1
|
Zhang W, He J, Wang Y, Jin H, Wang R. Scientific status analysis of exercise benefits for vascular cognitive impairment: Evidence of neuroinflammation. J Neuroimmunol 2025; 402:578574. [PMID: 40086400 DOI: 10.1016/j.jneuroim.2025.578574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/07/2025] [Accepted: 03/02/2025] [Indexed: 03/16/2025]
Abstract
Vascular cognitive impairment (VCI) is a syndrome characterized by cognitive decline resulting from insufficient perfusion to the entire brain or specific brain regions. The lack of a clear understanding of the mechanisms linking cerebrovascular disease to cognitive impairment has impeded the development of targeted treatments for VCI. Increasing evidence indicates that exercise may offer significant benefits for patients with VCI. This study explores how neuroinflammatory mechanisms mediate the effects of exercise on VCI, focusing on the broader biological processes involved. Exercise plays a crucial role in mitigating vascular risk factors, reducing oxidative stress, and promoting neurogenesis. Furthermore, exercise influences neuroinflammatory mediators and central immune cells via various signaling pathways. Different types and intensities of exercise, including resistance and endurance training, have been shown to differentially modulate neuroinflammation during the progression of VCI. This paper summarizes the current mechanisms of action and proposes exercise interventions targeting neuroinflammatory pathways, along with biomarker studies, to enhance our understanding of VCI pathogenesis and inform clinical practice. A more in-depth understanding of the inflammatory mechanisms underlying VCI may facilitate the development of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Wei Zhang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jing He
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuxin Wang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - He Jin
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Rong Wang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing, China; Beijing Institute of Major Brain Diseases, Beijing, China.
| |
Collapse
|
2
|
Zhang Z, Ru D, Liu Z, Guo Z, Zhu L, Zhang Y, Chu M, Wang Y, Zhao J. Integrative Multiomics Profiling of Mouse Hippocampus Reveals Transcriptional Upregulation of Interferon-Stimulated Genes Through PU.1 Regulator in Microglial Activation Induced by Chronic Cerebral Hypoperfusion. MedComm (Beijing) 2025; 6:e70157. [PMID: 40242160 PMCID: PMC11999893 DOI: 10.1002/mco2.70157] [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: 07/01/2024] [Revised: 12/31/2024] [Accepted: 02/19/2025] [Indexed: 04/18/2025] Open
Abstract
Chronic cerebral hypoperfusion (CCH) is a significant factor that accelerates cognitive deterioration, yet the mechanisms of hippocampal microglial activation in this context remain unclear. Using an integrative multiomics approach, we investigated the transcriptional and epigenomic landscape of microglial activation in a mouse model of CCH induced by bilateral common carotid artery stenosis. Behavioral assessments revealed cognitive impairments, while neuropathological analysis confirmed hippocampal damage. Proteomic and transcriptomic profiling uncovered significant upregulation of stress and inflammatory pathways, particularly the interferon (IFN) signaling cascade. Epigenomic analysis identified regions of open chromatin, suggesting active transcriptional regulation driven by the transcription factor (TF) PU.1. ChIP-nexus analysis further confirmed that PU.1 directly modulates the expression of IFN-stimulated genes (ISGs), which are pivotal in regulating microglial activation. Our findings demonstrate that PU.1 serves as a key regulator of the IFN-driven microglial response during CCH, mediated by enhanced chromatin accessibility and transcriptional activation of ISGs. This study highlights the critical role of PU.1 in microglial-mediated neuroinflammation and offers potential therapeutic targets for mitigating hippocampal damage associated with chronic cerebral ischemia.
Collapse
Affiliation(s)
- Zengyu Zhang
- Department of NeurologyMinhang HospitalFudan UniversityShanghaiChina
- Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Dewen Ru
- Shanghai Medical CollegeFudan UniversityShanghaiChina
- Department of NeurosurgeryJinshan HospitalFudan UniversityShanghaiChina
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Zhuohang Liu
- Department of NeurologyMinhang HospitalFudan UniversityShanghaiChina
- Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Zimin Guo
- Department of NeurologyShanghai Pudong Hospital, Fudan University Pudong Medical CenterShanghaiChina
| | - Lei Zhu
- Department of Vascular SurgeryHuashan Hospital, Fudan UniversityShanghaiChina
| | - Yuan Zhang
- Department of Vascular SurgeryShanghai Pudong Hospital, Fudan University Pudong Medical CenterShanghaiChina
| | - Min Chu
- Department of NeurologyMinhang HospitalFudan UniversityShanghaiChina
| | - Yong Wang
- Department of NeurologyZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Jing Zhao
- Department of NeurologyMinhang HospitalFudan UniversityShanghaiChina
- Institute of Healthy Yangtze River DeltaShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
3
|
Zhang J, Zhang Y, Zhang Y, Lu K, Zhang X, Wang X, Fu J. Farrerol alleviates cognitive impairments in chronic cerebral hypoperfusion via suppressing NLRP3 inflammasome-mediated pyroptosis. Int Immunopharmacol 2025; 153:114442. [PMID: 40112599 DOI: 10.1016/j.intimp.2025.114442] [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: 12/16/2024] [Revised: 02/21/2025] [Accepted: 03/04/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) serves as a critical pathological mechanism that contributes to the development of vascular dementia (VaD). NLRP3 inflammasome activation is a pivotal factor in promoting cognitive decline in CCH. Farrerol, a dihydroflavonoid derived from Rhododendron and other Ericaceae species, possesses anti-inflammatory, antioxidant, and neuroprotective properties. However, its role in regulating pyroptosis triggered by the NLRP3 inflammasome in CCH remains unclear. METHODS A permanent CCH rat model was generated by occluding the bilateral common carotid arteries (BCCAO), and cellular models of sustained hypoxia were used to mimic CCH in vitro. Eight weeks post-surgery, rats received farrerol treatment. Behavioral tests were conducted after four weeks of treatment. Brain tissues were analyzed via histological staining, immunofluorescence, qRT-PCR, ELISA, and Western blot. The anti-pyroptosis effects and mechanisms of farrerol were also tested in BV2 cells and primary microglia subjected to hypoglycemia and hypoxia conditions. RESULTS Farrerol markedly alleviated cognitive impairments and neural damage caused by CCH. In CCH rats, farrerol suppressed the activation of the NLRP3 inflammasome, decreased the levels of IL-1β and IL-18, and reduced pyroptosis. The in vitro experiments also demonstrated that farrerol could reduce chronic hypoxia-induced pyroptosis by inhibiting the NLRP3 inflammasome pathway. The cellular study further showed that the beneficial effects of farrerol in CCH are via modulating the MAPK-NF-κB pathway. CONCLUSION Farrerol mitigates CCH-induced cognitive dysfunction by inhibiting NLRP3 inflammasome-associated pyroptosis via modulating the MAPK-NF-κB signaling cascade. These findings underscore the potential of farrerol as a therapeutic candidate for CCH.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Neurology, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China; Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China
| | - Yaxuan Zhang
- Department of Neurology, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China; Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China
| | - Yueqi Zhang
- Department of Neurology, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China; Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China
| | - Kaili Lu
- Department of Neurology, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China; Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China
| | - Xiaojie Zhang
- Department of Neurology, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China; Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China
| | - Xiuzhe Wang
- Department of Neurology, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China; Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China.
| | - Jianliang Fu
- Department of Neurology, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China; Department of Neurology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China.
| |
Collapse
|
4
|
Lin H, Yin L, Liu W, Li R, Jiang T, Yang M, Cao Y, Wang S, Yu Y, Chen C, Guo X, Wang W, Liu H, Dai Y, Yan J, Lin Y, Ding Y, Ruan C, Yang L, Wu T, Tao J, Chen L. Muscle-Derived Small Extracellular Vesicles Mediate Exercise-Induced Cognitive Protection in Chronic Cerebral Hypoperfusion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2410209. [PMID: 40271743 DOI: 10.1002/advs.202410209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 04/06/2025] [Indexed: 04/25/2025]
Abstract
Physical exercise protects against cognitive impairment caused by chronic cerebral hypoperfusion (CCH). However, the mechanisms through which exercise sends signals from the periphery to the central nervous system remain incompletely understood. This study demonstrated that exercise promotes the secretion of muscle-derived small extracellular vesicles (sEVs), which facilitate interorgan communication between the muscle and the brain. Systematic delivery of muscle-derived sEVs enhances synaptic plasticity and alleviated cognitive impairment in CCH. Notably, miRNA sequencing reveal miR-17/20a-5p as key cargos in sEVs involved in the exercise-induced muscle-brain crosstalk. Muscle-derived sEVs are also identified as the primary source of swimming-induced miR-17/20a-5p in circulating sEVs. Mechanistically, miR-17/20a-5p binds to the DEP-domain containing mTOR-interacting protein (DEPTOR) and activates the mammalian target of rapamycin (mTOR) pathway in the hippocampus. Depletion of miR-17/20a-5p from muscle-derived sEVs impairs the exercise-induced enhancement of synaptic plasticity and cognitive function. Moreover, overexpression of DEPTOR in the hippocampus attenuates the cognitive benefits of exercise. Conversely, hippocampus-specific activation of mTOR reverses these effects, highlighting the crucial role of mTOR in mediating the positive effects of exercise. Collectively, these findings identify miR-17/20a-5p in muscle-derived sEVs as the exercise-induced myokine with potent effects on the brain, emphasizing the therapeutic potential of exercise in managing cognitive impairment.
Collapse
Affiliation(s)
- Huawei Lin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Lianhua Yin
- The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350003, China
| | - Weilin Liu
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Provincial and Ministerial Co-founded Collaborative Innovation Center of Rehabilitation Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Rui Li
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Tao Jiang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Minguang Yang
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Provincial and Ministerial Co-founded Collaborative Innovation Center of Rehabilitation Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Key Laboratory of Cognitive Rehabilitation of Fujian Province, Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350001, China
- Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Yajun Cao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Sinuo Wang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Yan Yu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Cong Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Xiaoqin Guo
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Wenju Wang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Huanhuan Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Yaling Dai
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Jiamin Yan
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Yanting Lin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Yanyi Ding
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Chendong Ruan
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Provincial and Ministerial Co-founded Collaborative Innovation Center of Rehabilitation Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Key Laboratory of Cognitive Rehabilitation of Fujian Province, Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350001, China
| | - Lei Yang
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Key Laboratory of Cognitive Rehabilitation of Fujian Province, Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350001, China
| | - Tiecheng Wu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Key Laboratory of Cognitive Rehabilitation of Fujian Province, Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350001, China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Provincial and Ministerial Co-founded Collaborative Innovation Center of Rehabilitation Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Key Laboratory of Cognitive Rehabilitation of Fujian Province, Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350001, China
| | - Lidan Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Provincial and Ministerial Co-founded Collaborative Innovation Center of Rehabilitation Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Key Laboratory of Cognitive Rehabilitation of Fujian Province, Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350001, China
- Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| |
Collapse
|
5
|
Wu X, Yang Q, Xie Y, Xia L, Li J, An W, Lu X. Drug-targeted Mendelian randomization analysis combined with transcriptome sequencing to explore the molecular mechanisms associated with cognitive impairment. J Alzheimers Dis 2025:13872877251335891. [PMID: 40267292 DOI: 10.1177/13872877251335891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
BackgroundCurrent therapies for cognitive impairment, including Alzheimer's disease (AD) and mild cognitive impairment, are limited by a lack of universal treatment and adverse effects associated with polypharmacy. Investigating genetic and molecular mechanisms underlying cognitive decline is critical for the development of targeted therapeutics.ObjectiveTo identify causal genes and potential therapeutic targets for cognitive impairment through integrative genomic analyses.MethodsGenome-wide association study data on cognitive impairment were combined with the expression quantitative trait loci (eQTL) data from the eQTLGen consortium. Mendelian randomization (MR) and colocalization analyses were employed to infer causal relationships. Gene Set Enrichment Analysis and Gene Set Variation Analysis evaluated the pathway and functional differences. Immune cell infiltration patterns and the immunometabolic pathways were assessed, followed by drug target prediction.ResultsMR analysis identified seven gene-eQTL pairs significantly associated with cognitive impairment. SMR colocalization prioritized three key genes: HNMT (histamine metabolism), TNFSF8 (inflammatory signaling), and S1PR5 (sphingolipid signaling). HNMT, TNFSF8, and S1PR5 had 39, 24, and 30 predicted targeted drugs, respectively, including arsenic trioxide, aspirin, and immunomodulators.ConclusionsThis study implicates HNMT, TNFSF8, and S1PR5 as potential therapeutic targets for cognitive impairment. Further validation is required to confirm their clinical relevance.
Collapse
Affiliation(s)
- Xixi Wu
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Qingyan Yang
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Yudi Xie
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Lingfeng Xia
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Jiatao Li
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Wenting An
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Xiao Lu
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| |
Collapse
|
6
|
Lin Y, Zhang Q, Chen L, Liu Y, Lin X, Peng X, Cao H, Lei Y, Wang X. Neomycin affects cardiovascular and hematopoietic system via the PI3K/Akt pathway in zebrafish larvae. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 296:118203. [PMID: 40262245 DOI: 10.1016/j.ecoenv.2025.118203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/05/2025] [Accepted: 04/13/2025] [Indexed: 04/24/2025]
Abstract
Neomycin, a widely used aminoglycoside antibiotic, poses potential risks to organism and the environment that remain incompletely evaluated. This study systematically evaluates its toxic effects on zebrafish embryos across physiological, cellular, molecular, and behavioral dimensions. At the physiological level, neomycin exposure induces severe developmental abnormalities, including yolk sac edema, reduced body length, and craniofacial malformations. Developmental disorders of the cardiovascular and hematopoietic systems are confirmed in exposed larvae. In addition, zebrafish larvae exposed to neomycin exhibit significant locomotor deficits, including reduced swimming speed, distance traveled, and impaired responsiveness to light-dark stimulation, indicating reduced activity. Mechanically, neomycin triggers oxidative stress through a dose-dependent elevation of reactive oxygen species (ROS) levels and induces cellular apoptosis through the PI3K/Akt signaling pathway. Collectively, our findings demonstrate that neomycin exerts toxic effects on zebrafish embryonic development, highlighting concerns regarding neomycin exposure risks during early pregnancy and providing critical insights into its potential environmental hazards.
Collapse
Affiliation(s)
- Yuan Lin
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350013, China; Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350011, China
| | - Qiuping Zhang
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350013, China; Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350011, China
| | - Lu Chen
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350013, China
| | - Yingying Liu
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350013, China; Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350011, China
| | - Xiaoxi Lin
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350013, China
| | - Xiaoyan Peng
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350013, China
| | - Hua Cao
- Shengli Clinical Medical College of Fujian Medical University Fujian Provincial Hospital, Fuzhou 350001, China
| | - Yuqing Lei
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350013, China.
| | - Xinrui Wang
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350013, China.
| |
Collapse
|
7
|
Altahrawi AY, James AW, Shah ZA. The Role of Oxidative Stress and Inflammation in the Pathogenesis and Treatment of Vascular Dementia. Cells 2025; 14:609. [PMID: 40277934 DOI: 10.3390/cells14080609] [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: 03/11/2025] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
Vascular dementia (VaD) is a heterogeneous group of brain disorders caused by cerebrovascular pathologies and the second most common cause of dementia, accounting for over 20% of cases and posing an important global health concern. VaD can be caused by cerebral infarction or injury in critical brain regions, including the speech area of the dominant hemisphere or arcuate fasciculus of the dominant hemisphere, leading to notable cognitive impairment. Although the exact causes of dementia remain multifactorial and complex, oxidative stress (reactive oxygen species), neuroinflammation (TNFα, IL-6, and IL-1β), and inflammasomes are considered central mechanisms in its pathology. These conditions contribute to neuronal damage, synaptic dysfunction, and cognitive decline. Thus, antioxidants and anti-inflammatory agents have emerged as potential therapeutic targets in dementia. Recent studies emphasize that cerebrovascular disease plays a dual role: first, as a primary cause of cognitive impairment and then as a contributor to the manifestation of dementia driven by other factors, such as Alzheimer's disease and other neurodegenerative conditions. This comprehensive review of VaD focuses on molecular mechanisms and their consequences. We provided up-to-date knowledge about epidemiology, pathophysiological mechanisms, and current therapeutic approaches for VaD.
Collapse
Affiliation(s)
- Aseel Y Altahrawi
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Antonisamy William James
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Zahoor A Shah
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
8
|
Liu D, Zhao Y, Liu R, Qiao B, Lu X, Bei Y, Niu Y, Yang X. Traditional Chinese medicine as a viable option for managing vascular cognitive impairment: A ray of hope. Medicine (Baltimore) 2025; 104:e41694. [PMID: 40101029 PMCID: PMC11922442 DOI: 10.1097/md.0000000000041694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
Vascular cognitive impairment (VCI) is a prevalent cognitive disorder resulting from cerebrovascular disease and encompasses a spectrum of cognitive deficits, ranging from mild impairment to vascular dementia (VD). VCI is responsible for a minimum of 20% to 40% of all cases of dementia, with its prevalence ranking second only to Alzheimer's disease on a global scale. The pathogenesis of VCI is complex and includes a lack of cholinergic nerve cells, inflammation, oxidative stress, alterations in the blood-brain barrier, and cell apoptosis. Current guideline-recommended drugs have unsatisfactory therapeutic effects. However, traditional Chinese medicine (TCM) has long been associated with treating dementia, and numerous studies regarding treating dementia with TCM have been conducted. The etiology and pathogenesis of VaD are linked to deficiencies in the spleen and kidney, as well as phlegm turbidity. Treatment involves benefiting the spleen and kidney, improving blood circulation, removing blood stasis, and dispelling phlegm. Moreover, TCM presents benefits such as few adverse effects, low cost, long-term use suitability, and preventive effects. This review outlines the pathogenesis of VCI in both modern medicine and TCM, examines traditional prescriptions and single-agent ingredients with their pharmacological effects, emphasizes TCM's unique features, and explores its multi-targeted approach to treating VCI.
Collapse
Affiliation(s)
- Di Liu
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, China
- Department of Pain, Heze Municipal Hospital, Heze, China
| | - YueYu Zhao
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, China
| | - RunFeng Liu
- Department of Traditional Chinese Medicine, Weifang People's Hospital, Weifang, China
| | - BaoGuang Qiao
- Department of Pain, Heze Municipal Hospital, Heze, China
| | - XinRu Lu
- College of Medical, Shandong Yingcai University, Jinan, China
| | - YuanYuan Bei
- Shandong Jiaotong College Hospital, Jinan, China
| | - Yin Niu
- Department of Endocrinology, People's Hospital of Dingtao District, Heze, China
| | - XiaoNi Yang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
9
|
Yang Q, Chen Q, Zhang KB, Liu Y, Zheng JC, Hu DX, Luo J. Sinomenine alleviates neuroinflammation in chronic cerebral hypoperfusion by promoting M2 microglial polarization and inhibiting neuronal pyroptosis via exosomal miRNA-223-3p. Acta Neuropathol Commun 2025; 13:48. [PMID: 40045356 PMCID: PMC11881310 DOI: 10.1186/s40478-025-01950-z] [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: 09/20/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
Chronic cerebral hypoperfusion (CCH) is a major contributor to vascular dementia, with neuroinflammation playing a central role in its pathogenesis. Sinomenine (SINO), a natural alkaloid derived from traditional Chinese medicine, has shown significant anti-inflammatory and neuroprotective properties. However, its efficacy and mechanism of action in CCH remain unclear. In this study, we established a CCH rat model through bilateral common carotid artery occlusion and administered 10 mg/kg of SINO daily. Behavioral tests demonstrated that SINO significantly improved cognitive and memory functions in CCH rats. Histological analysis revealed that SINO effectively reduced neuroinflammation and damage in the hippocampal CA1, CA3, and DG regions. Mechanistically, SINO promoted microglial polarization from the M1 to M2 phenotype, markedly inhibiting the release of pro-inflammatory cytokines, including IL-1β, IL-6, and TNF-α. Further exploration of its neuroprotective mechanism showed that exosomes from SINO-treated microglia were enriched with miRNA-223-3p, which suppressed NLRP3-mediated pyroptosis in neurons. While our findings highlight the therapeutic potential of SINO, further studies are needed to validate its safety and efficacy in diverse populations and chronic settings. In summary, this study not only demonstrates SINO's regulatory effect on microglial polarization in CCH but also unveils a novel neuroprotective mechanism through exosomal miRNA-223-3p delivery, providing a solid theoretical foundation for SINO's potential as a treatment for CCH.
Collapse
Affiliation(s)
- Qu Yang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Precision Cell Therapy, Nanchang, Jiangxi, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- The National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Qi Chen
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Kai-Bing Zhang
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yu Liu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- The National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Jia-Cheng Zheng
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Dong-Xia Hu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
- The National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Jun Luo
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
- Jiangxi Province Key Laboratory of Precision Cell Therapy, Nanchang, Jiangxi, 330006, China.
- The National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
10
|
Xiao J, Wu M, Li H, Zhang S, Deng J, Wu B. TIGAR alleviates cognitive impairment in rats with chronic cerebral hypoperfusion by suppressing oxidative stress and pyroptosis. Am J Transl Res 2025; 17:1223-1236. [PMID: 40092124 PMCID: PMC11909525 DOI: 10.62347/nwqs1671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 01/18/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND It is postulated that oxidative stress and pyroptosis, which are induced by chronic cerebral hypoperfusion (CCH), contribute to the pathogenesis of vascular cognitive impairment (VCI). The protective role of TIGAR in neurologic illnesses has been the subject of extensive examination, yet its role in models of CCH-induced cognitive impairment remains unexplored. The objective of this study was to ascertain whether TIGAR is a neuroprotective agent in rats with CCH that reduces oxidative stress and pyroptosis. METHODS A CCH model was established in rats through the use of bilateral common carotid artery occlusion (BCCAO). The effects of TIGAR on cognitive function and anxiety-depressive behaviors in rats with CCH were examined. To this end, the Y-maze and open field tests were employed. Nissl and hematoxylin-eosin (H&E) staining were used to assess histologic changes in the CA1 area of the hippocampus. Hippocampal glutathione (GSH) activity, malondialdehyde (MDA) content, and NADPH/NADP+ ratio were measured using the WST-8 colorimetric method to assess oxidative stress. The expression of TIGAR and pyroptosis-related proteins was assessed by western blotting. RESULTS A model of CCH-induced cognitive impairment was successfully established. Four weeks after BCCAO, cerebral blood flow returned to normal in the rats, and cognitive impairment and anxiety-depression-like behavior developed. In rats with CCH, MDA levels increased, GSH activity and NADPH/NADP+ ratio decreased, and pyroptosis-related protein expression increased. The pathologic findings indicated that there was an exacerbation of neuronal injury in the CA1 area of the hippocampus and that the cells were loosely arranged. In rats with CCH, overexpression of TIGAR reduced pyroptosis-associated protein expression while increasing MDA content, GSH activity, and NADPH/NADP+ ratio. It promoted neuronal cell survival, and improved cognitive function and anxiety-depression-like behavior. CONCLUSION Overexpression of TIGAR reduced CCH-induced oxidative stress and pyroptosis and ameliorated cognitive dysfunction and anxiety-depression-like behavior in rats. These findings suggest that TIGAR counteracts oxidative stress and prevents pyroptosis, making it a promising target for the treatment of VCI.
Collapse
Affiliation(s)
- Jie Xiao
- Department of Internal Medicine, Nanchong Mental Health Center Nanchong 637000, Sichuan, China
- Innovation Centre for Science and Technology, North Sichuan Medical College Nanchong 637000, Sichuan, China
| | - Min Wu
- Department of Internal Medicine, Nanchong Mental Health Center Nanchong 637000, Sichuan, China
- Innovation Centre for Science and Technology, North Sichuan Medical College Nanchong 637000, Sichuan, China
| | - Hailong Li
- Innovation Centre for Science and Technology, North Sichuan Medical College Nanchong 637000, Sichuan, China
- Department of Gastroenterology, Wusheng People's Hospital Guang'an 638500, Sichuan, China
| | - Shufan Zhang
- Department of Internal Medicine, Nanchong Mental Health Center Nanchong 637000, Sichuan, China
- Innovation Centre for Science and Technology, North Sichuan Medical College Nanchong 637000, Sichuan, China
| | - Jing Deng
- Department of Internal Medicine, Nanchong Mental Health Center Nanchong 637000, Sichuan, China
- Innovation Centre for Science and Technology, North Sichuan Medical College Nanchong 637000, Sichuan, China
| | - Bihua Wu
- Department of Internal Medicine, Nanchong Mental Health Center Nanchong 637000, Sichuan, China
| |
Collapse
|
11
|
Yawoot N, Tocharus J, Tocharus C. Toll-Like Receptor 4-Mediated Neuroinflammation: Updates on Pathological Roles and Therapeutic Strategies in Chronic Cerebral Hypoperfusion. Mol Neurobiol 2025:10.1007/s12035-025-04718-7. [PMID: 39875782 DOI: 10.1007/s12035-025-04718-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/21/2025] [Indexed: 01/30/2025]
Abstract
Neuroinflammation has been acknowledged as being one of the main pathologies that occur following chronic cerebral hypoperfusion (CCH). Since it significantly contributes to neuronal cell damage and thereby leads to cognitive impairment, the signals related to inflammation in hypoperfusion injury have been extensively investigated over the past few years. Toll-like receptor 4 (TLR4) is the key receptor responsible for immune and inflammatory reactions. It has been reported that TLR4 is involved in the pathology of several diseases and has emerged as a therapeutic target for developing a variety of anti-inflammatory compounds. This study explored the pathological roles of TLR4 that potentially cause the promotion of neuroinflammation in CCH damage. The evidence pertinent to the activation of TLR4 and its downstream inflammatory cascades following CCH are also summarized. This study also demonstrated the therapeutic potential of TLR4 inhibition, whether through drugs, substances, or other treatment strategies, in models of CCH-induced neurological dysfunction. The limitations of the accumulated evidence are addressed and discussed in this study. A deeper understanding of the roles of TLR4 in neuroinflammation following CCH damage may help inform the machinery behind pathological processes for advancing further neuroscientific research and developing therapeutic strategies for vascular dementia.
Collapse
Affiliation(s)
- Nuttapong Yawoot
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand.
- Center of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand.
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| |
Collapse
|
12
|
Patil S, Patel D, Kata R, Teichner E, Subtirelu R, Ayubcha C, Werner T, Alavi A. Molecular Imaging with PET in the Assessment of Vascular Dementia and Cerebrovascular Disease. PET Clin 2025; 20:121-131. [PMID: 39477719 DOI: 10.1016/j.cpet.2024.09.001] [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] [Indexed: 11/17/2024]
Abstract
Vascular dementia (VaD) is a unique form of cognitive decline caused by impairment of blood flow to the brain. Atherosclerosis is strongly associated with VaD as plaque accumulation can lead to tissue hypoperfusion or stroke. VaD and atherosclerosis are both diagnosed relatively late in their disease courses, prompting the need for novel diagnostic approaches such as PET to visualize subclinical pathophysiologic changes. This review discusses the use of PET in the assessment of VaD and cerebrovascular disease, focusing on the application of [18F] fluorodeoxyglucose to study neurometabolism and [18F] sodium fluoride to quantify arterial calcification.
Collapse
Affiliation(s)
- Shiv Patil
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Darshil Patel
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rithvik Kata
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Eric Teichner
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Robert Subtirelu
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Cyrus Ayubcha
- Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Thomas Werner
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Abass Alavi
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
13
|
Yang Y, Lu SR, Xu Q, Yu J, Wang Z, Zhang BS, Hong K. Predictive value of nutritional status and serological indicators in elderly patients with mild cognitive impairment. World J Psychiatry 2024; 14:1925-1935. [PMID: 39704370 PMCID: PMC11622028 DOI: 10.5498/wjp.v14.i12.1925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/09/2024] [Accepted: 11/08/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Mild cognitive impairment (MCI) in elderly individuals is a transitional stage between normal cognition and dementia. Understanding the risk factors for MCI and identifying those at high risk are extremely important for the elderly population. AIM To analyze the risk factors for MCI in the elderly population and construct a clinical prediction model. METHODS Total 295 elderly individuals presenting with memory loss diagnosed at Wuxi People's Hospital between March 2021 and March 2024 were included. Comprehensive demographic, clinical, and serological data were collected for analysis. Participants were categorized into either an MCI group or a normal group based on their performance on the Montreal Cognitive Assessment Scale. An elaborate clinical predictive model was developed to predict the likelihood of MCI in stroke patients; its accuracy was evaluated using area under curve values and calibration curves. RESULTS The results of the study showed that old age, hypertension, diabetes, hyperlipidemia, smoking, high-salt diet, high-cholesterol diet, decreased red blood count, increased neutrophil lymphocyte ratio and increased low-density lipoprotein cholesterol were risk factors for the onset of MCI, with A high vitamin diet and elevated high-density lipoprotein cholesterol being protective factors. In addition, the prediction model constructed in this study exhibits good degrees of differentiation and calibration. CONCLUSION The risk factors for MCI are diverse. Early identification of individuals at high risk of MCI can better intervene and improve their quality of life of MCI patients.
Collapse
Affiliation(s)
- Ying Yang
- Department of Geriatrics, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, Jiangsu Province, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi People's Hospital, Wuxi 214023, Jiangsu Province, China
| | - Shou-Rong Lu
- Department of Geriatrics, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, Jiangsu Province, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi People's Hospital, Wuxi 214023, Jiangsu Province, China
| | - Qiao Xu
- Department of Geriatrics, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, Jiangsu Province, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi People's Hospital, Wuxi 214023, Jiangsu Province, China
| | - Jie Yu
- Department of Geriatrics, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, Jiangsu Province, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi People's Hospital, Wuxi 214023, Jiangsu Province, China
| | - Zhuo Wang
- Department of Geriatrics, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, Jiangsu Province, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi People's Hospital, Wuxi 214023, Jiangsu Province, China
| | - Bing-Shan Zhang
- Department of Geriatrics, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, Jiangsu Province, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi People's Hospital, Wuxi 214023, Jiangsu Province, China
| | - Kan Hong
- Department of Geriatrics, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, Jiangsu Province, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi People's Hospital, Wuxi 214023, Jiangsu Province, China
| |
Collapse
|
14
|
Zhong G, Wang X, Zhang Q, Zhang X, Fang X, Li S, Pan Y, Ma Y, Wang X, Wan T, Wang Q. Exploring the therapeutic implications of natural compounds modulating apoptosis in vascular dementia. Phytother Res 2024; 38:5270-5289. [PMID: 39223915 DOI: 10.1002/ptr.8316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Vascular dementia (VaD) is a prevalent form of dementia stemming from cerebrovascular disease, manifesting in memory impairment and executive dysfunction, thereby imposing a substantial societal burden. Unfortunately, no drugs have been approved for the treatment of VaD due to its intricate pathogenesis, and the development of innovative and efficacious medications is urgently needed. Apoptosis, a programmed cell death process crucial for eliminating damaged or unwanted cells within an organism, assumes pivotal roles in embryonic development and tissue homeostasis maintenance. An increasing body of evidence indicates that apoptosis may significantly influence the onset and progression of VaD, and numerous natural compounds have demonstrated significant therapeutic potential. Here, we discuss the molecular mechanisms underlying apoptosis and its correlation with VaD. We also provide a crucial reference for developing innovative pharmaceuticals by systematically reviewing the latest research progress concerning the neuroprotective effects of natural compounds on VaD by regulating apoptosis. Further high-quality clinical studies are imperative to firmly ascertain these natural compounds' clinical efficacy and safety profiles in the treatment of VaD.
Collapse
Affiliation(s)
- Guangcheng Zhong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyue Wang
- Department of Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qian Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xueying Zhang
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, China
| | - Xiaoling Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuting Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yaru Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yujie Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuejing Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ting Wan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
15
|
Ru D, Zhang Z, Liu M, Fan X, Wang Y, Yan Y, Wang E. Downregulation of Notch Signaling-Stimulated Genes in Neurovascular Unit Alterations Induced by Chronic Cerebral Hypoperfusion. Immun Inflamm Dis 2024; 12:e70082. [PMID: 39607309 PMCID: PMC11603426 DOI: 10.1002/iid3.70082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/09/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) is a key contributor to vascular cognitive impairment (VCI) and is typically associated with blood-brain barrier (BBB) damage. This study investigates the pathological mechanisms underlying CCH-induced neurovascular unit (NVU) alterations. METHODS A mouse model of CCH was established using the bilateral common carotid artery stenosis (BCAS) procedure. Decreased cerebral blood flow (CBF) and impaired BBB integrity were assessed. Brain microvessel (BMV)-specific transcriptome profiles were analyzed using RNA-seq, supplemented with published single-cell RNA-seq data. RESULTS RNA-seq revealed neuroinflammation-related gene activation and significant downregulation of Notch signaling pathway genes in BMVs post-BCAS. Upregulated differentially expressed genes (DEGs) were enriched in microglia/macrophages, while downregulated DEGs were prominent in endothelial cells and pericytes. Enhanced activation of vascular-associated microglia (VAM) was linked to neurovascular alterations. CONCLUSION CCH induces significant NVU changes, marked by microglia-associated neuroinflammation and Notch signaling downregulation. These insights highlight potential therapeutic targets for treating neuroinflammatory and vascular-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Dewen Ru
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
- Department of Neurosurgery, Huashan HospitalFudan UniversityShanghaiChina
- Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Zengyu Zhang
- Shanghai Medical CollegeFudan UniversityShanghaiChina
- Department of Neurology, Minhang HospitalFudan UniversityShanghaiChina
| | - Meng Liu
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
| | - Xuhui Fan
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
| | - Yuqi Wang
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
| | - Yufeng Yan
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
| | - Ersong Wang
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
16
|
Ding H, Wang J, Liu S, Xie Y, Zhang X, Yu J. Association between fibrosis-4 index and cognitive impairment in elderly patients with hypertension: A cross-sectional study. J Clin Hypertens (Greenwich) 2024; 26:1246-1255. [PMID: 39276132 PMCID: PMC11555535 DOI: 10.1111/jch.14890] [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: 03/20/2024] [Revised: 06/24/2024] [Accepted: 08/09/2024] [Indexed: 09/16/2024]
Abstract
The fibrosis-4 index (FIB-4) is a noninvasive fibrosis test that is recommended for patients who are at risk of developing hepatic fibrosis. The aim of the study was to explore the correlation between FIB-4 index and the decline of cognitive function among older patients with hypertension. The study used a cross-sectional design to analyze data obtained from the NHANES 2011-2014. The significance of the FIB-4 index correlation with cognitive function in individuals over the age of 60 was evaluated via multivariate regression models. The nonlinear link was described and fitted smoothed curves. There were a total of 2039 participants in the study, and those with a higher FIB-4 index were more susceptible to developing cognitive decline. In the completely adjusted model, the association remained statistically significant between the FIB-4 index and poor cognitive function as measured by CERAD: Total Score (OR = 0.72, 0.57-0.91), Animal Fluency Score (OR = 0.66, 0.48-0.91), and Digit Symbol Score (OR = 0.36, 0.17-0.77). A nonlinear association was found between the FIB-4 and poor cognitive ability: Total Score, CERAD: Score Delayed Recall, Digit Symbol Score, and Animal Fluency Score. In elderly patients with hypertension, a high FIB-4 index is correlated with an increased prevalence of cognitive decline. Hence, the FIB-4 index could potentially serve as a valuable tool for determining individuals with hypertension who are susceptible to both liver-related complications and cognitive impairment.
Collapse
Affiliation(s)
- Hong Ding
- Department of Cardiovascular MedicineLanzhou University Second HospitalLanzhouChina
| | - Jingtao Wang
- Department of Cardiovascular MedicineLanzhou University Second HospitalLanzhouChina
| | - Shu Liu
- Department of Cardiovascular MedicineLanzhou University Second HospitalLanzhouChina
| | - Yafei Xie
- Department of Cardiovascular MedicineLanzhou University Second HospitalLanzhouChina
| | - Xiaowei Zhang
- Department of Cardiovascular MedicineLanzhou University Second HospitalLanzhouChina
| | - Jing Yu
- Department of Cardiovascular MedicineLanzhou University Second HospitalLanzhouChina
| |
Collapse
|
17
|
Wang D, Zhang X, Huang Z, Li Y, Wang X, Wang J, Zhao Y, Lv Q, Wu M, Zha M, Yuan K, Zhu W, Xu G, Xie Y. Theta-burst transcranial magnetic stimulation attenuates chronic ischemic demyelination and vascular cognitive impairment in mice. Exp Neurol 2024; 383:115022. [PMID: 39442857 DOI: 10.1016/j.expneurol.2024.115022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/11/2024] [Accepted: 10/20/2024] [Indexed: 10/25/2024]
Abstract
Vascular cognitive impairment and dementia (VCID) is mainly caused by chronic cerebral hypoperfusion and subsequent white matter lesions. Noninvasive transcranial magnetic stimulation has been utilized in treating various neurological disorders. However, the function of theta-burst transcranial magnetic stimulation on VCID remains to be defined. We utilized 4-week bilateral carotid artery stenosis model of male mice to mimic VCID. Intermittent theta-burst stimulation (iTBS) or consecutive theta-burst stimulation (cTBS) was administered for 14 consecutive days. Through luxol fast blue staining, electron microscopy and immunofluorescence, we found that iTBS, not cTBS, significantly improved demyelination, axonal damage and β-amyloid deposition, without affecting cerebral blood flow in VCID mice. At cellular levels, iTBS rescued the loss of mature oligodendrocytes, promoted precursor cell differentiation, and inhibited pro-inflammatory activation of astrocytes and microglia. Notably, iTBS attenuated cognitive deterioration in both short-term retention and long-term spatial memory of VCID mice as indicated by serial neurobehavioral tests. To explore the molecular involvement of iTBS, mRNA sequencing was carried out. By real-time PCR and combined RNA fluorescence in situ hybridization with immunofluorescence, iTBS was confirmed to increase Rxrg expression specifically in mature oligodendrocytes. Collectively, iTBS could ameliorate vascular cognitive dysfunction, probably via mitigating white matter lesions and neuroinflammation in the corpus callosum. Rxrg signaling in mature oligodendrocytes, which was increased by iTBS, might be a potential target for VCID treatment.
Collapse
Affiliation(s)
- Di Wang
- Department of Neurology, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, Jiangsu 210000, China
| | - Xiaohao Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Zhenqian Huang
- Department of Neurology, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, Jiangsu 210000, China
| | - Yunzi Li
- Department of Neurology, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, Jiangsu 210000, China
| | - Xinyi Wang
- Department of Neurology, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, Jiangsu 210000, China
| | - Jia Wang
- Department of Neurology, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, Jiangsu 210000, China
| | - Ying Zhao
- Department of Neurology, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, Jiangsu 210000, China
| | - Qiushi Lv
- Department of Neurology, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, Jiangsu 210000, China
| | - Min Wu
- Department of Neurology, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Mingming Zha
- Department of Neurology, First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang 310006, China
| | - Kang Yuan
- Department of Neurology, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, Jiangsu 210000, China
| | - Wusheng Zhu
- Department of Neurology, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, Jiangsu 210000, China
| | - Gelin Xu
- Department of Neurology, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, Jiangsu 210000, China.
| | - Yi Xie
- Department of Neurology, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, Jiangsu 210000, China; Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, China.
| |
Collapse
|
18
|
Cheng W, Wang Y, Cheng C, Chen X, Zhang L, Huang W. Single-cell RNA Sequencing Identifies a Novel Subtype of Microglia with High Cd74 Expression that Facilitates White Matter Inflammation During Chronic Cerebral Hypoperfusion. Neurochem Res 2024; 49:2821-2841. [PMID: 39012534 DOI: 10.1007/s11064-024-04206-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 05/17/2024] [Accepted: 07/05/2024] [Indexed: 07/17/2024]
Abstract
Vascular dementia (VaD) causes progressive cognitive decline in the elderly population, but there is short of available therapeutic measures. Microglia-mediated neuroinflammation is vigorously involved in the pathogenesis of VaD, but the traditional classification of microglial M1/M2 phenotypes remains restrictive and controversial. This study aims to investigate whether microglia transform into novel subtypes in VaD. Chronic cerebral hypoperfusion (CCH) rat model was constructed to mimic VaD. Microglia were isolated via magnetic-activated cell sorting and analyzed by single-cell RNA sequencing (scRNA-seq) and bioinformatics. The findings inferred from scRNA-seq and bioinformatics were further validated through in vivo experiments. In this study, microglia were divided into eight clusters. The proportion of MG5 cluster was significantly increased in the white matter of the CCH group compared with the Sham group and was named chronic ischemia-associated microglia (CIAM). Immunity- and inflammation-related genes, including RT1-Db1, RT1-Da, RT1-Ba, Cd74, Spp1, C3, and Cd68, were markedly upregulated in CIAM. Enrichment analysis illustrated that CIAM possessed the function of evoking neuroinflammation. Further studies unveiled that Cd74 is associated with the most abundant GO terms involved in inflammation as well as cell proliferation and differentiation. In addition, microglia-specific Cd74 knockdown mediated by adeno-associated virus decreased the abundance of CIAM in the white matter, thereby mitigating inflammatory cytokine levels, alleviating white matter lesions, and improving cognitive impairment for CCH rats. These findings indicate that Cd74 is the core molecule of CIAM to trigger neuroinflammation and induce microglial differentiation to CIAM, suggesting that Cd74 may be a potential therapeutic target for VaD.
Collapse
Affiliation(s)
- Wenchao Cheng
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Yuhan Wang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Chang Cheng
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiuying Chen
- Department of Neurology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Lan Zhang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Wen Huang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China.
- Department of Neurology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing, China.
| |
Collapse
|
19
|
Tran-Chi VL, Maes M, Nantachai G, Hemrungrojn S, Solmi M, Stoyanov D, Stoyanova K, Tunvirachaisakul C. Cytokine dysregulation in amnestic mild cognitive impairment. Sci Rep 2024; 14:22486. [PMID: 39341896 PMCID: PMC11439069 DOI: 10.1038/s41598-024-73099-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
The pathophysiology of amnestic Mild Cognitive Impairment (aMCI) is largely unknown, although some papers found signs of immune activation. To assess the cytokine network in aMCI after excluding patients with major depression (MDD) and to examine the immune profiles of quantitative aMCI (qMCI) and distress symptoms of old age (DSOA) scores. A case-control study was conducted on 61 Thai aMCI participants and 60 healthy old adults (both without MDD). The Bio-Plex Pro human cytokine 27-plex test kit was used to assay cytokines/chemokines/growth factors in fasting plasma samples. aMCI is characterized by a significant immunosuppression, and reductions in T helper 1 (Th)1 and T cell growth profiles, the immune-inflammatory responses system, interleukin (IL)1β, IL6, IL7, IL12p70, IL13, GM-CSF, and MCP-1. These 7 cytokines/chemokines exhibit neuroprotective effects at physiologic concentrations. In multivariate analyses, three neurotoxic chemokines, CCL11, CCL5, and CXCL8, emerged as significant predictors of aMCI. Logistic regression showed that aMCI was best predicted by combining IL7, IL1β, MCP-1, years of education (all inversely associated) and CCL5 (positively associated). We found that 38.2% of the variance in the qMCI score was explained by IL7, IL1β, MCP-1, IL13, years of education (inversely associated) and CCL5 (positively associated). The DSOA was not associated with any immune data. An imbalance between lowered levels of neuroprotective cytokines and chemokines, and relative increases in neurotoxic chemokines are key factors in aMCI. Future MCI research should always control for the confounding effects of affective symptoms.
Collapse
Affiliation(s)
- Vinh-Long Tran-Chi
- Ph.D. Programme in Clinical Sciences, School of Global Health, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, China.
- Research Institute, Medical University of Plovdiv, Plovdiv, Bulgaria.
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.
- Kyung Hee University, Dongdaemun-gu, Seoul, South Korea.
- Center of Excellence in Cognitive Impairment and Dementia, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
- Cognitive Fitness and Biopsychiatry Technology Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Strategic Research and Innovation Program for the Development of MU-PLOVDIV-(SRIPD-MUP), European Union-NextGenerationEU, Plovdiv, Bulgaria.
| | - Gallayaporn Nantachai
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Medical Services, Somdet Phra Sungharaj Nyanasumvara Geriatric Hospital, Ministry of Public Health, Chon Buri, Chon Buri Province, Thailand
| | - Solaphat Hemrungrojn
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Cognitive Fitness and Biopsychiatry Technology Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Marco Solmi
- Department of Psychiatry, University of Ottawa, Ottawa, Ontario, Canada
- Regional Centre for the Treatment of Eating Disorders and On Track, The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ottawa, Ontario, Canada
- Ottawa Hospital Research Institute (OHRI), Clinical Epidemiology Program, University of Ottawa, Ottawa, Ontario, Canada
- Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany
| | - Drozdstoy Stoyanov
- Research Institute, Medical University of Plovdiv, Plovdiv, Bulgaria
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
- Strategic Research and Innovation Program for the Development of MU-PLOVDIV-(SRIPD-MUP), European Union-NextGenerationEU, Plovdiv, Bulgaria
| | - Kristina Stoyanova
- Research Institute, Medical University of Plovdiv, Plovdiv, Bulgaria
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
- Strategic Research and Innovation Program for the Development of MU-PLOVDIV-(SRIPD-MUP), European Union-NextGenerationEU, Plovdiv, Bulgaria
| | - Chavit Tunvirachaisakul
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Center of Excellence in Cognitive Impairment and Dementia, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
20
|
Li W, Gong Q, Zhu W, Ali T, Yu ZJ, Li S, Yu X. AMPA receptor potentiation alleviates NLRP3 knockout-induced fear generalization in mice. Biochem Biophys Res Commun 2024; 722:150074. [PMID: 38805785 DOI: 10.1016/j.bbrc.2024.150074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/07/2024] [Indexed: 05/30/2024]
Abstract
Genetic knockout and pharmaceutical inhibition of the NLRP3 inflammasome enhances the extinction of contextual fear memory, which is attributed to its role in neuronal and synaptic dysregulation, concurrent with neurotransmitter function disturbances. This study aimed to determine whether NLRP3 plays a role in generalizing fear via the inflammatory axis. We established the NLRP3 KO mice model, followed by behavioral and biochemical analyses. The NLRP3 KO mice displayed impaired fear generalization, lower neuroinflammation levels, and dysregulated neurotransmitter function. Additionally, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, but not the inhibition of NMDA or 5-HT2C receptors, resulted in fear generalization in NLRP3 KO mice because TAT-GluA2 3Y, but not SB242084 and D-cycloserine, treated blocked NLRP3 deprivation effects on fear generalization. Thus, global knockout of NLRP3 is associated with aberrant fear generalization, possibly through AMPA receptor signaling.
Collapse
Affiliation(s)
- Weifen Li
- Department of Infectious Diseases and Shenzhen key laboratory for endogenous infections, the 6th Affiliated Hospital of Shenzhen University Health Science Center, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China; State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Qichao Gong
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Wenhui Zhu
- Southern Medical University, Nanfang Hospital, Department of Laboratory Medicine, Guangzhou, 510515, Guangdong, China.
| | - Tahir Ali
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Zhi-Jian Yu
- Department of Infectious Diseases and Shenzhen key laboratory for endogenous infections, the 6th Affiliated Hospital of Shenzhen University Health Science Center, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China.
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Xiaoming Yu
- Cancer Center, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China.
| |
Collapse
|
21
|
Du Y, He J, Xu Y, Wu X, Cheng H, Yu J, Wang X, An Y, Wu Y, Guo W. SIRT6 prevent chronic cerebral hypoperfusion induced cognitive impairment by remodeling mitochondrial dynamics in a STAT5-PGAM5-Drp1 dependent manner. J Transl Med 2024; 22:788. [PMID: 39183280 PMCID: PMC11346289 DOI: 10.1186/s12967-024-05566-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024] Open
Abstract
Vascular dementia (VaD) is a prevalent form of dementia resulting from chronic cerebral hypoperfusion (CCH). However, the pathogenic mechanisms of VaD and corresponding therapeutic strategies are not well understood. Sirtuin 6 (SIRT6) has been implicated in various biological processes, including cellular metabolism, DNA repair, redox homeostasis, and aging. Nevertheless, its functional relevance in VaD remains unexplored. In this study, we utilized a bilateral common carotid artery stenosis (BCAS) mouse model of VaD to investigate the role of SIRT6. We detected a significant decrease in neuronal SIRT6 protein expression following CCH. Intriguingly, neuron-specific ablation of Sirt6 in mice exacerbated neuronal damage and cognitive deficits after CCH. Conversely, treatment with MDL-800, an agonist of SIRT6, effectively mitigated neuronal loss and facilitated neurological recovery. Mechanistically, SIRT6 inhibited excessive mitochondrial fission by suppressing the CCH-induced STAT5-PGAM5-Drp1 signaling cascade. Additionally, the gene expression of monocyte SIRT6 in patients with asymptomatic carotid stenosis showed a correlation with cognitive outcomes, suggesting translational implications in human subjects. Our findings provide the first evidence that SIRT6 prevents cognitive impairment induced by CCH, and mechanistically, this protection is achieved through the remodeling of mitochondrial dynamics in a STAT5-PGAM5-Drp1-dependent manner.
Collapse
Affiliation(s)
- Yong Du
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, 710032, China
| | - Jiaqing He
- Department of Neurosurgery, Xi'an Medical University, Xi'an, Shaanxi, 710032, China
| | - Yanni Xu
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, 710032, China
| | - Xun Wu
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, 710032, China
| | - Hongbo Cheng
- Department of Neurosurgery, The second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Jiegang Yu
- Department of Neurosurgery, The second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Xiaoliang Wang
- Department of Neurosurgery, The second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Yaqing An
- Department of Emergency, The second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Yang Wu
- Department of Neurosurgery, The second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China.
| | - Wei Guo
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
22
|
Dmytriv TR, Duve KV, Storey KB, Lushchak VI. Vicious cycle of oxidative stress and neuroinflammation in pathophysiology of chronic vascular encephalopathy. Front Physiol 2024; 15:1443604. [PMID: 39161701 PMCID: PMC11330875 DOI: 10.3389/fphys.2024.1443604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/23/2024] [Indexed: 08/21/2024] Open
Abstract
Chronic vascular encephalopathy (CVE) is a frequent cause of vascular mild cognitive impairment and dementia, which significantly worsens the quality of life, especially in the elderly population. CVE is a result of chronic cerebral hypoperfusion, characterized by prolonged limited blood flow to the brain. This causes insufficient oxygenation of the brain leading to hypoxia. The latter can trigger a series of events associated with the development of oxidative/reductive stresses and neuroinflammation. Addressing the gap in knowledge regarding oxidative and reductive stresses in the development of vascular disorders and neuroinflammation can give a start to new directions of research in the context of CVE. In this review, we consider the hypoxia-induced molecular challenges involved in the pathophysiology of CVE, focusing on oxidative stress and neuroinflammation, which are combined in a vicious cycle of neurodegeneration. We also briefly describe therapeutic approaches to the treatment of CVE and outline the prospects for the use of sulforaphane, an isothiocyanate common in cruciferous plants, and vitamin D to break the vicious cycle and alleviate the cognitive impairments characteristic of patients with CVE.
Collapse
Affiliation(s)
- Tetiana R. Dmytriv
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| | - Khrystyna V. Duve
- Department of Neurology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | | | - Volodymyr I. Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| |
Collapse
|
23
|
Xu LL, Yang S, Zhou LQ, Chu YH, Pang XW, You YF, Zhang H, Zhang LY, Zhu LF, Chen L, Shang K, Xiao J, Wang W, Tian DS, Qin C. Bruton's tyrosine kinase inhibition ameliorated neuroinflammation during chronic white matter ischemia. J Neuroinflammation 2024; 21:195. [PMID: 39097747 PMCID: PMC11297596 DOI: 10.1186/s12974-024-03187-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024] Open
Abstract
Chronic cerebral hypoperfusion (CCH), a disease afflicting numerous individuals worldwide, is a primary cause of cognitive deficits, the pathogenesis of which remains poorly understood. Bruton's tyrosine kinase inhibition (BTKi) is considered a promising strategy to regulate inflammatory responses within the brain, a crucial process that is assumed to drive ischemic demyelination progression. However, the potential role of BTKi in CCH has not been investigated so far. In the present study, we elucidated potential therapeutic roles of BTK in both in vitro hypoxia and in vivo ischemic demyelination model. We found that cerebral hypoperfusion induced white matter injury, cognitive impairments, microglial BTK activation, along with a series of microglia responses associated with inflammation, oxidative stress, mitochondrial dysfunction, and ferroptosis. Tolebrutinib treatment suppressed both the activation of microglia and microglial BTK expression. Meanwhile, microglia-related inflammation and ferroptosis processes were attenuated evidently, contributing to lower levels of disease severity. Taken together, BTKi ameliorated white matter injury and cognitive impairments induced by CCH, possibly via skewing microglia polarization towards anti-inflammatory and homeostatic phenotypes, as well as decreasing microglial oxidative stress damage and ferroptosis, which exhibits promising therapeutic potential in chronic cerebral hypoperfusion-induced demyelination.
Collapse
Affiliation(s)
- Lu-Lu Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yun-Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Wei Pang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yun-Fan You
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lu-Yang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li-Fang Zhu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lian Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ke Shang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun Xiao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
24
|
Yang C, He Y, Ren S, Ding Y, Liu X, Li X, Sun H, Jiao D, Zhang H, Wang Y, Sun L. Hydrogen Attenuates Cognitive Impairment in Rat Models of Vascular Dementia by Inhibiting Oxidative Stress and NLRP3 Inflammasome Activation. Adv Healthc Mater 2024; 13:e2400400. [PMID: 38769944 DOI: 10.1002/adhm.202400400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/09/2024] [Indexed: 05/22/2024]
Abstract
Vascular dementia (VaD) is the second most common form of dementia worldwide. Oxidative stress and neuroinflammation are important factors contributing to cognitive dysfunction in patients with VaD. The antioxidant and anti-inflammatory properties of hydrogen are increasingly being utilized in neurological disorders, but conventional hydrogen delivery has the disadvantage of inefficiency. Therefore, magnesium silicide nanosheets (MSNs) are used to release hydrogen in vivo in larger quantities and for longer periods of time to explore the appropriate dosage and regimen. In this study, it is observed that hydrogen improved learning and working memory in VaD rats in the Morris water maze and Y-maze, which elicits improved cognitive function. Nissl staining of neurons shows that hydrogen treatment significantly improves edema in neuronal cells. The expression and activation of reactive oxygen species (ROS), Thioredoxin-interacting protein (TXNIP), NOD-like receptor protein 3 (NLRP3), caspase-1, and IL-1β in the hippocampus are measured via ELISA, Western blotting, real-time qPCR, and immunofluorescence. The results show that oxidative stress indicators and inflammasome-related factors are significantly decreased after 7dMSN treatment. Therefore, it is concluded that hydrogen can ameliorate neurological damage and cognitive dysfunction in VaD rats by inhibiting ROS/NLRP3/IL-1β-related oxidative stress and inflammation.
Collapse
Affiliation(s)
- Congwen Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Yuxuan He
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Shuang Ren
- Department of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Yiqin Ding
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Xinru Liu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Xue Li
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Hao Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Dezhi Jiao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Haolin Zhang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Yingshuai Wang
- Department of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Lin Sun
- School of Psychology, Shandong Second Medical University, Weifang, Shandong, 261053, China
| |
Collapse
|
25
|
E R, Wang Y. Vagus Nerve Stimulation for Improvement of Vascular Cognitive Impairment. Neuropsychiatr Dis Treat 2024; 20:1445-1451. [PMID: 39072312 PMCID: PMC11283790 DOI: 10.2147/ndt.s465249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/13/2024] [Indexed: 07/30/2024] Open
Abstract
Vagus nerve stimulation (VNS) is acknowledged as a highly effective therapy for various neurological conditions, including refractory epilepsy, depression, Alzheimer's disease (AD), migraine, and stroke. Presently, there is an increasing focus on understanding the impact of VNS on cognitive aspects. Numerous studies suggest that VNS suppresses the body's inflammatory response, leading to enhanced cognitive function in patients. Vascular cognitive impairment (VCI) is a severe cognitive dysfunction syndrome resulting from prolonged chronic cerebral hypoperfusion (CCH), where the primary pathogenesis is CCH-induced neuroinflammation. In this paper, we present a comprehensive overview of the research advancements in using VNS for treating VCI and discuss that VNS improves cognitive function in VCI patients by suppressing neuroinflammation, offering insights into a potential novel approach for addressing this condition.
Collapse
Affiliation(s)
- Ridengnaxi E
- Department of Rehabilitation Medicine, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, People’s Republic of China
| | - Yan Wang
- Department of Rehabilitation Medicine, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, People’s Republic of China
| |
Collapse
|
26
|
Wei N, Zhang LM, Xu JJ, Li SL, Xue R, Ma SL, Li C, Sun MM, Chen KS. Astaxanthin Rescues Memory Impairments in Rats with Vascular Dementia by Protecting Against Neuronal Death in the Hippocampus. Neuromolecular Med 2024; 26:29. [PMID: 39014255 DOI: 10.1007/s12017-024-08796-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024]
Abstract
Vascular dementia (VaD) is a cognitive disorder characterized by a decline in cognitive function resulting from cerebrovascular disease. The hippocampus is particularly susceptible to ischemic insults, leading to memory deficits in VaD. Astaxanthin (AST) has shown potential therapeutic effects in neurodegenerative diseases. However, the mechanisms underlying its protective effects in VaD and against hippocampal neuronal death remain unclear. In this study, We used the bilateral common carotid artery occlusion (BCCAO) method to establish a chronic cerebral hypoperfusion (CCH) rat model of VaD and administered a gastric infusion of AST at 25 mg/kg per day for 4 weeks to explore its therapeutic effects. Memory impairments were assessed using Y-maze and Morris water maze tests. We also performed biochemical analyses to evaluate levels of hippocampal neuronal death and apoptosis-related proteins, as well as the impact of astaxanthin on the PI3K/Akt/mTOR pathway and oxidative stress. Our results demonstrated that AST significantly rescued memory impairments in VaD rats. Furthermore, astaxanthin treatment protected against hippocampal neuronal death and attenuated apoptosis. We also observed that AST modulated the PI3K/Akt/mTOR pathway, suggesting its involvement in promoting neuronal survival and synaptic plasticity. Additionally, AST exhibited antioxidant properties, mitigating oxidative stress in the hippocampus. These findings provide valuable insights into the potential therapeutic effects of AST in VaD. By elucidating the mechanisms underlying the actions of AST, this study highlights the importance of protecting hippocampal neurons and suggests potential targets for intervention in VaD. There are still some unanswered questions include long-term effects and optimal dosage of the use in human. Further research is warranted to fully understand the therapeutic potential of AST and its application in the clinical treatment of VaD.
Collapse
Affiliation(s)
- Na Wei
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China.
- Henan Key Laboratory of Tumor Pathology, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China.
- Department of Pathology, School of Basic Medicine, Zhengzhou University, No.100 Ke Xue Avenue, Zhengzhou, 450002, People's Republic of China.
| | - Luo-Man Zhang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Henan Key Laboratory of Tumor Pathology, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, No.100 Ke Xue Avenue, Zhengzhou, 450002, People's Republic of China
| | - Jing-Jing Xu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Henan Key Laboratory of Tumor Pathology, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, No.100 Ke Xue Avenue, Zhengzhou, 450002, People's Republic of China
| | - Sheng-Lei Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Henan Key Laboratory of Tumor Pathology, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, No.100 Ke Xue Avenue, Zhengzhou, 450002, People's Republic of China
| | - Rui Xue
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
| | - Sheng-Li Ma
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
| | - Cai Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Dong Road, Zhengzhou, 450002, People's Republic of China
| | - Miao-Miao Sun
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Henan Key Laboratory of Tumor Pathology, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, No.100 Ke Xue Avenue, Zhengzhou, 450002, People's Republic of China
| | - Kui-Sheng Chen
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Henan Key Laboratory of Tumor Pathology, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, No.100 Ke Xue Avenue, Zhengzhou, 450002, People's Republic of China
| |
Collapse
|
27
|
Rajeev V, Tabassum NI, Fann DY, Chen CP, Lai MK, Arumugam TV. Intermittent Metabolic Switching and Vascular Cognitive Impairment. J Obes Metab Syndr 2024; 33:92-107. [PMID: 38736362 PMCID: PMC11224924 DOI: 10.7570/jomes24010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/25/2024] [Accepted: 05/08/2024] [Indexed: 05/14/2024] Open
Abstract
Intermittent fasting (IF), a dietary pattern alternating between eating and fasting periods within a 24-hour cycle, has garnered recognition for its potential to enhance both healthspan and lifespan in animal models and humans. It also shows promise in alleviating age-related diseases, including neurodegeneration. Vascular cognitive impairment (VCI) spans a severity range from mild cognitive deficits to severe cognitive deficits and loss of function in vascular dementia. Chronic cerebral hypoperfusion has emerged as a significant contributor to VCI, instigating vascular pathologies such as microbleeds, blood-brain barrier dysfunction, neuronal loss, and white matter lesions. Preclinical studies in rodents strongly suggest that IF has the potential to attenuate pathological mechanisms, including excitotoxicity, oxidative stress, inflammation, and cell death pathways in VCI models. Hence, this supports evaluating IF in clinical trials for both existing and at-risk VCI patients. This review compiles existing data supporting IF's potential in treating VCI-related vascular and neuronal pathologies, emphasizing the mechanisms by which IF may mitigate these issues. Hence providing a comprehensive overview of the available data supporting IF's potential in treating VCI by emphasizing the underlying mechanisms that make IF a promising intervention for VCI.
Collapse
Affiliation(s)
- Vismitha Rajeev
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nishat I. Tabassum
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Australia
| | - David Y. Fann
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Christopher P. Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
| | - Mitchell K.P. Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
| | - Thiruma V. Arumugam
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Australia
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
28
|
Zheng ZJ, Zhu LZ, Qiu H, Zheng WYX, You PT, Chen SH, Hu CL, Huang JR, Zhou YJ. Neferine inhibits BMECs pyroptosis and maintains blood-brain barrier integrity in ischemic stroke by triggering a cascade reaction of PGC-1α. Sci Rep 2024; 14:14438. [PMID: 38910141 PMCID: PMC11194274 DOI: 10.1038/s41598-024-64815-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/13/2024] [Indexed: 06/25/2024] Open
Abstract
Blood-brain barrier disruption is a critical pathological event in the progression of ischemic stroke (IS). Most studies regarding the therapeutic potential of neferine (Nef) on IS have focused on neuroprotective effect. However, whether Nef attenuates BBB disruption during IS is unclear. We here used mice underwent transient middle cerebral artery occlusion (tMCAO) in vivo and bEnd.3 cells exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) injury in vitro to simulate cerebral ischemia. We showed that Nef reduced neurobehavioral dysfunction and protected brain microvascular endothelial cells and BBB integrity. Molecular docking, short interfering (Si) RNA and plasmid transfection results showed us that PGC-1α was the most binding affinity of biological activity protein for Nef. And verification experiments were showed that Nef upregulated PGC-1α expression to reduce mitochondrial oxidative stress and promote TJ proteins expression, further improves the integrity of BBB in mice. Intriguingly, our study showed that neferine is a natural PGC-1α activator and illustrated the mechanism of specific binding site. Furthermore, we have demonstrated Nef reduced mitochondria oxidative damage and ameliorates endothelial inflammation by inhibiting pyroptosis to improve BBB permeability through triggering a cascade reaction of PGC-1α via regulation of PGC-1α/NLRP3/GSDMD signaling pathway to maintain the integrity of BBB in ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Zi-Jian Zheng
- Department of Pharmacy, Gongan Hospital of Traditional Chinese Medicine, Jingzhou, 434300, China
- Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Li-Zhi Zhu
- Department of Pharmacy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, 518055, China
| | - Han Qiu
- Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, 3002 West Sungang Rd, Shenzhen, 518020, China
| | - Wu-Yin-Xiao Zheng
- Department of Pharmacy, Gongan Hospital of Traditional Chinese Medicine, Jingzhou, 434300, China
- Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Peng-Tao You
- Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Shu-He Chen
- Department of Pharmacy, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
- Hubei Shizhen Laboratory, Wuhan, 430061, China
| | - Chun-Ling Hu
- Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Jun-Rong Huang
- Department of Pharmacy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, 518055, China
| | - Ya-Jun Zhou
- Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, 3002 West Sungang Rd, Shenzhen, 518020, China.
- Department of Pharmacy, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China.
- Hubei Shizhen Laboratory, Wuhan, 430061, China.
| |
Collapse
|
29
|
Wang C, Liu Z, Cai J, Xu X. The regulatory effect of intermittent fasting on inflammasome activation in health and disease. Nutr Rev 2024; 82:978-987. [PMID: 37634143 DOI: 10.1093/nutrit/nuad104] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023] Open
Abstract
Intermittent fasting (IF), one of the most popular diets, can regulate inflammation and promote health; however, the detailed molecular mechanisms are not fully understood. The present review aims to provide an overview of recent preclinical and clinical studies that have examined the effect of IF on inflammasome signaling, and to discuss the translational gap between preclinical and clinical studies. Three databases (PubMed, Web of Science, and Embase) were searched to identify all relevant preclinical and clinical studies up to October 30, 2022. A total of 1544 studies were identified through the database searches, and 29 preclinical and 10 clinical studies were included. Twenty-three of the 29 preclinical studies reported that IF treatment could reduce inflammasome activation in neurological diseases, metabolic and cardiovascular diseases, immune and inflammatory diseases, gastrointestinal diseases, and pulmonary diseases, and 7 of the 10 clinical studies demonstrated reduced inflammasome activation after IF intervention in both healthy and obese participants. Among various IF regimens, time-restricted eating seemed to be the most effective one in terms of inflammasome regulation, and the efficacy of IF might increase over time. This review highlights the regulatory effect of IF on inflammasome activation in health and disease. Future studies using different IF regimens, in various populations, are needed in order to evaluate its potential to be used alone or as an adjunct therapy in humans to improve health and counteract diseases.
Collapse
Affiliation(s)
- Chenchen Wang
- Center for Molecular Metabolism, Nanjing University of Science and Technology, Nanjing, P. R. China
| | - Zhiqin Liu
- Center for Molecular Metabolism, Nanjing University of Science and Technology, Nanjing, P. R. China
| | - Jinpeng Cai
- Center for Molecular Metabolism, Nanjing University of Science and Technology, Nanjing, P. R. China
| | - Xi Xu
- Center for Molecular Metabolism, Nanjing University of Science and Technology, Nanjing, P. R. China
| |
Collapse
|
30
|
Zhang H, Shang J, Li W, Gao D, Zhang J. Increased Expression of VCAM1 on Brain Endothelial Cells Drives Blood-Brain Barrier Impairment Following Chronic Cerebral Hypoperfusion. ACS Chem Neurosci 2024; 15:2028-2041. [PMID: 38710594 PMCID: PMC11099957 DOI: 10.1021/acschemneuro.4c00039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/08/2024] Open
Abstract
Chronic cerebral hypoperfusion (CCH)-triggered blood-brain barrier (BBB) dysfunction is a core pathological change occurring in vascular dementia (VD). Despite the recent advances in the exploration of the structural basis of BBB impairment and the routes of entry of harmful compounds after a BBB leakage, the molecular mechanisms inducing BBB impairment remain largely unknown in terms of VD. Here, we employed a CCH-induced VD model and discovered increased vascular cell adhesion molecule 1 (VCAM1) expression on the brain endothelial cells (ECs). The expression of VCAM1 was directly correlated with the severity of BBB impairment. Moreover, the VCAM1 expression was associated with different regional white matter lesions. Furthermore, a compound that could block VCAM1 activation, K-7174, was also found to alleviate BBB leakage and protect the white matter integrity, whereas pharmacological manipulation of the BBB leakage did not affect the VCAM1 expression. Thus, our results demonstrated that VCAM1 is an important regulator that leads to BBB dysfunction following CCH. Blocking VCAM1-mediated BBB impairment may thus offer a new strategy to treat CCH-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Huiwen Zhang
- Department
of Neurology, Zhengzhou University People’s
Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan 450003, China
| | - Junkui Shang
- Department
of Neurology, Zhengzhou University People’s
Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan 450003, China
| | - Wei Li
- Department
of Neurology, Zhengzhou University People’s
Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan 450003, China
| | - Dandan Gao
- Department
of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430072, China
| | - Jiewen Zhang
- Department
of Neurology, Zhengzhou University People’s
Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan 450003, China
| |
Collapse
|
31
|
Lou T, Wu H, Feng M, Liu L, Yang X, Pan M, Wei Z, Zhang Y, Shi L, Qu B, Yang H, Cong S, Chen K, Liu J, Li Y, Jia Z, Xiao H. Integration of metabolomics and transcriptomics reveals that Da Chuanxiong Formula improves vascular cognitive impairment via ACSL4/GPX4 mediated ferroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117868. [PMID: 38325668 DOI: 10.1016/j.jep.2024.117868] [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: 11/30/2023] [Revised: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 02/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Da Chuanxiong Formula (DCX) is a traditional herbal compound composed of Gastrodia elata Bl. and Ligusticum chuanxiong Hort, which could significantly enhance blood circulation and neuroprotection, showing promise in treating Vascular Cognitive Impairment (VCI). AIM OF STUDY This study aims to elucidate the potential of DCX in treating VCI and its underlying mechanism. MATERIALS AND METHODS Firstly, the cognitive behavior level, blood flow changes, and brain pathology changes were evaluated through techniques such as the Morris water maze, step-down, laser speckle, coagulation analysis, and pathological staining to appraise the DCX efficacy. Then, the DCX targeting pathways were decoded by merging metabolomics with transcriptomics. Finally, the levels of reactive oxygen species (ROS), Fe2+, and lipid peroxidation related to the targeting signaling pathways of DCX were detected by kit, and the expression levels of mRNAs or proteins related to ferroptosis were determined by qPCR or Western blot assays respectively. RESULTS DCX improved cognitive abilities and cerebral perfusion significantly, and mitigated pathological damage in the hippocampal region of VCI model rats. Metabolomics revealed that DCX was able to call back 33 metabolites in plasma and 32 metabolites in brain samples, and the majority of the differential metabolites are phospholipid metabolites. Transcriptomic analysis revealed that DCX regulated a total of 3081 genes, with the ferroptosis pathway exhibiting the greatest impact. DCX inhibited ferroptosis of VCI rates by decreasing the levels of ferrous iron, ROS, and malondialdehyde (MDA) while increasing the level of superoxide dismutase (SOD) and glutathione (GSH) in VCI rats. Moreover, the mRNA and protein levels of ACSL4, LPCAT3, ALOX15, and GPX4, which are related to lipid metabolism in ferroptosis, were also regulated by DCX. CONCLUSION Our research findings indicated that DCX could inhibit ferroptosis through the ACSL4/GPX4 signaling pathway, thereby exerting its therapeutic benefits on VCI.
Collapse
Affiliation(s)
- Tianyu Lou
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Hao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Menghan Feng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Lirong Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoqin Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Mingxia Pan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zuying Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yinhuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China; Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
| | - Lixia Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Biqiong Qu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Haolan Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shiyu Cong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Kui Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Liu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yueting Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhixin Jia
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hongbin Xiao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China; Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
32
|
Xu F, Xin Q, Ren M, Shi P, Wang B. Inhibition of piezo1 prevents chronic cerebral hypoperfusion-induced cognitive impairment and blood brain barrier disruption. Neurochem Int 2024; 175:105702. [PMID: 38401846 DOI: 10.1016/j.neuint.2024.105702] [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: 07/26/2023] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 02/26/2024]
Abstract
Chronic cerebral hypoperfusion (CCH) plays a critical role in the onset and progression of vascular dementia (VD), which is now recognized as the second most common form of dementia after Alzheimer's disease (AD). The mechanosensitive piezo1 channel has been identified to play important roles in several neurological disorders. However, the roles and possible mechanisms of piezo1 in CCH-induced cognitive decline and blood brain barrier (BBB) disruption, as well as the underlying mechanisms remain elusive. In this study, the CCH model was established by bilateral common carotid artery occlusion in rats and by oxygen and glucose deprivation/reoxygenation (OGD/R) in bEnd.3 cells. The results demonstrated that the antagonist of piezo1 GsMTx4 ameliorated CCH-induced cognitive dysfunction and mitigated cerebral edema. Furthermore, this study indicated that GsMTx4 improved the permeability and integrity of BBB and protected cerebral microvasculature after CCH. In vitro, GsMTx4 improved cell viability, promoted the ability of cell motility and migration, and inhibited the degradation of BBB integrity-related proteins by inhibiting NLRP3 inflammasome activation. In addition, NLRP3 agonist abolished the beneficial effects of GsMTx4. Collectively, our results demonstrate that piezo1 might be involved in CCH-induced cognitive impairment and BBB damage, which may be at least partially mediated through regulation of NLRP3 inflammasome.
Collapse
Affiliation(s)
- Fei Xu
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Department of Vascular Surgery, Jining NO.1 People's Hospital, Jining, 272000, China
| | - Qing Xin
- Department of Physiology, Jining Medical University, Jining, 272000, China
| | - Mengyao Ren
- Department of Physiology, Jining Medical University, Jining, 272000, China
| | - Peixin Shi
- Department of Physiology, Jining Medical University, Jining, 272000, China
| | - Bing Wang
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
33
|
Wang Z, Han B, Qi J, Cao X, Gu H, Sun J. Chuanzhitongluo capsule improves cognitive impairment in mice with chronic cerebral hypoperfusion via the cholinergic anti-inflammatory pathway. Exp Gerontol 2024; 189:112407. [PMID: 38522309 DOI: 10.1016/j.exger.2024.112407] [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: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024]
Abstract
Vascular cognitive impairment (VCI) has become a common disease-causing cognitive deficit in humans, second only to Alzheimer's Disease (AD). Chuanzhitongluo capsule (CZTL) is a Traditional Chinese Medicine (TCM) preparation known for its effective protection against cerebral ischemia. However, its potential to ameliorate VCI remains unclear. This study aimed to investigate the cognitive improvement effects of CZTL in a mouse model of VCI. Chronic cerebral hypoperfusion (CCH) was induced in mice by bilateral common carotid artery stenosis (BCAS) to simulate the pathological changes associated with VCI. Spatial learning and memory abilities were assessed using the Morris Water Maze (MWM). RNA sequencing (RNA-Seq) was employed to identify differentially expressed genes (DEGs) in the hippocampus. Levels of inflammatory factors were measured through enzyme-linked immunosorbent assay (ELISA), while immunofluorescence (IF) determined the expression intensity of target proteins. Western Blot (WB) confirmed the final action pathway. Results indicated that CZTL significantly improved the spatial learning and memory abilities of CCH mice, along with alterations in gene expression profiles in the hippocampus. It also reduced neuroinflammation in the hippocampus and upregulated the choline acetyltransferase (ChAT) and α7 subunit-containing nicotinic acetylcholine receptor (α7nAChR), which are in synaptic plasticity and neuronal development. Moreover, CZTL inhibited the NF-κB signaling pathway. In conclusion, CZTL may alleviate neuroinflammation induced by CCH and improve cognitive impairment in CCH mice by regulating the cholinergic anti-inflammatory pathway (CAIP) involving ChAT/α7nAChR/NF-κB.
Collapse
Affiliation(s)
- Zhiyuan Wang
- Institute of Integrative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Han
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianjiao Qi
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xuelei Cao
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Huali Gu
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Jinping Sun
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
34
|
Lim YA, Tan LS, Lee WT, Sim WL, Lv Y, Takakuni M, Saito S, Ihara M, Arumugam TV, Chen C, Wong FWS, Dawe GS. Hope for vascular cognitive impairment: Ac-YVAD-cmk as a novel treatment against white matter rarefaction. PLoS One 2024; 19:e0299703. [PMID: 38630707 PMCID: PMC11023579 DOI: 10.1371/journal.pone.0299703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 02/14/2024] [Indexed: 04/19/2024] Open
Abstract
Vascular cognitive impairment (VCI) is the second leading cause of dementia with limited treatment options, characterised by cerebral hypoperfusion-induced white matter rarefaction (WMR). Subcortical VCI is the most common form of VCI, but the underlying reasons for region susceptibility remain elusive. Recent studies employing the bilateral cortical artery stenosis (BCAS) method demonstrate that various inflammasomes regulate white matter injury and blood-brain barrier dysfunction but whether caspase-1 inhibition will be beneficial remains unclear. To address this, we performed BCAS on C57/BL6 mice to study the effects of Ac-YVAD-cmk, a caspase-1 inhibitor, on the subcortical and cortical regions. Cerebral blood flow (CBF), WMR, neuroinflammation and the expression of tight junction-related proteins associated with blood-brain barrier integrity were assessed 15 days post BCAS. We observed that Ac-YVAD-cmk restored CBF, attenuated BCAS-induced WMR and restored subcortical myelin expression. Within the subcortical region, BCAS activated the NLRP3/caspase-1/interleukin-1beta axis only within the subcortical region, which was attenuated by Ac-YVAD-cmk. Although we observed that BCAS induced significant increases in VCAM-1 expression in both brain regions that were attenuated with Ac-YVAD-cmk, only ZO-1 and occludin were observed to be significantly altered in the subcortical region. Here we show that caspase-1 may contribute to subcortical regional susceptibility in a mouse model of VCI. In addition, our results support further investigations into the potential of Ac-YVAD-cmk as a novel treatment strategy against subcortical VCI and other conditions exhibiting cerebral hypoperfusion-induced WMR.
Collapse
Affiliation(s)
- Yun-An Lim
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Li Si Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Thye Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Liang Sim
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Yang Lv
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Maki Takakuni
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Saito
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
- Japan Society for the Promotion of Science, Chiyoda-ku, Tokyo, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | | | - Christopher Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Fred Wai-Shiu Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gavin Stewart Dawe
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
35
|
Rahman MS, Islam R, Bhuiyan MIH. Ion transporter cascade, reactive astrogliosis and cerebrovascular diseases. Front Pharmacol 2024; 15:1374408. [PMID: 38659577 PMCID: PMC11041382 DOI: 10.3389/fphar.2024.1374408] [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: 01/22/2024] [Accepted: 03/21/2024] [Indexed: 04/26/2024] Open
Abstract
Cerebrovascular diseases and their sequalae, such as ischemic stroke, chronic cerebral hypoperfusion, and vascular dementia are significant contributors to adult disability and cognitive impairment in the modern world. Astrocytes are an integral part of the neurovascular unit in the CNS and play a pivotal role in CNS homeostasis, including ionic and pH balance, neurotransmission, cerebral blood flow, and metabolism. Astrocytes respond to cerebral insults, inflammation, and diseases through unique molecular, morphological, and functional changes, collectively known as reactive astrogliosis. The function of reactive astrocytes has been a subject of debate. Initially, astrocytes were thought to primarily play a supportive role in maintaining the structure and function of the nervous system. However, recent studies suggest that reactive astrocytes may have both beneficial and detrimental effects. For example, in chronic cerebral hypoperfusion, reactive astrocytes can cause oligodendrocyte death and demyelination. In this review, we will summarize the (1) roles of ion transporter cascade in reactive astrogliosis, (2) role of reactive astrocytes in vascular dementia and related dementias, and (3) potential therapeutic approaches for dementing disorders targeting reactive astrocytes. Understanding the relationship between ion transporter cascade, reactive astrogliosis, and cerebrovascular diseases may reveal mechanisms and targets for the development of therapies for brain diseases associated with reactive astrogliosis.
Collapse
Affiliation(s)
- Md Shamim Rahman
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX, United States
| | | | - Mohammad Iqbal H. Bhuiyan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX, United States
| |
Collapse
|
36
|
Chong JR, Chai YL, Yam ATY, Hilal S, Vrooman H, Venketasubramanian N, Blennow K, Zetterberg H, Ashton NJ, Chen CP, Lai MKP. Association of plasma GFAP with elevated brain amyloid is dependent on severity of white matter lesions in an Asian cognitively impaired cohort. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12576. [PMID: 38605996 PMCID: PMC11007806 DOI: 10.1002/dad2.12576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/25/2024] [Accepted: 03/01/2024] [Indexed: 04/13/2024]
Abstract
INTRODUCTION While elevated blood glial fibrillary acidic protein (GFAP) has been associated with brain amyloid pathology, whether this association occurs in populations with high cerebral small vessel disease (CSVD) concomitance remains unclear. METHODS Using a Singapore-based cohort of cognitively impaired subjects, we assessed associations between plasma GFAP and neuroimaging measures of brain amyloid and CSVD, including white matter hyperintensities (WMH). We also examined the diagnostic performance of plasma GFAP in detecting brain amyloid beta positivity (Aβ+). RESULTS When stratified by WMH status, elevated brain amyloid was associated with higher plasma GFAP only in the WMH- group (β = 0.383; P < 0.001). The diagnostic performance of plasma GFAP in identifying Aβ+ was significantly higher in the WMH- group (area under the curve [AUC] = 0.896) than in the WMH+ group (AUC = 0.712, P = 0.008). DISCUSSION The biomarker utility of plasma GFAP in detecting brain amyloid pathology is dependent on the severity of concomitant WMH. Highlight Glial fibrillary acidic protein (GFAP)'s association with brain amyloid is unclear in populations with high cerebral small vessel disease (CSVD).Plasma GFAP was measured in a cohort with CSVD and brain amyloid.Plasma GFAP was better in detecting amyloid in patients with low CSVD versus high CSVD.Biomarker utility of GFAP in detecting brain amyloid depends on the severity of CSVD.
Collapse
Affiliation(s)
- Joyce R. Chong
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| | - Yuek Ling Chai
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| | - Amelia T. Y. Yam
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| | - Saima Hilal
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
- Saw Swee Hock School of Public HealthNational University of Singapore and National University Health SystemKent RidgeSingapore
- Department of Radiology and Nuclear MedicineErasmus Medical CenterRotterdamthe Netherlands
| | - Henri Vrooman
- Department of Radiology and Nuclear MedicineErasmus Medical CenterRotterdamthe Netherlands
| | | | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGöteborgSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGöteborgSweden
- Department of Neurodegenerative DiseaseThe UCL Queen Square Institute of NeurologyLondonUK
| | - Nicholas J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGöteborgSweden
| | - Christopher P. Chen
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| | - Mitchell K. P. Lai
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| |
Collapse
|
37
|
Jearjaroen P, Thangwong P, Tocharus C, Chaichompoo W, Suksamrarn A, Tocharus J. Hexahydrocurcumin attenuated demyelination and improved cognitive impairment in chronic cerebral hypoperfusion rats. Inflammopharmacology 2024; 32:1531-1544. [PMID: 38153537 DOI: 10.1007/s10787-023-01406-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/27/2023] [Indexed: 12/29/2023]
Abstract
Age-related white matter lesions (WML) frequently present vascular problems by decreasing cerebral blood supply, resulting in the condition known as chronic cerebral hypoperfusion (CCH). This study aimed to investigate the effect of hexahydrocurcumin (HHC) on the processes of demyelination and remyelination induced by the model of the Bilateral Common Carotid Artery Occlusion (BCCAO) for 29 days to mimic the CCH condition. The pathological appearance of myelin integrity was significantly altered by CCH, as evidenced by Transmission Electron Microscopy (TEM) and Luxol Fast Blue (LFB) staining. In addition, CCH activated A1-astrocytes and reactive-microglia by increasing the expression of Glial fibrillary acidic protein (GFAP), complement 3 (C3d) and pro-inflammatory cytokines. However, S100a10 expression, a marker of neuroprotective astrocytes, was suppressed, as were regenerative factors including (IGF-1) and Transglutaminase 2 (TGM2). Therefore, the maturation step was obstructed as shown by decreases in the levels of myelin basic protein (MBP) and the proteins related with lipid synthesis. Cognitive function was therefore impaired in the CCH model, as evidenced by the Morris water maze test. By contrast, HHC treatment significantly improved myelin integrity, and inhibited A1-astrocytes and reactive-microglial activity. Consequently, pro-inflammatory cytokines and A1-astrocytes were attenuated, and regenerative factors increased assisting myelin maturation and hence improving cognitive performance. In conclusion, HHC improves cognitive function and also the integrity of white matter in CCH rats by reducing demyelination, and pro-inflammatory cytokine production and promoting the process of remyelination.
Collapse
Affiliation(s)
- Pranglada Jearjaroen
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Phakkawat Thangwong
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chianqg Mai, Thailand
| | - Waraluck Chaichompoo
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Apichart Suksamrarn
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Functional Food Research Center for Well-Being, Multidisciplinary Research Institute, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
38
|
Xie Y, Han R, Li Y, Li W, Zhang S, Wu Y, Zhao Y, Liu R, Wu J, Jiang W, Chen X. P2X7 receptor antagonists modulate experimental autoimmune neuritis via regulation of NLRP3 inflammasome activation and Th17 and Th1 cell differentiation. J Neuroinflammation 2024; 21:73. [PMID: 38528529 PMCID: PMC10964508 DOI: 10.1186/s12974-024-03057-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/27/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Guillain-Barré syndrome (GBS), a post-infectious, immune-mediated, acute demyelinating disease of the peripheral nerves and nerve roots, represents the most prevalent and severe acute paralyzing neuropathy. Purinergic P2X7 receptors (P2X7R) play a crucial role in central nervous system inflammation. However, little is known about their role in the immune-inflammatory response within the peripheral nervous system. METHODS Initially, we assessed the expression of purinergic P2X7R in the peripheral blood of patients with GBS using flow cytometry and qRT-PCR. Next, we explored the expression of P2 X7R in CD4+ T cells, CD8+ T cells, and macrophages within the sciatic nerves and spleens of rats using immunofluorescence labeling and flow cytometry. The P2X7R antagonist brilliant blue G (BBG) was employed to examine its therapeutic impact on rats with experimental autoimmune neuritis (EAN) induced by immunization with the P0180 - 199 peptide. We analyzed CD4+ T cell differentiation in splenic mononuclear cells using flow cytometry, assessed Th17 cell differentiation in the sciatic nerve through immunofluorescence staining, and examined the expression of pro-inflammatory cytokine mRNA using RT-PCR. Additionally, we performed protein blotting to assess the expression of P2X7R and NLRP3-related inflammatory proteins within the sciatic nerve. Lastly, we utilized flow cytometry and immunofluorescence labeling to examine the expression of NLRP3 on CD4+ T cells in rats with EAN. RESULTS P2X7R expression was elevated not only in the peripheral blood of patients with GBS but also in rats with EAN. In rats with EAN, inhibiting P2X7R with BBG alleviated neurological symptoms, reduced demyelination, decreased inflammatory cell infiltration of the peripheral nerves, and improved nerve conduction. BBG also limited the production of pro-inflammatory molecules, down-regulated the expression of P2X7R and NLRP3, and suppressed the differentiation of Th1 and Th17 cells, thus protecting against EAN. These effects collectively contribute to modifying the inflammatory environment and enhancing outcomes in EAN rats. CONCLUSIONS Suppression of P2X7R relieved EAN manifestation by regulating CD4+ T cell differentiation and NLRP3 inflammasome activation. This finding underscores the potential significance of P2X7R as a target for anti-inflammatory treatments, advancing research and management of GBS.
Collapse
Affiliation(s)
- Yuhan Xie
- Department of Neurology, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300052, China
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Ranran Han
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yulin Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Weiya Li
- Department of Neurology, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300052, China
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Shichao Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300102, China
| | - Yu Wu
- Department of Neurology, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Yuexin Zhao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Rongrong Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jie Wu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Wei Jiang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Xiuju Chen
- Department of Neurology, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300052, China.
| |
Collapse
|
39
|
Qu J, Wu L, Zhang M, Kan M, Chen H, Shi Y, Wang S, Wang X, Chen F. Serum autophagy-related gene 5 level in stroke patients: correlation with CD4+ T cells and cognition impairment during a 3-year follow-up. Braz J Med Biol Res 2024; 57:e13019. [PMID: 38511768 PMCID: PMC10946239 DOI: 10.1590/1414-431x2024e13019] [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: 05/19/2023] [Accepted: 02/04/2024] [Indexed: 03/22/2024] Open
Abstract
Autophagy-related gene (ATG) 5 regulates blood lipids, chronic inflammation, CD4+ T-cell differentiation, and neuronal death and is involved in post-stroke cognitive impairment. This study aimed to explore the correlation of serum ATG5 with CD4+ T cells and cognition impairment in stroke patients. Peripheral blood was collected from 180 stroke patients for serum ATG5 and T helper (Th) 1, Th2, Th17, and regulatory T (Treg) cell detection via enzyme-linked immunosorbent assays and flow cytometry. The Mini-Mental State Examination (MMSE) scale was completed at enrollment, year (Y)1, Y2, and Y3 in stroke patients. Serum ATG5 was also measured in 50 healthy controls (HCs). Serum ATG5 was elevated in stroke patients compared to HCs (P<0.001) and was positively correlated to Th2 cells (P=0.022), Th17 cells (P<0.001), and Th17/Treg ratio (P<0.001) in stroke patients but not correlated with Th1 cells, Th1/Th2 ratio, or Treg cells (all P>0.050). Serum ATG5 (P=0.037), Th1 cells (P=0.022), Th17 cells (P=0.002), and Th17/Treg ratio (P=0.018) were elevated in stroke patients with MMSE score-identified cognition impairment vs those without cognition impairment, whereas Th2 cells, Th1/Th2 ratio, and Treg cells were not different between them (all P>0.050). Importantly, serum ATG5 was negatively linked with MMSE score at enrollment (P=0.004), Y1 (P=0.002), Y2 (P=0.014), and Y3 (P=0.001); moreover, it was positively related to 2-year (P=0.024) and 3-year (P=0.012) MMSE score decline in stroke patients. Serum ATG5 was positively correlated with Th2 and Th17 cells and estimated cognitive function decline in stroke patients.
Collapse
Affiliation(s)
- Juanjuan Qu
- Department of Emergency, Handan Central Hospital, Handan, China
| | - Linxia Wu
- Department of Emergency, Handan Central Hospital, Handan, China
| | - Meng Zhang
- Department of Emergency, Handan Central Hospital, Handan, China
| | - Minchen Kan
- Department of Emergency, Handan Central Hospital, Handan, China
| | - Huimin Chen
- Department of Emergency, Handan Central Hospital, Handan, China
| | - Yanqing Shi
- Department of Emergency, Handan Central Hospital, Handan, China
| | - Shuangyu Wang
- Department of Nephrology, Handan Central Hospital, Handan, China
| | - Xiaohua Wang
- Department of Neonatal Ward, Handan Central Hospital, Handan, China
| | - Fan Chen
- Department of Emergency, Handan Central Hospital, Handan, China
| |
Collapse
|
40
|
Wang Y, Cheng W, Chen X, Cheng C, Zhang L, Huang W. Serum Proteomics Identified TAFI as a Potential Molecule Facilitating the Migration of Peripheral Monocytes to Damaged White Matter During Chronic Cerebral Hypoperfusion. Neurochem Res 2024; 49:597-616. [PMID: 37978153 DOI: 10.1007/s11064-023-04050-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 11/19/2023]
Abstract
Neuroinflammation is assumed as the critical pathophysiologic mechanism of white matter lesions (WMLs), and infiltrated peripheral monocyte-derived macrophages are implicated in the development of neuroinflammation. This study sought to explore the blood molecules that promote the migration of peripheral monocytes to the sites of WMLs. The serum protein expression profiles of patients and Sprague-Dawley rat models with WMLs were detected by data-independent acquisition (DIA) proteomics technique. Compared with corresponding control groups, we acquired 62 and 41 differentially expressed proteins (DEPs) in the serum of patients and model rats with WMLs respectively. Bioinformatics investigations demonstrated that these DEPs were linked to various Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and Gene Ontology (GO) terms involved in neuroinflammation. Afterward, we identified thrombin-activatable fibrinolysis inhibitor (TAFI) as a shared and overexpressed protein in clinical and animal serum samples, which was further verified by enzyme-linked immunosorbent assay. Additionally, an upregulation of TAFI was also observed in the white matter of rat models, and the inhibition of TAFI impeded the migration of peripheral monocytes to the area of WMLs. In vitro experiments suggested that TAFI could enhance the migration ability of RAW264.7 cells and increase the expression of Ccr2. Our study demonstrates that neuroinflammatory signals can be detected in the peripheral blood of WMLs patients and model rats. TAFI may serve as a potential protein that promotes the migration of peripheral monocytes to WMLs regions, thereby providing a novel molecular target for further investigation into the interaction between the central and peripheral immune systems.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Wenchao Cheng
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiuying Chen
- Department of Neurology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Chang Cheng
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Lan Zhang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Wen Huang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China.
- Department of Neurology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing, China.
| |
Collapse
|
41
|
Cheng W, Wang Y, Zhang L, Cheng C, Chen X, Huang W. The Impact of Dipyridamole on Disease-Associated Microglia Phenotypic Transformation in White Matter Lesions Induced by Chronic Cerebral Hypoperfusion. Neurochem Res 2024; 49:744-757. [PMID: 38102341 DOI: 10.1007/s11064-023-04066-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/31/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023]
Abstract
White matter lesions (WMLs) resulting from chronic cerebral hypoperfusion (CCH) are the leading cause of vascular dementia (VaD). This study aimed to investigate whether dipyridamole could alleviate WMLs by regulating the phenotype of disease-associated microglia (DAM) through equilibrative nucleoside transporter 2 (ENT2) and adenosine A2A receptor (Adora2a) and to clarify the underlying molecular mechanisms. CCH rat models were constructed to mimic VaD. Morris water maze and Luxol Fast Blue staining were employed to assess cognitive function and quantify the severity of WMLs, respectively. Immunofluorescent staining was performed to analyze the activation of glial cells and the phenotypic transformation of DAM. Additionally, levels of ENT2, proteins in the NF-κB and ERK1/2 pathways and inflammatory cytokines were detected. The results indicated that dipyridamole diminished the activation and proliferation of microglia and astrocytes, increased the expression of myelin basic protein and ameliorated WMLs and cognitive decline in CCH rats. Further study revealed that dipyridamole decreased the expression of ENT2 and inhibited the activation of ERK1/2 and NF-κB signaling pathways, which ultimately converted DAM to anti-inflammatory phenotype and suppressed the levels of TNF-α, IL-1β, IL-6 in WMLs. However, Adora2a inhibitor (SCH58261) attenuated above effects. Our study demonstrates that dipyridamole facilitates the conversion of DAM to the anti-inflammatory phenotype through ENT2/Adora2a pathway and inhibits the activation of ERK1/2 and NF-κB signaling pathways, thereby alleviating neuroinflammation in WMLs. The current findings establish the basis for using dipyridamole to treat VaD.
Collapse
Affiliation(s)
- Wenchao Cheng
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Yuhan Wang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Lan Zhang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Chang Cheng
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiuying Chen
- Department of Neurology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Wen Huang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China.
- Department of Neurology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing, China.
| |
Collapse
|
42
|
Pavuluri K, Huston J, Ehman RL, Manduca A, Jack CR, Senjem ML, Vemuri P, Murphy MC. Associations between vascular health, brain stiffness and global cognitive function. Brain Commun 2024; 6:fcae073. [PMID: 38505229 PMCID: PMC10950054 DOI: 10.1093/braincomms/fcae073] [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: 07/13/2023] [Revised: 12/19/2023] [Accepted: 02/27/2024] [Indexed: 03/21/2024] Open
Abstract
Vascular brain injury results in loss of structural and functional connectivity and leads to cognitive impairment. Its various manifestations, including microinfarcts, microhaemorrhages and white matter hyperintensities, result in microstructural tissue integrity loss and secondary neurodegeneration. Among these, tissue microstructural alteration is a relatively early event compared with atrophy along the aging and neurodegeneration continuum. Understanding its association with cognition may provide the opportunity to further elucidate the relationship between vascular health and clinical outcomes. Magnetic resonance elastography offers a non-invasive approach to evaluate tissue mechanical properties, providing a window into the microstructural integrity of the brain. This retrospective study evaluated brain stiffness as a potential biomarker for vascular brain injury and its role in mediating the impact of vascular dysfunction on cognitive impairment. Seventy-five participants from the Mayo Clinic Study of Aging underwent brain imaging using a 3T MR imager with a spin-echo echo-planar imaging sequence for magnetic resonance elastography and T1- and T2-weighted pulse sequences. This study evaluated the effects of vascular biomarkers (white matter hyperintensities and cardiometabolic condition score) on brain stiffness using voxelwise analysis. Partial correlation analysis explored associations between brain stiffness, white matter hyperintensities, cardiometabolic condition and global cognition. Mediation analysis determined the role of stiffness in mediating the relationship between vascular biomarkers and cognitive performance. Statistical significance was set at P-values < 0.05. Diagnostic accuracy of magnetic resonance elastography stiffness for white matter hyperintensities and cardiometabolic condition was evaluated using receiver operator characteristic curves. Voxelwise linear regression analysis indicated white matter hyperintensities negatively correlate with brain stiffness, specifically in periventricular regions with high white matter hyperintensity levels. A negative association between cardiovascular risk factors and stiffness was also observed across the brain. No significant patterns of stiffness changes were associated with amyloid load. Global stiffness (µ) negatively correlated with both white matter hyperintensities and cardiometabolic condition when all other covariables including amyloid load were controlled. The positive correlation between white matter hyperintensities and cardiometabolic condition weakened and became statistically insignificant when controlling for other covariables. Brain stiffness and global cognition were positively correlated, maintaining statistical significance after adjusting for all covariables. These findings suggest mechanical alterations are associated with cognitive dysfunction and vascular brain injury. Brain stiffness significantly mediated the indirect effects of white matter hyperintensities and cardiometabolic condition on global cognition. Local cerebrovascular diseases (assessed by white matter hyperintensities) and systemic vascular risk factors (assessed by cardiometabolic condition) impact brain stiffness with spatially and statistically distinct effects. Global brain stiffness is a significant mediator between vascular disease measures and cognitive function, highlighting the value of magnetic resonance elastography-based mechanical assessments in understanding this relationship.
Collapse
Affiliation(s)
| | - John Huston
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Richard L Ehman
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Armando Manduca
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthew L Senjem
- Department of Information Technology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | |
Collapse
|
43
|
Zeylan M, Senyuz S, Picón-Pagès P, García-Elías A, Tajes M, Muñoz FJ, Oliva B, Garcia-Ojalvo J, Barbu E, Vicente R, Nattel S, Ois A, Puig-Pijoan A, Keskin O, Gursoy A. Shared Proteins and Pathways of Cardiovascular and Cognitive Diseases: Relation to Vascular Cognitive Impairment. J Proteome Res 2024; 23:560-573. [PMID: 38252700 PMCID: PMC10846560 DOI: 10.1021/acs.jproteome.3c00289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/29/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024]
Abstract
One of the primary goals of systems medicine is the detection of putative proteins and pathways involved in disease progression and pathological phenotypes. Vascular cognitive impairment (VCI) is a heterogeneous condition manifesting as cognitive impairment resulting from vascular factors. The precise mechanisms underlying this relationship remain unclear, which poses challenges for experimental research. Here, we applied computational approaches like systems biology to unveil and select relevant proteins and pathways related to VCI by studying the crosstalk between cardiovascular and cognitive diseases. In addition, we specifically included signals related to oxidative stress, a common etiologic factor tightly linked to aging, a major determinant of VCI. Our results show that pathways associated with oxidative stress are quite relevant, as most of the prioritized vascular cognitive genes and proteins were enriched in these pathways. Our analysis provided a short list of proteins that could be contributing to VCI: DOLK, TSC1, ATP1A1, MAPK14, YWHAZ, CREB3, HSPB1, PRDX6, and LMNA. Moreover, our experimental results suggest a high implication of glycative stress, generating oxidative processes and post-translational protein modifications through advanced glycation end-products (AGEs). We propose that these products interact with their specific receptors (RAGE) and Notch signaling to contribute to the etiology of VCI.
Collapse
Affiliation(s)
- Melisa
E. Zeylan
- Computational
Sciences and Engineering, Graduate School of Science and Engineering, Koç University, Istanbul 34450, Türkiye
| | - Simge Senyuz
- Computational
Sciences and Engineering, Graduate School of Science and Engineering, Koç University, Istanbul 34450, Türkiye
| | - Pol Picón-Pagès
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Anna García-Elías
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Marta Tajes
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Francisco J. Muñoz
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Baldomero Oliva
- Laboratory
of Structural Bioinformatics (GRIB), Department of Medicine and Life
Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Jordi Garcia-Ojalvo
- Laboratory
of Dynamical Systems Biology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Eduard Barbu
- Institute
of Computer Science, University of Tartu, Tartu, 50090, Estonia
| | - Raul Vicente
- Institute
of Computer Science, University of Tartu, Tartu, 50090, Estonia
| | - Stanley Nattel
- Department
of Medicine and Research Center, Montreal Heart Institute and Université
de Montréal; Institute of Pharmacology, West German Heart and
Vascular Center, University Duisburg-Essen,
Germany; IHU LIRYC and Fondation Bordeaux Université, Bordeaux 33000, France
| | - Angel Ois
- Department
of Neurology, Hospital Del Mar. Hospital
Del Mar - Medical Research Institute and Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Albert Puig-Pijoan
- Department
of Neurology, Hospital Del Mar. Hospital
Del Mar - Medical Research Institute and Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Ozlem Keskin
- Department
of Chemical and Biological Engineering, Koç University, Istanbul 34450, Türkiye
| | - Attila Gursoy
- Department
of Computer Engineering, Koç University, Istanbul 34450, Türkiye
| |
Collapse
|
44
|
Meng J, Han L, Xu H, Zhang L, Liu Z, Zhou Y, Zhang X, Luo H, Zhang YW. TREM2 regulates microglial phagocytosis of synapses in innate immune tolerance. Int Immunopharmacol 2024; 127:111445. [PMID: 38147777 DOI: 10.1016/j.intimp.2023.111445] [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: 11/08/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 12/28/2023]
Abstract
Increasing evidence indicates that innate immune cells also possess immunological memory. Microglia are brain-resident innate immune cells and execute inflammatory and phagocytic functions upon environmental stimulation, during which processes triggering receptor expressed on myeloid cells 2 (TREM2) plays an important regulatory role. However, although microglia are known to exhibit innate immune memory related to inflammation when subjected to continuous inflammatory stimuli, whether microglia exhibit innate immune memory related to phagocytosis and whether TREM2 participates in innate immune memory of microglia remain unknown. Herein, we treated WT and Trem2 KO mice with peripheral injection of lipopolysaccharides (LPS) to induce microglial activation or microglial immune tolerance. We found that Tnfα and Il-1β expression levels in the hippocampi were significantly elevated after 1xLPS and then dramatically decreased after 4xLPS in both WT and Trem2 KO mice; and their level changes were indistinguishable between WT and Trem2 KO mice. Moreover, 1xLPS significantly promoted microglial phagocytosis of synapses and caused microglial morphology changes resembling activated status in both WT and Trem2 KO mice. However, 4xLPS significantly reduced synapse phagocytosis and largely reversed morphology changes in WT microglia. While 4xLPS had no effect on reducing synapse phagocytosis in Trem2 KO microglia. RNA-seq analysis revealed that TREM2 deficiency reprogrammed complement and phagosome-related transcriptional landscape during immune tolerance. Our results demonstrate that microglia also exhibit immune tolerance related to phagocytosis of synapses and that TREM2 plays a crucial role in this process possibly through regulating complement system and phagosome-related gene expressions.
Collapse
Affiliation(s)
- Jian Meng
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| | - Linkun Han
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Hui Xu
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Lingliang Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhaoji Liu
- Department of Neurology, Zhongshan Hospital Xiamen University, Xiamen, Fujian 361004, China
| | - Yunqiang Zhou
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Xian Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Hong Luo
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yun-Wu Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
45
|
Zhan T, Tang S, Du J, Liu J, Yu B, Yang Y, Xie Y, Qiu Y, Li G, Gao Y. Implication of lncRNA MSTRG.81401 in Hippocampal Pyroptosis Induced by P2X7 Receptor in Type 2 Diabetic Rats with Neuropathic Pain Combined with Depression. Int J Mol Sci 2024; 25:1186. [PMID: 38256257 PMCID: PMC10816120 DOI: 10.3390/ijms25021186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/02/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
Major depressive disorder (MDD) is a common complication of diabetes and is often observed alongside diabetic neuropathic pain (DNP) as a comorbidity in diabetic patients. Long non-coding RNA (lncRNA) plays an important role in various pathophysiological processes. The P2X7 receptor is responsible for triggering inflammatory responses, such as pyroptosis, linked to pain and depression. The aim of this study was to investigate the effect of lncRNA MSTRG.81401 on hippocampal pyroptosis induced by the P2X7 receptor in diabetic rats with DNP combined with MDD (DNP + MDD). Our results showed that the expression of lncRNA MSTRG.81401 was significantly elevated in the hippocampus of DNP + MDD rats compared with the control group. Following the administration of shRNA targeting lncRNA MSTRG.81401, a notable elevation in mechanical and thermal pain thresholds was observed in rats with comorbid DNP and MDD. Additionally, significant improvements in depression-like behaviors were evident in the open-field test (OFT), sucrose preference test (SPT), and forced swim test (FST). In the DNP + MDD rats, elevated levels in hippocampal P2X7 receptor mRNA and protein were observed, along with increased co-expression of P2X7 and the astrocytic marker glial fibrillary acidic protein (GFAP). Meanwhile, in DNP + MDD rats, the heightened mRNA expression of NOD-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein (ASC), pyroptosis-related protein Gasdermin D (GSDMD), caspase-1, IL-1β, IL-18, and TNF-α was detected, in addition to increased serum levels of IL-1β, IL-18 and TNF-α. After shRNA treatment with lncRNA MSTRG.81401, the above abnormal changes in indicators for pyroptosis and inflammation were improved. Therefore, our study demonstrates that shRNA of lncRNA MSTRG.81401 can alleviate the pain and depression-like behaviors in diabetic rats associated with the comorbidity of DNP and MDD by inhibiting the hippocampal P2X7 receptor-mediated pyroptosis pathway and pro-inflammatory responses. This suggests that the P2X7R/NLRP3/caspase-1 implicated pyroptosis and inflammatory scenario may serve as a potential target for the management of comorbid DNP and MDD in diabetes.
Collapse
Affiliation(s)
- Ting Zhan
- Department of Physiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (T.Z.); (S.T.); (J.D.); (Y.Y.); (Y.X.); (Y.Q.); (G.L.)
| | - Shanshan Tang
- Department of Physiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (T.Z.); (S.T.); (J.D.); (Y.Y.); (Y.X.); (Y.Q.); (G.L.)
| | - Junpei Du
- Department of Physiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (T.Z.); (S.T.); (J.D.); (Y.Y.); (Y.X.); (Y.Q.); (G.L.)
| | - Jingshuang Liu
- Joint Program of Nanchang University and Queen Mary University of London, Nanchang University, Nanchang 330006, China;
| | - Bodong Yu
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China;
| | - Yuxin Yang
- Department of Physiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (T.Z.); (S.T.); (J.D.); (Y.Y.); (Y.X.); (Y.Q.); (G.L.)
| | - Yuting Xie
- Department of Physiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (T.Z.); (S.T.); (J.D.); (Y.Y.); (Y.X.); (Y.Q.); (G.L.)
| | - Yanting Qiu
- Department of Physiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (T.Z.); (S.T.); (J.D.); (Y.Y.); (Y.X.); (Y.Q.); (G.L.)
| | - Guodong Li
- Department of Physiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (T.Z.); (S.T.); (J.D.); (Y.Y.); (Y.X.); (Y.Q.); (G.L.)
| | - Yun Gao
- Department of Physiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (T.Z.); (S.T.); (J.D.); (Y.Y.); (Y.X.); (Y.Q.); (G.L.)
- Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang 330006, China
| |
Collapse
|
46
|
Kalaria RN, Akinyemi RO, Paddick SM, Ihara M. Current perspectives on prevention of vascular cognitive impairment and promotion of vascular brain health. Expert Rev Neurother 2024; 24:25-44. [PMID: 37916306 PMCID: PMC10872925 DOI: 10.1080/14737175.2023.2273393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/17/2023] [Indexed: 11/03/2023]
Abstract
INTRODUCTION The true global burden of vascular cognitive impairment (VCI) is unknown. Reducing risk factors for stroke and cardiovascular disease would inevitably curtail VCI. AREAS COVERED The authors review current diagnosis, epidemiology, and risk factors for VCI. VCI increases in older age and by inheritance of known genetic traits. They emphasize modifiable risk factors identified by the 2020 Lancet Dementia Commission. The most profound risks for VCI also include lower education, cardiometabolic factors, and compromised cognitive reserve. Finally, they discuss pharmacological and non-pharmacological interventions. EXPERT OPINION By virtue of the high frequencies of stroke and cardiovascular disease the global prevalence of VCI is expectedly higher than prevalent neurodegenerative disorders causing dementia. Since ~ 90% of the global burden of stroke can be attributed to modifiable risk factors, a formidable opportunity arises to reduce the burden of not only stroke but VCI outcomes including progression from mild to the major in form of vascular dementia. Strict control of vascular risk factors and secondary prevention of cerebrovascular disease via pharmacological interventions will impact on burden of VCI. Non-pharmacological measures by adopting healthy diets and encouraging physical and cognitive activities and urging multidomain approaches are important for prevention of VCI and preservation of vascular brain health.
Collapse
Affiliation(s)
- Raj N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Rufus O Akinyemi
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Stella-Maria Paddick
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Centre, Osaka, Japan
| |
Collapse
|
47
|
Gusev E, Sarapultsev A. Interplay of G-proteins and Serotonin in the Neuroimmunoinflammatory Model of Chronic Stress and Depression: A Narrative Review. Curr Pharm Des 2024; 30:180-214. [PMID: 38151838 DOI: 10.2174/0113816128285578231218102020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/29/2023] [Indexed: 12/29/2023]
Abstract
INTRODUCTION This narrative review addresses the clinical challenges in stress-related disorders such as depression, focusing on the interplay between neuron-specific and pro-inflammatory mechanisms at the cellular, cerebral, and systemic levels. OBJECTIVE We aim to elucidate the molecular mechanisms linking chronic psychological stress with low-grade neuroinflammation in key brain regions, particularly focusing on the roles of G proteins and serotonin (5-HT) receptors. METHODS This comprehensive review of the literature employs systematic, narrative, and scoping review methodologies, combined with systemic approaches to general pathology. It synthesizes current research on shared signaling pathways involved in stress responses and neuroinflammation, including calcium-dependent mechanisms, mitogen-activated protein kinases, and key transcription factors like NF-κB and p53. The review also focuses on the role of G protein-coupled neurotransmitter receptors (GPCRs) in immune and pro-inflammatory responses, with a detailed analysis of how 13 of 14 types of human 5-HT receptors contribute to depression and neuroinflammation. RESULTS The review reveals a complex interaction between neurotransmitter signals and immunoinflammatory responses in stress-related pathologies. It highlights the role of GPCRs and canonical inflammatory mediators in influencing both pathological and physiological processes in nervous tissue. CONCLUSION The proposed Neuroimmunoinflammatory Stress Model (NIIS Model) suggests that proinflammatory signaling pathways, mediated by metabotropic and ionotropic neurotransmitter receptors, are crucial for maintaining neuronal homeostasis. Chronic mental stress can disrupt this balance, leading to increased pro-inflammatory states in the brain and contributing to neuropsychiatric and psychosomatic disorders, including depression. This model integrates traditional theories on depression pathogenesis, offering a comprehensive understanding of the multifaceted nature of the condition.
Collapse
Affiliation(s)
- Evgenii Gusev
- Laboratory of Inflammation Immunology, Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, Ekaterinburg 620049, Russia
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, Chelyabinsk 454080, Russia
| | - Alexey Sarapultsev
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, Chelyabinsk 454080, Russia
- Laboratory of Immunopathophysiology, Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, Ekaterinburg 620049, Russia
| |
Collapse
|
48
|
Niu Z, Yu M, Xu P, Liu R, Li S, Wu C, Huang B, Ye X, Hu J, Xu Y, Lu S. Effect of 40 Hz light flicker on cognitive impairment and transcriptome of hippocampus in right unilateral common carotid artery occlusion mice. Sci Rep 2023; 13:21361. [PMID: 38049571 PMCID: PMC10695931 DOI: 10.1038/s41598-023-48897-6] [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: 10/14/2023] [Revised: 11/18/2023] [Accepted: 11/30/2023] [Indexed: 12/06/2023] Open
Abstract
Vascular cognitive impairment caused by chronic cerebral hypoperfusion (CCH) seriously affects the quality of life of elderly patients. However, there is no effective treatment to control this disease. This study investigated the potential neuroprotective effect of the 40 Hz light flicker in a mouse model of CCH. CCH was induced in male C57 mice by right unilateral common carotid artery occlusion (rUCCAO), leading to chronic brain injury. The mice underwent 40 Hz light flicker stimulation for 30 days after surgery. The results showed that 40 Hz light flicker treatment ameliorated memory deficits after rUCCAO and alleviated the damage to neurons in the frontal lobe and hippocampus. Light flicker administration at 40 Hz decreased IL-1β and TNF-α levels in the frontal lobe and hippocampus, but immunohistochemistry showed that it did not induce angiogenesis in mice with rUCCAO. Gene expression profiling revealed that the induction of genes was mainly enriched in inflammatory-related pathways. Our findings demonstrate that 40 Hz light flicker can suppress cognitive impairment caused by rUCCAO and that this effect may be involved in the attenuation of neuroinflammation.
Collapse
Affiliation(s)
- Zhaorui Niu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Minjie Yu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Peixia Xu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Renchuan Liu
- Zhejiang Xinyue Health Consulting Service Medical Institution, Hangzhou, 310003, China
| | - Shangda Li
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Congchong Wu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Bochao Huang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
- Zhejiang Xinyue Health Consulting Service Medical Institution, Hangzhou, 310003, China
| | - Xinyi Ye
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Jianbo Hu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China
- Brain Research Institute of Zhejiang University, Hangzhou, 310003, China
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, 310003, China
| | - Yi Xu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China.
- Brain Research Institute of Zhejiang University, Hangzhou, 310003, China.
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, 310003, China.
- Zhejiang Xinyue Health Consulting Service Medical Institution, Hangzhou, 310003, China.
| | - Shaojia Lu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China.
- Brain Research Institute of Zhejiang University, Hangzhou, 310003, China.
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, 310003, China.
| |
Collapse
|
49
|
Wong LJ, Lee BWL, Sng YJ, Poh L, Rajeev V, Selvaraji S, Drummond GR, Sobey CG, Arumugam TV, Fann DY. Inflammasome Activation Mediates Apoptotic and Pyroptotic Death in Astrocytes Under Ischemic Conditions. Neuromolecular Med 2023; 25:533-544. [PMID: 37646911 DOI: 10.1007/s12017-023-08753-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/20/2023] [Indexed: 09/01/2023]
Abstract
Inflammation is a hallmark mechanism of ischemic stroke-induced brain injury. Recent studies have shown that an intracellular multimeric protein complex known as an inflammasome is a key factor for inducing an inflammatory response, and apoptotic and pyroptotic cell death in ischemic stroke. Inflammasome assembly leads to the activation of pro-inflammatory caspases, and the maturation and secretion of pro-inflammatory cytokines IL-1β and IL-18. While the role of inflammasomes in ischemic stroke-induced neuronal death, and microglial activation and cell death have been established, little is known about the role of inflammasomes in astrocytes under ischemic conditions. In this study, we investigated the expression and activation of inflammasome components in protoplasmic and fibrous astrocytes under ischemic conditions. We found that both protoplasmic and fibrous astrocytes expressed a differential increase in inflammasome protein components, and that their activation promoted maturation of IL-1β and IL-18, and secretion of IL-1β, as well as initiating apoptotic and pyroptotic cell death. Pharmacological inhibition of caspase-1 decreased expression of cleaved caspase-1 and production of mature IL-1β, and protected against inflammasome-mediated apoptotic and pyroptotic cell death. Overall, this study provides novel insights into the role of inflammasome signaling in astrocytes under ischemic conditions.
Collapse
Affiliation(s)
- Lap Jack Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Bernice Woon Li Lee
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yi Jing Sng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Luting Poh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vismitha Rajeev
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sharmelee Selvaraji
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
| | - Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research and Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research and Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Thiruma V Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Centre for Cardiovascular Biology and Disease Research and Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia.
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.
| | - David Y Fann
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Healthy Longevity Translational Research Programme (HLTRP), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
50
|
Tan S, Chen W, Kong G, Wei L, Xie Y. Peripheral inflammation and neurocognitive impairment: correlations, underlying mechanisms, and therapeutic implications. Front Aging Neurosci 2023; 15:1305790. [PMID: 38094503 PMCID: PMC10716308 DOI: 10.3389/fnagi.2023.1305790] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/10/2023] [Indexed: 08/22/2024] Open
Abstract
Cognitive impairments, such as learning and memory deficits, may occur in susceptible populations including the elderly and patients who are chronically ill or have experienced stressful events, including surgery, infection, and trauma. Accumulating lines of evidence suggested that peripheral inflammation featured by the recruitment of peripheral immune cells and the release of pro-inflammatory cytokines may be activated during aging and these conditions, participating in peripheral immune system-brain communication. Lots of progress has been achieved in deciphering the core bridging mechanism connecting peripheral inflammation and cognitive impairments, which may be helpful in developing early diagnosis, prognosis evaluation, and prevention methods based on peripheral blood circulation system sampling and intervention. In this review, we summarized the evolving evidence on the prevalence of peripheral inflammation-associated neurocognitive impairments and discussed the research advances in the underlying mechanisms. We also highlighted the prevention and treatment strategies against peripheral inflammation-associated cognitive dysfunction.
Collapse
Affiliation(s)
- Siyou Tan
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Anesthesiology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Wenyan Chen
- Department of Anesthesiology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Gaoyin Kong
- Department of Anesthesiology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Lai Wei
- Department of Anesthesiology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yubo Xie
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|