of CLDN5 in CNS diseases. However, it is noteworthy that CLDN5 also plays a significant role in tumor growth and metastasis. In addition, abnormal CLDN5 expression is involved in the development of respiratory diseases, intestinal diseases, cardiac diseases, and diabetic ocular complications. This paper aims to review the structure, expression, and regulation of CLDN5, focusing on its role in tumors, including its expression and regulation, effects on malignant phenotypes, and clinical significance. Furthermore, this paper will provide an overview of the role and mechanisms of CLDN5 in respiratory diseases, intestinal diseases, cardiac diseases, and diabetic ocular complications.
Collapse
Affiliation(s)
- Yao Ling
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, China; Bethune Second Clinical Medical College of Jilin University, Changchun, China
| | - Xinxin Kang
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, China; Bethune Second Clinical Medical College of Jilin University, Changchun, China
| | - Ying Yi
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, China; Bethune Second Clinical Medical College of Jilin University, Changchun, China
| | - Shenao Feng
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, China; Bethune Second Clinical Medical College of Jilin University, Changchun, China
| | - Guanshen Ma
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, China; Bethune Second Clinical Medical College of Jilin University, Changchun, China
| | - Huinan Qu
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
31
|
Halvorson CS, Sánchez-Lafuente CL, Johnston JN, Kalynchuk LE, Caruncho HJ. Molecular Mechanisms of Reelin in the Enteric Nervous System and the Microbiota-Gut-Brain Axis: Implications for Depression and Antidepressant Therapy. Int J Mol Sci 2024; 25:814. [PMID: 38255890 PMCID: PMC10815176 DOI: 10.3390/ijms25020814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/30/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Current pharmacological treatments for depression fail to produce adequate remission in a significant proportion of patients. Increasingly, other systems, such as the microbiome-gut-brain axis, are being looked at as putative novel avenues for depression treatment. Dysbiosis and dysregulation along this axis are highly comorbid with the severity of depression symptoms. The endogenous extracellular matrix protein reelin is present in all intestinal layers as well as in myenteric and submucosal ganglia, and its receptors are also present in the gut. Reelin secretion from subepithelial myofibroblasts regulates cellular migration along the crypt-villus axis in the small intestine and colon. Reelin brain expression is downregulated in mood and psychotic disorders, and reelin injections have fast antidepressant-like effects in animal models of depression. This review seeks to discuss the roles of reelin in the gastrointestinal system and propose a putative role for reelin actions in the microbiota-gut-brain axis in the pathogenesis and treatment of depression, primarily reflecting on alterations in gut epithelial cell renewal and in the clustering of serotonin transporters.
Collapse
Affiliation(s)
- Ciara S. Halvorson
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Rd., Victoria, BC V8P 5C2, Canada; (C.S.H.); (C.L.S.-L.); (L.E.K.)
| | - Carla Liria Sánchez-Lafuente
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Rd., Victoria, BC V8P 5C2, Canada; (C.S.H.); (C.L.S.-L.); (L.E.K.)
| | - Jenessa N. Johnston
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Lisa E. Kalynchuk
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Rd., Victoria, BC V8P 5C2, Canada; (C.S.H.); (C.L.S.-L.); (L.E.K.)
| | - Hector J. Caruncho
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Rd., Victoria, BC V8P 5C2, Canada; (C.S.H.); (C.L.S.-L.); (L.E.K.)
| |
Collapse
|