1
|
Li E, Niu W, Lu C, Wang M, Xu X, Xu K, Xu P. Interoception and aging. Ageing Res Rev 2025; 108:102743. [PMID: 40188990 DOI: 10.1016/j.arr.2025.102743] [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: 12/04/2024] [Revised: 03/04/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Interoception refers to the body's perception and regulation of internal physiological states and involves complex neural mechanisms and sensory systems. The current definition of interoception falls short of capturing the breadth of related research; here, we propose an updated definition. Homeostasis, a foundational principle of integrated physiology, is the process by which organisms dynamically maintain optimal balance across all conditions through neural, endocrine, and behavioral functions. This review examines the role of interoception in body homeostasis. Aging is a complex process influenced by multiple factors and involving multiple levels, including physical, psychological, and cognitive. However, interoceptive and aging interoceptive interactions are lacking. A new perspective on interoception and aging holds significant implications for understanding how aging regulates interoception and how interoception affects the aging process. Finally, we summarize that arachidonic acid metabolites show promise as biomarkers of interoception-aging. The aim of this study is to comprehensively analyze interoceptive-aging interactions, understand the aging mechanism from a novel perspective, and provide a theoretical basis for exploring anti-aging strategies.
Collapse
Affiliation(s)
- Erliang Li
- Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, China; Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an 710000, China.
| | - Wenjing Niu
- Changlefang Community Health Service Center, Xi'an 710000, China
| | - Chao Lu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, China
| | - Min Wang
- Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, China; Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an 710000, China
| | - Xin Xu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, China
| | - Ke Xu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, China.
| | - Peng Xu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, China; Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an 710000, China.
| |
Collapse
|
2
|
Song D, Li Y, Yang LL, Luo YX, Yao XQ. Bridging systemic metabolic dysfunction and Alzheimer's disease: the liver interface. Mol Neurodegener 2025; 20:61. [PMID: 40437610 PMCID: PMC12121119 DOI: 10.1186/s13024-025-00849-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 05/09/2025] [Indexed: 06/01/2025] Open
Abstract
Alzheimer's disease (AD) is increasingly recognized as a systemic disorder with a substantial metabolic disorder component, where the liver significantly impacts the brain via the liver-brain axis. Key mechanisms include the liver's role in clearing peripheral β-amyloid (Aβ), the influence of hepatic enzymes and metabolites on cognitive decline, and the systemic effects of metabolic disorders on AD progression. Hepatokines, liver-secreted proteins including fibroblast growth factor (FGF)-21, selenoprotein P (SELENOP), Fetuin-A, Midbrain astrocyte-derived neurotrophic factor (MANF), apolipoprotein J (ApoJ), sex hormone-binding globulin (SHBG), Adropin and Angiopoietin-like protein 3 (ANGPTL3), could regulate insulin sensitivity, lipid metabolism, oxidative stress, immune responses, and neurotrophic support. These pathways are closely linked to core AD pathologies, including Aβ aggregation, tau hyperphosphorylation, neuroinflammation, oxidative stress and mitochondrial dysfunction. Lifestyle interventions, including exercise and dietary modifications, that regulate hepatokines expression may offer novel preventive and therapeutic strategies for AD. This review synthesizes current knowledge on the liver-brain crosstalk in AD, emphasizing the mechanistic role of liver in bridging metabolic dysfunction with neurodegeneration and underscores the diagnostic and therapeutic potential of hepatokines in addressing AD's complex pathology.
Collapse
Affiliation(s)
- Dan Song
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Yang Li
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Ling-Ling Yang
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Ya-Xi Luo
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Xiu-Qing Yao
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
- Chongqing Municipality Clinical Research Center for Geriatric Medicine, No. 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
- Department of Rehabilitation Therapy, Chongqing Medical University, No. 1 Medical College Road, Yuzhong District, Chongqing, 400000, China.
| |
Collapse
|
3
|
Pan L, Xie L, Yang W, Feng S, Mao W, Ye L, Cheng H, Wu X, Mao X. The role of brain-liver-gut Axis in neurological disorders. BURNS & TRAUMA 2025; 13:tkaf011. [PMID: 40321299 PMCID: PMC12048006 DOI: 10.1093/burnst/tkaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 05/08/2025]
Abstract
In recent years, with the increasing volume of related research, it has become apparent that the liver and gut play important roles in the pathogenesis of neurological disorders. Considering the interactions among the brain, liver, and gut, the brain-liver-gut axis has been proposed and gradually recognized. In this article, we summarized the complex network of interactions within the brain-liver-gut axis, encompassing the vagus nerve, barrier permeability, immunity and inflammation, the blood-brain barrier, gut microbial metabolites, the gut barrier, neurotoxic metabolites, and beta-amyloid (Aβ) metabolism. We also elaborated on the impact of the brain-liver-gut axis on various neurological disorders. Furthermore, we outline several therapies aimed at modulating the brain-liver-gut axis, including antibiotics, probiotics and prebiotics, fecal microbiota transplantation (FMT), vagus nerve stimulation (VNS), and dietary interventions. The focus is on elucidating possible mechanisms underlying neurological disorders pathogenesis and identifying effective treatments that are based on our understanding of the brain-liver-gut axis.
Collapse
Affiliation(s)
- Li Pan
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Lizheng Xie
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230022, China
| | - Wenpei Yang
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230022, China
| | - Shi Feng
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Wenbao Mao
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Lei Ye
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Hongwei Cheng
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Xiao Wu
- Department of Emergency, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Xiang Mao
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230022, China
| |
Collapse
|
4
|
Wang S, Qi C, Rajpurohit C, Ghosh B, Xiong W, Wang B, Qi Y, Hwang SH, Hammock BD, Li H, Gan L, Zheng H. Inhibition of soluble epoxide hydrolase confers neuroprotection and restores microglial homeostasis in a tauopathy mouse model. Mol Neurodegener 2025; 20:44. [PMID: 40264187 PMCID: PMC12016400 DOI: 10.1186/s13024-025-00844-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND The epoxyeicosatrienoic acids (EETs) are derivatives of the arachidonic acid metabolism with anti-inflammatory activities. However, their efficacy is limited due to the rapid hydrolysis by soluble epoxide hydrolase (sEH). Inhibition of sEH has been shown to stabilize the EETs and reduce neuroinflammation in Aβ mouse models of Alzheimer's disease (AD). However, the role of the sEH-EET signaling pathway in other CNS cell types and neurodegenerative conditions are less understood. METHODS Here we investigated the mechanisms and functional role of the sEH-EET axis in tauopathy by treating PS19 mice with a small molecule sEH inhibitor TPPU and by crossing the PS19 mice with Ephx2 (gene encoding sEH) knockout mice. This was followed by single-nucleus RNA-sequencing (snRNA-seq), biochemical and immunohistochemical analysis, and behavioral assessments. Additionally, we examined the effects of the sEH-EET pathway in primary microglia cultures and human induced pluripotent stem cell (iPSC)-derived neurons exhibiting seeding-induced Tau inclusions. RESULTS sEH inhibition improved cognitive function, rescued neuronal cell loss, and reduced Tau pathology and microglial reactivity. snRNA-seq revealed that TPPU treatment upregulated genes involved in actin cytoskeleton and excitatory synaptic pathways. Treatment of human iPSC-derived neurons with TPPU enhanced synaptic density without affecting Tau accumulation, suggesting a cell-autonomous neuroprotective effect of sEH blockade. Furthermore, sEH inhibition reversed disease-associated and interferon-responsive microglial states in PS19 mice, while EET supplementation promoted Tau phagocytosis and clearance in primary microglia cultures. CONCLUSION These findings demonstrate that sEH blockade or EET augmentation confers therapeutic benefit in neurodegenerative tauopathies by simultaneously targeting neuronal and microglial pathways.
Collapse
Affiliation(s)
- Shuo Wang
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Chuangye Qi
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Chetan Rajpurohit
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Baijayanti Ghosh
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Wen Xiong
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Baiping Wang
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Yanyan Qi
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Sung Hee Hwang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Bruce D Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Hongjie Li
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
5
|
Wang Q, Hu FR, Gou XC, Wang S, Ji NC. Aerobic Exercise Ameliorates Alzheimer's Disease-Like Pathology by Regulating Hepatic Phagocytosis of Aβ. FRONT BIOSCI-LANDMRK 2025; 30:36597. [PMID: 40302344 DOI: 10.31083/fbl36597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/19/2025] [Accepted: 03/21/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disease which significantly and negatively affects families and society. Aerobic exercise serves as a non-pharmacological strategy, potentially safeguarding against cognitive decline and lowering the risk of AD. However, how aerobic exercise ameliorates AD remains unknown. This study investigated the effects of two types of aerobic exercise, including aerobic interval training (AIT) and aerobic continuous training (ACT), on cognitive and exploratory function, brain histopathology, and hepatic amyloid beta (Aβ) clearance in amyloid precursor protein/presenilin-1 double transgenic (APP/PS1) transgenic mice. METHODS Twenty-four six-month-old male APP/PS1 transgenic mice (body weight: 20-22 g) were used to establish the AD model. APP/PS1 transgenic mice were randomly assigned to one of the three groups: rest (AD group, n = 8), aerobic interval training (AIT group, n = 8), and aerobic continuous training (ACT group, n = 8). The exploration ability and anxiety of AD mice were measured using the open-field test. Learning and memory of AD mice were detected using the novel object recognition test, Y-maze test, and Morris water maze test. Neuronal damage was analyzed using hematoxylin and eosin staining and Nissl staining. Aβ deposition in the brain was detected using a thioflavin-S fluorescence assay and immunofluorescence. The mechanisms underlying hepatic Aβ clearance were investigated using an immunofluorescence assay and western blotting. Data were analyzed using one-way ANOVA with Tukey's post hoc test, and p < 0.05 was deemed statistically significant. RESULTS The results revealed that both AIT and ACT improved the recognition memory and exploration ability of mice after 8 weeks of intervention. Additionally, both forms of aerobic exercise significantly mitigated neuronal damage and Aβ deposition in the brain and improved the hepatic clearance of Aβ. CONCLUSIONS Our findings indicated that AIT and ACT can improve cognitive deficits in APP/PS1 mice, potentially by increasing the hepatic phagocytic capacity of Aβ. Hepatic clearance of Aβ may serve as a supplementary mechanism by which aerobic exercise can improve AD.
Collapse
Affiliation(s)
- Qing Wang
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, 710021 Xi'an, Shaanxi, China
- Engineering Research Center of Brain Diseases Drug Development, Universities of Shaanxi Province, Xi'an Medical University, 710021 Xi'an, Shaanxi, China
| | - Feng-Rui Hu
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, 710021 Xi'an, Shaanxi, China
- Engineering Research Center of Brain Diseases Drug Development, Universities of Shaanxi Province, Xi'an Medical University, 710021 Xi'an, Shaanxi, China
| | - Xing-Chun Gou
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, 710021 Xi'an, Shaanxi, China
- Engineering Research Center of Brain Diseases Drug Development, Universities of Shaanxi Province, Xi'an Medical University, 710021 Xi'an, Shaanxi, China
| | - Shan Wang
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, 710021 Xi'an, Shaanxi, China
- Engineering Research Center of Brain Diseases Drug Development, Universities of Shaanxi Province, Xi'an Medical University, 710021 Xi'an, Shaanxi, China
| | - Nai-Chun Ji
- Center for Blockchain & Healthcare Service, Xi'an Medical University, 710021 Xi'an, Shaanxi, China
| |
Collapse
|
6
|
Yue C, Chen B, Pan F, Wang Z, Yu H, Liu G, Li W, Wang R, Tang Y. TCnet: A Novel Strategy to Predict Target Combination of Alzheimer's Disease via Network-Based Methods. J Chem Inf Model 2025; 65:3866-3878. [PMID: 40172120 DOI: 10.1021/acs.jcim.5c00172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder with an unclear pathogenesis; the traditional ″single gene-single target-single drug″ strategy is insufficient for effective treatment. This study explores a novel strategy for the multitarget therapy of AD by integrating multiomics data and employing network analysis. Different from conventional single-target methods, TCnet adopts a mechanism-driven strategy, utilizing multiomics data to decompose disease mechanisms, construct potential target combinations, and prioritize the optimal combinations using a scoring function. TCnet not only advances our understanding of disease mechanisms but also facilitates large-scale drug screening. This approach was further employed to screen active compounds from Huang-Lian-Jie-Du-Tang (HLJDT), identifying quercetin as a candidate targeting GSK3β and ADAM17. Subsequent in vitro experiments confirmed the neuroprotective and anti-inflammatory effects of quercetin. Overall, TCnet offers a promising approach for predicting target combinations and provides new insights and directions for drug discovery in AD.
Collapse
Affiliation(s)
- Chengyuan Yue
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Baiyu Chen
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Fei Pan
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Ze Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Hongbo Yu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Rui Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
7
|
Zheng S, Guo J, Zheng R, Ji Y, Zhong Q, Yin H. A Naturalistic Prospective Study of the Prognostic Impact of EPHX2 in Major Depressive Disorder: Impulsivity may be an Important Factor. Depress Anxiety 2025; 2025:8124403. [PMID: 40259894 PMCID: PMC12009677 DOI: 10.1155/da/8124403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 03/20/2025] [Indexed: 04/23/2025] Open
Abstract
Background: Major depressive disorder (MDD) is a leading cause of disability worldwide. The pathophysiology of MDD remains unclear, which limits the development of treatments for MDD. Recently, epoxide hydrolase 2 (EPHX2) has been found to be associated with MDD. Our previous study revealed an association between EPHX2 expression and suicide. However, the effect of EPHX2 on the prognosis of MDD and suicide remains unclear. Previous studies have found that impulsivity at baseline can be a significant predictor of clinical improvement in patients with MDD. Therefore, we inferred that EPHX2 could be associated with the treatment effect of MDD, and impulsivity could mediate the effect of EPHX2 on the treatment effect of MDD. Methods: This naturalistic prospective study included 117 participants with MDD, who were assessed, using clinical questionnaires, cognitive function, and treatment information, at baseline, 2 weeks, and 1, 2, and 3 months. A linear mixed-effects model was used to investigate longitudinal changes in the severity of symptoms, risk of suicide, and cognitive function. Results: The interactive effects of impulsivity and EPHX2 polymorphisms on the risk of suicide (measured by the Columbia-Suicide Severity Rating Scale) were significantly different for rs11288636, rs68012435, and rs11288636. The interactive effects between polymorphisms and time on depression severity (measured by the Hamilton Depression Scale-24) were significantly different and including after adjustment for the total impulsivity score. Conclusions: This study suggests that EPHX2 polymorphisms are associated with the prognosis of MDD, and impulsivity could be a critical factor for the change in suicide risk among different EPHX2 genotypes. Trial Registration: ClinicalTrials.gov identifier: NCT05575713.
Collapse
Affiliation(s)
- Shuqiong Zheng
- Department of Psychiatry, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Jia Guo
- Department of Biostatistics, Columbia University, New York, New York, USA
| | - Rongxin Zheng
- Department of Psychiatry, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Yujia Ji
- Department of Psychiatry, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Quan Zhong
- Department of Psychiatry, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Honglei Yin
- Department of Psychiatry, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Cui Y, Zhi SM, Ding PF, Zhu T, Chen XX, Liu XZ, Sheng B, Li XJ, Wang J, Zhang JT, Xu MX, Jiang YX, Hang CH, Li W. Silybin attenuates microglia-mediated neuroinflammation via inhibition of STING in experimental subarachnoid hemorrhage. Int Immunopharmacol 2025; 151:114305. [PMID: 39986195 DOI: 10.1016/j.intimp.2025.114305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND The primary cause of subarachnoid hemorrhage (SAH) is the rupture of intracranial aneurysms. Over-activation of microglia following SAH is a primary driving force in early brain injury (EBI), which is a leading cause of poor outcomes. Silybin is a flavonoid compound extracted from Silybum marianum, a plant belonging to the Asteraceae family. Its anti-inflammatory and antioxidant properties could provide neuroprotective effects. The mechanism of silybin on EBI after SAH is unclear. PURPOSE To determine the therapeutic effect of silybin on SAH and its underlying mechanisms. METHODS We used a prechiasmatic autologous arterial blood injection in vivo and hemoglobin in vitro to establish experimental SAH model. Dexamethasone was used as a positive control drug. We evaluated the neuroprotective effect of silybin on the in vivo SAH model by neurological function scores, rotarod test, and open field test, and explored the protective effect of silybin on neuroinflammation and apoptosis after SAH by quantitative polymerase chain reaction (qPCR), western blot (WB), Immunofluorescence (IF) and TUNEL staining. IF staining of CD86 and CD206 was used to assess microglial phenotype polarization. Then we used WB and IF labeling of STING to explore the effect of silybin on the STING pathway after SAH, and used a combination of transcriptomics and non-targeted metabolomics to study the potential mechanism of silybin in detail, and verified the essential genes by qPCR. We also extracted cerebrospinal fluid from SAH patients and detected the expression level of STING in cerebrospinal fluid by enzyme-linked immunosorbent assay (ELISA) to clarify the association between STING and neural function. RESULTS Results showed that silybin ameliorated neuronal damage and improved short-term neurological function, and reduced inflammatory damage and neuronal apoptosis in SAH mice. Silybin inhibited the expression levels of TNF-α, IL-1β and NLRP3, and promoted the expression levels of CD206, Arg1 and IL-10. Notably, Silybin promoted M2 microglia polarization. Further studies found that silybin reduced the mRNA and protein levels of the stimulator of interferon genes (STING) in microglia. And the use of a specific activator of STING (CMA) disrupted the protective effect of silybin. A total of 358 differential expression genes were identified using transcriptomics, and 150 different metabolites abundance were identified using metabolomic screening. Analysis of the effects of STING on transcriptomics and metabolomics revealed that STING might impact metabolic pathways, including linoleic acid metabolism. The qPCR results confirmed the decreased expression of essential proteins involved in the pathway. Finally, we found that increased STING expression in the cerebrospinal fluid of SAH patients was associated with decreased neurological function scores and poor prognosis. CONCLUSION Silybin had a therapeutic effect on SAH. The underlying mechanism involves linoleic acid metabolism, which is associated with the differential genes and metabolites detected in the study. This study presented a pharmacological rationale for using silybin to treat SAH.
Collapse
Affiliation(s)
- Yue Cui
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University
| | - Si-Min Zhi
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University
| | - Peng-Fei Ding
- Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Ting Zhu
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University
| | - Xiang-Xin Chen
- Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xun-Zhi Liu
- Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Bin Sheng
- Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiao-Jian Li
- Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jie Wang
- Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jia-Tong Zhang
- Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Meng-Xiao Xu
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University
| | - Yong-Xiao Jiang
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University
| | - Chun-Hua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
9
|
Asimakidou E, Saipuljumri EN, Lo CH, Zeng J. Role of metabolic dysfunction and inflammation along the liver-brain axis in animal models with obesity-induced neurodegeneration. Neural Regen Res 2025; 20:1069-1076. [PMID: 38989938 PMCID: PMC11438328 DOI: 10.4103/nrr.nrr-d-23-01770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 04/26/2024] [Indexed: 07/12/2024] Open
Abstract
The interaction between metabolic dysfunction and inflammation is central to the development of neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Obesity-related conditions like type 2 diabetes and non-alcoholic fatty liver disease exacerbate this relationship. Peripheral lipid accumulation, particularly in the liver, initiates a cascade of inflammatory processes that extend to the brain, influencing critical metabolic regulatory regions. Ceramide and palmitate, key lipid components, along with lipid transporters lipocalin-2 and apolipoprotein E, contribute to neuroinflammation by disrupting blood-brain barrier integrity and promoting gliosis. Peripheral insulin resistance further exacerbates brain insulin resistance and neuroinflammation. Preclinical interventions targeting peripheral lipid metabolism and insulin signaling pathways have shown promise in reducing neuroinflammation in animal models. However, translating these findings to clinical practice requires further investigation into human subjects. In conclusion, metabolic dysfunction, peripheral inflammation, and insulin resistance are integral to neuroinflammation and neurodegeneration. Understanding these complex mechanisms holds potential for identifying novel therapeutic targets and improving outcomes for neurodegenerative diseases.
Collapse
Affiliation(s)
- Evridiki Asimakidou
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Eka Norfaishanty Saipuljumri
- School of Applied Science, Republic Polytechnic, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Chih Hung Lo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jialiu Zeng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
10
|
Jiang Q, Liu J, Huang S, Wang XY, Chen X, Liu GH, Ye K, Song W, Masters CL, Wang J, Wang YJ. Antiageing strategy for neurodegenerative diseases: from mechanisms to clinical advances. Signal Transduct Target Ther 2025; 10:76. [PMID: 40059211 PMCID: PMC11891338 DOI: 10.1038/s41392-025-02145-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/29/2024] [Accepted: 01/15/2025] [Indexed: 05/13/2025] Open
Abstract
In the context of global ageing, the prevalence of neurodegenerative diseases and dementia, such as Alzheimer's disease (AD), is increasing. However, the current symptomatic and disease-modifying therapies have achieved limited benefits for neurodegenerative diseases in clinical settings. Halting the progress of neurodegeneration and cognitive decline or even improving impaired cognition and function are the clinically meaningful goals of treatments for neurodegenerative diseases. Ageing is the primary risk factor for neurodegenerative diseases and their associated comorbidities, such as vascular pathologies, in elderly individuals. Thus, we aim to elucidate the role of ageing in neurodegenerative diseases from the perspective of a complex system, in which the brain is the core and peripheral organs and tissues form a holistic network to support brain functions. During ageing, the progressive deterioration of the structure and function of the entire body hampers its active and adaptive responses to various stimuli, thereby rendering individuals more vulnerable to neurodegenerative diseases. Consequently, we propose that the prevention and treatment of neurodegenerative diseases should be grounded in holistic antiageing and rejuvenation means complemented by interventions targeting disease-specific pathogenic events. This integrated approach is a promising strategy to effectively prevent, pause or slow down the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Qiu Jiang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Jie Liu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Shan Huang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Xuan-Yue Wang
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
| | - Xiaowei Chen
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
- Brain Research Center, Third Military Medical University, Chongqing, China
| | - Guang-Hui Liu
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Keqiang Ye
- Faculty of Life and Health Sciences, and Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weihong Song
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province. Zhejiang Clinical Research Center for Mental Disorders, School of Mental Health and The Affiliated Kangning Hospital, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia.
| | - Jun Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, China.
| |
Collapse
|
11
|
Zhang X, Peng L, Kuang S, Wang T, Wu W, Zuo S, Chen C, Ye J, Zheng G, Guo Y, He Y. Lactate accumulation from HIF-1α-mediated PMN-MDSC glycolysis restricts brain injury after acute hypoxia in neonates. J Neuroinflammation 2025; 22:59. [PMID: 40025545 PMCID: PMC11871681 DOI: 10.1186/s12974-025-03385-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 02/17/2025] [Indexed: 03/04/2025] Open
Abstract
Fetal intrauterine distress (FD) during delivery can cause fetal intrauterine hypoxia, posing significant risks to the fetus, mother, and newborns. While studies highlight the role of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) in neonatal diseases and tumor hypoxia, their specific involvement in newborns experiencing fetal distress during delivery (FDNB) is not well understood. Here, we found elevated PMN-MDSC activation, increased glycolysis, enhanced lactate production, and upregulated HIF-1α expression in the blood of FDNB neonates compared to healthy newborns (NNB). Importantly, PMN-MDSC levels were inversely correlated with neuron-specific enolase (NSE), a marker for neurological injury. In neonatal mice subjected to acute hypoxia, a 48-h exposure led to a shift from exacerbation to amelioration of brain damage when compared with a 24-h period. This change was associated with a reduction in microglial activation, a decrease in the expression of inflammatory factors within the microglia, alongside increased peripheral PMN-MDSC activation. Depleting PMN-MDSCs led to heightened microglial activation and aggravated brain injury. Mechanistically, enhanced activation of PMN-MDSCs promotes HIF-1α accumulation while enhancing glycolysis and lactate release, thereby mitigating neonatal brain injury. Notably, lactate supplementation in hypoxic mice rescued brain damage caused by insufficient PMN-MDSC activation due to HIF-1α deficiency. Our study clarifies the role of lactate in peripheral PMN-MDSCs after acute hypoxia and its effects on microglial activation and subsequent brain injury.
Collapse
Affiliation(s)
- Xiaogang Zhang
- Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Immunology, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Laiqin Peng
- Department of Gynecology and Obstetrics, Huizhou Central People's Hospital, Huizhou, China
| | - Shuyi Kuang
- Department of Immunology, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Tianci Wang
- Department of Immunology, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Weibin Wu
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaowen Zuo
- Department of Immunology, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Chunling Chen
- Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University; Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences,, Guangzhou, China
| | - Jiaxiu Ye
- Department of Gynecology and Obstetrics, Huizhou Central People's Hospital, Huizhou, China
| | - Guilang Zheng
- Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University; Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences,, Guangzhou, China.
| | - Yuxiong Guo
- Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University; Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences,, Guangzhou, China.
| | - Yumei He
- Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
- Department of Immunology, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Wu B, Liu Y, Li H, Zhu L, Zeng L, Zhang Z, Peng W. Liver as a new target organ in Alzheimer's disease: insight from cholesterol metabolism and its role in amyloid-beta clearance. Neural Regen Res 2025; 20:695-714. [PMID: 38886936 PMCID: PMC11433892 DOI: 10.4103/1673-5374.391305] [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: 05/31/2023] [Revised: 09/14/2023] [Accepted: 11/07/2023] [Indexed: 06/20/2024] Open
Abstract
Alzheimer's disease, the primary cause of dementia, is characterized by neuropathologies, such as amyloid plaques, synaptic and neuronal degeneration, and neurofibrillary tangles. Although amyloid plaques are the primary characteristic of Alzheimer's disease in the central nervous system and peripheral organs, targeting amyloid-beta clearance in the central nervous system has shown limited clinical efficacy in Alzheimer's disease treatment. Metabolic abnormalities are commonly observed in patients with Alzheimer's disease. The liver is the primary peripheral organ involved in amyloid-beta metabolism, playing a crucial role in the pathophysiology of Alzheimer's disease. Notably, impaired cholesterol metabolism in the liver may exacerbate the development of Alzheimer's disease. In this review, we explore the underlying causes of Alzheimer's disease and elucidate the role of the liver in amyloid-beta clearance and cholesterol metabolism. Furthermore, we propose that restoring normal cholesterol metabolism in the liver could represent a promising therapeutic strategy for addressing Alzheimer's disease.
Collapse
Affiliation(s)
- Beibei Wu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yuqing Liu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Hongli Li
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Lemei Zhu
- Academician Workstation, Changsha Medical University, Changsha, Hunan Province, China
| | - Lingfeng Zeng
- Academician Workstation, Changsha Medical University, Changsha, Hunan Province, China
| | - Zhen Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Yangsheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
- Qinhuangdao Shanhaiguan Pharmaceutical Co., Ltd, Qinhuangdao, Hebei Province, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Mental Disorder, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
13
|
Qiao M, Ni J, Qing H, Qiu Y, Quan Z. Role of Peripheral NLRP3 Inflammasome in Cognitive Impairments: Insights of Non-central Factors. Mol Neurobiol 2025:10.1007/s12035-025-04779-8. [PMID: 40000575 DOI: 10.1007/s12035-025-04779-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/13/2025] [Indexed: 02/27/2025]
Abstract
Cognitive impairments are common clinical manifestation of Alzheimer's disease, vascular dementia, type 2 diabetes mellitus, and autoimmune diseases. Emerging evidence has suggested a strong correlation between peripheral chronic inflammation and cognitive impairments. For example, nearly 40% of individuals with inflammatory bowel disease also suffer from cognitive impairments. In this condition, NLRP3 inflammasome (NLRP3-I) generating pro-inflammatory cytokines like IL-1β serves as a significant effector, and its persistence exerts adverse effects to both periphery and the brain. Moreover, investigations on serum biomarkers of mild cognitive impairments have shown NLRP3-I components' upregulation, suggesting the involvement of peripheral inflammasome pathway in this disorder. Here, we systematically reviewed the current knowledge of NLRP3-I in inflammatory disease to uncover its potential role in bridging peripheral chronic inflammation and cognitive impairments. This review summarizes the molecular features and ignition process of NLRP3-I in inflammatory response. Meanwhile, various effects of NLRP3-I involved in peripheral inflammation-associated disease are also reviewed, especially its chronic disturbances to brain homeostasis and cognitive function through routes including gut-brain, liver-brain, and kidney-brain axes. In addition, current promising compounds and their targets relative to NLRP3-I are discussed in the context of cognitive impairments. Through the detailed investigation, this review highlights the critical role of peripheral NLRP3-I in the pathogenesis of cognitive disorders, and offers novel perspectives for developing effective therapeutic interventions for diseases associated with cognitive impairments. The present review outlines the current knowledge on the ignition of NLRP3-I in inflammatory disease and more importantly, emphasizes the role of peripheral NLRP3-I as a causal pathway in the development of cognitive disorders. Although major efforts to restrain cognitive decline are mainly focused on the central nervous system, it has become clear that disturbances from peripheral immune are closely associated with the dysfunctional brain. Therefore, attenuation of these inflammatory changes through inhibiting the NLRP3-I pathway in early inflammatory disease may reduce future risk of cognitive impairments, and in the meantime, considerations on such pathogenesis for combined drug therapy will be required in the clinical evaluation of cognitive disorders.
Collapse
Affiliation(s)
- Mengfan Qiao
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
- Department of Biology, Shenzhen MSU-BIT University, Shenzhen, 518172, China
| | - Yunjie Qiu
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
14
|
Wang Z, Yin Z, Sun G, Zhang D, Zhang J. Genetic evidence for the liver-brain axis: lipid metabolism and neurodegenerative disease risk. Lipids Health Dis 2025; 24:41. [PMID: 39923073 PMCID: PMC11806572 DOI: 10.1186/s12944-025-02455-3] [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: 12/04/2024] [Accepted: 01/29/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND The liver‒brain axis is critical in neurodegenerative diseases (NDs), with lipid metabolism influencing neuroinflammation and microglial function. A systematic investigation of the genetic relationship between lipid metabolism abnormalities and ND, namely, Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS), is lacking. To assess potential causal links between ND and six lipid parameters, two-sample Mendelian randomization (MR) was used. METHODS Large-scale European ancestry GWAS data for lipid parameters and ND (AD, ALS, PD, and MS) were used. Genetic variants demonstrating significant correlations (P < 5 × 10-8) with lipid metabolism parameters were identified and employed as instrumental variables (IVs) after proper validation. The research incorporated UK Biobank genomic data to examine associations between genetic variants and lipid metabolism parameters. The analysis included primary MR, sensitivity analyses, and multivariable MR, which considered potential mediators. RESULTS MR via the inverse-variance weighted method revealed causal effects of cholesterol (CHOL, OR = 1.10, 95% CI: 1.03-1.18, P = 4.23 × 10⁻3) and low-density lipoprotein cholesterol (LDLC, OR = 1.10, 95% CI: 1.03-1.17, P = 3.28 × 10⁻3) on the risk of ALS, which were validated across multiple methods. Potential correlations were observed between ApoB and ALS and inversely correlated with AD, whereas no significant associations were found for PD or MS. CHOL and LDLC associations with ALS demonstrated no significant heterogeneity or pleiotropy, supporting their reliability. CONCLUSIONS Higher CHOL and LDLC levels were associated with increased ALS risk, suggesting a potential causal link, and supporting the liver‒brain axis hypothesis in ND. Current genetic evidence does not support a significant role for lipid metabolism in PD and MS etiology, suggesting the relationship between lipid metabolism and other NDs may be more complex and warrants further investigation.
Collapse
Affiliation(s)
- Zeyu Wang
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zixiao Yin
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Neurostimulation, Beijing, 100070, China
| | - Guangyong Sun
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Dong Zhang
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Jianguo Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Neurostimulation, Beijing, 100070, China.
| |
Collapse
|
15
|
He J, Xu P, Xu T, Yu H, Wang L, Chen R, Zhang K, Yao Y, Xie Y, Yang Q, Wu W, Sun D, Wu D. Therapeutic potential of hydrogen-rich water in zebrafish model of Alzheimer's disease: targeting oxidative stress, inflammation, and the gut-brain axis. Front Aging Neurosci 2025; 16:1515092. [PMID: 39839307 PMCID: PMC11746902 DOI: 10.3389/fnagi.2024.1515092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder, with amyloid-beta (Aβ) aggregation playing a key role in its pathogenesis. Aβ-induced oxidative stress leads to neuronal damage, mitochondrial dysfunction, and apoptosis, making antioxidative strategies promising for AD treatment. This study investigates the effects of hydrogen-rich water (HRW) in a zebrafish AD model. Zebrafish were exposed to aluminum chloride to induce AD-like pathology and then treated with HRW using a nanobubble device. Behavioral assays, ELISA, Hematoxylin-eosin (H&E) staining, and reactive oxygen species (ROS) and neutrophil fluorescence labeling were employed to assess HRW's impact. Additionally, 16S rRNA sequencing analyzed HRW's effect on gut microbiota. HRW can significantly improve cognitive impairment and depression-like behavior in zebrafish AD model, reduce Aβ deposition (p < 0.0001), regulate liver Soluble epoxide hydrolase (sEH) levels (p < 0.05), reduce neuroinflammation, and reduce oxidative stress. Furthermore, HRW reduced the number of harmful bacteria linked to AD pathology by restoring the balance of microbiota in the gut. These findings suggest that HRW has potential as a therapeutic strategy for AD by targeting oxidative stress, inflammation, and gut-brain axis modulation.
Collapse
Affiliation(s)
- Jiaxuan He
- Institute of Life Sciences and Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Peiye Xu
- Institute of Life Sciences and Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Ting Xu
- Institute of Life Sciences and Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Haiyang Yu
- Institute of Life Sciences and Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Lei Wang
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Kun Zhang
- Chongqing Municipality Clinical Research Center for Geriatric Diseases, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Yueliang Yao
- Fuzhou Innovation Center for AI Drug, Fuzhou Medical College of Nanchang University, Fuzhou, China
| | - Yanyan Xie
- The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Chongqing University, Chongqing, China
| | - Da Sun
- Institute of Life Sciences and Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Dejun Wu
- Department of Geriatric Medicine, Quzhou People’s Hospital, Quzhou, China
| |
Collapse
|
16
|
Kim JW, Byun MS, Yi D, Jung JH, Kong N, Chang YY, Jung G, Ahn H, Lee JY, Kang KM, Sohn CH, Lee YS, Kim YK, Lee DY. Liver function and Alzheimer's brain pathologies: A longitudinal study: Liver and Alzheimer's pathologies. J Prev Alzheimers Dis 2025; 12:100012. [PMID: 39800466 PMCID: PMC12065704 DOI: 10.1016/j.tjpad.2024.100012] [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] [Indexed: 02/27/2025]
Abstract
IMPORTANCE The neuropathological links underlying the association between changes in liver function and AD have not yet been clearly elucidated. OBJECTIVE We aimed to examine the relationship between liver function markers and longitudinal changes in Alzheimer's disease (AD) core pathologies. DESIGN Data from the Korean Brain Aging Study for the Early Diagnosis and Prediction of Alzheimer's Disease, a longitudinal cohort study initiated in 2014, were utilized. SETTING Community and memory clinic setting. PARTICIPANTS Three hundred forty-seven older adults. MAIN OUTCOME AND MEASURES Participants underwent baseline and 2-year follow-up evaluations, including liver function assessments and various brain imaging techniques, such as amyloid and tau PET, FDG-PET, and MRI). Liver function indicators [alanine aminotransferase (ALT), aspartate aminotransferase (AST), and total bilirubin] were examined as exposure variables. RESULTS Higher baseline ALT levels were associated with a greater increase in beta-amyloid deposition over 2 years [β = 0.166, Bonferroni-corrected P (PB) = 0.012], while lower total bilirubin levels were associated with a greater increase in tau deposition over the same period (β = -0.570, PB < 0.001). In contrast, AST alone showed no significant association with changes of AD pathologies. CONCLUSIONS AND RELEVANCE The findings suggest a possible link between lower liver function and the accumulation of core AD pathologies in the brain. These results also support the possibility that the liver-brain axis could be a potential target for therapeutic or preventive strategies against AD.
Collapse
Affiliation(s)
- Jee Wook Kim
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, 7 Keunjaebong-gil, Hwaseong, Gyeonggi, 18450, Republic of Korea; Department of Psychiatry, Hallym University College of Medicine, Chuncheon, Gangwon, 24252, Republic of Korea.
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea; Department of Psychiatry, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, 03080, Republic of Korea.
| | - Joon Hyung Jung
- Department of Psychiatry, Chungbuk National University Hospital, Cheongju, 28644, Republic of Korea.
| | - Nayeong Kong
- Department of Psychiatry, Keimyung University Dongsan Hospital, Daegu, 42601, Republic of Korea.
| | - Yoon Young Chang
- Department of Psychiatry, Inje University Sanggye Paik Hospital, Seoul, 01757, Republic of Korea.
| | - Gijung Jung
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea.
| | - Hyejin Ahn
- Interdisciplinary Program of Cognitive Science, Seoul National University College of Humanities, Seoul, 08826, Republic of Korea.
| | - Jun-Young Lee
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, 07061, Republic of Korea.
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, 03080, Republic of Korea.
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, 03080, Republic of Korea.
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, 07061, Republic of Korea.
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea; Department of Psychiatry, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, 03080, Republic of Korea; Interdisciplinary Program of Cognitive Science, Seoul National University College of Humanities, Seoul, 08826, Republic of Korea.
| |
Collapse
|
17
|
Liu Q, Wang YX, Ge ZH, Zhu MZ, Ding J, Wang H, Liu SM, Liu RC, Li C, Yu MJ, Feng Y, Zhu XH, Liang JH. Discovery of glycosidated glycyrrhetinic acid derivatives: Natural product-based soluble epoxide hydrolase inhibitors. Eur J Med Chem 2024; 280:116937. [PMID: 39413443 DOI: 10.1016/j.ejmech.2024.116937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/26/2024] [Accepted: 10/02/2024] [Indexed: 10/18/2024]
Abstract
There are few reports on soluble epoxide hydrolase (sEH) structure-activity relationship studies using natural product-based scaffolds. In this study, we discovered that C-30 urea derivatives of glycyrrhetinic acid such as 33, rather than C-20/C-3 urea derivatives, possess in vitro sEH inhibitory capabilities. Furthermore, we explored the impact of stereoconfigurations at C-3 and C-18 positions, and glycosidic bonds at the 3-OH on the compound's activity. Consequently, a glycoside of 33, specifically 49Cα containing alpha-oriented mannose, exhibited promising in vivo efficacy in alleviating carrageenan-induced paw edema and acetic acid-induced writhing. Meanwhile, 49Cα demonstrated potential in mitigating acute pancreatitis by modulating the ratios of anti-inflammatory epoxyeicosatrienoic acids (EETs) to pro-inflammatory dihydroxyeicosatrienoic acids (DHETs). The co-crystal structure of sEH in complex with 49Cα revealed that the N-tetrahydropyranylmethylene urea hydrogen bonded with the residues within the sEH tunnel, contrasting with the mannose component that extended beyond the tunnel's confines. Our findings highlight 49Cα (coded LQ-38) as a promising candidate for anti-inflammatory and analgesic effects, and pave the way for the future rational design of triterpenoid-based sEH inhibitors.
Collapse
Affiliation(s)
- Qian Liu
- Key Laboratory of Medicinal Molecule Science and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 102488, China
| | - Yi-Xin Wang
- Key Laboratory of Medicinal Molecule Science and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 102488, China
| | - Zi-Hao Ge
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Min-Zhen Zhu
- Research Center for Brain Health, PazhouLab, Guangzhou, 510330, China
| | - Jing Ding
- Key Laboratory of Medicinal Molecule Science and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 102488, China
| | - Hao Wang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Si-Meng Liu
- Key Laboratory of Medicinal Molecule Science and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 102488, China
| | - Rui-Chen Liu
- Key Laboratory of Medicinal Molecule Science and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 102488, China
| | - Chun Li
- Key Lab for Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| | - Ming-Jia Yu
- Key Laboratory of Medicinal Molecule Science and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 102488, China
| | - Yue Feng
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Xin-Hong Zhu
- Research Center for Brain Health, PazhouLab, Guangzhou, 510330, China.
| | - Jian-Hua Liang
- Key Laboratory of Medicinal Molecule Science and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 102488, China.
| |
Collapse
|
18
|
Yousef A, Fang L, Heidari M, Kranrod J, Seubert JM. The role of CYP-sEH derived lipid mediators in regulating mitochondrial biology and cellular senescence: implications for the aging heart. Front Pharmacol 2024; 15:1486717. [PMID: 39703395 PMCID: PMC11655241 DOI: 10.3389/fphar.2024.1486717] [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: 08/26/2024] [Accepted: 10/28/2024] [Indexed: 12/21/2024] Open
Abstract
Cellular senescence is a condition characterized by stable, irreversible cell cycle arrest linked to the aging process. The accumulation of senescent cells in the cardiac muscle can contribute to various cardiovascular diseases (CVD). Telomere shortening, epigenetic modifications, DNA damage, mitochondrial dysfunction, and oxidative stress are known contributors to the onset of cellular senescence in the heart. The link between mitochondrial processes and cellular senescence contributed to the age-related decline in cardiac function. These include changes in mitochondrial functions and behaviours that arise from various factors, including impaired dynamics, dysregulated biogenesis, mitophagy, mitochondrial DNA (mtDNA), reduced respiratory capacity, and mitochondrial structural changes. Thus, regulation of mitochondrial biology has a role in cellular senescence and cardiac function in aging hearts. Targeting senescent cells may provide a novel therapeutic approach for treating and preventing CVD associated with aging. CYP epoxygenases metabolize N-3 and N-6 polyunsaturated fatty acids (PUFA) into epoxylipids that are readily hydrolyzed to diol products by soluble epoxide hydrolase (sEH). Increasing epoxylipids levels or inhibition of sEH has demonstrated protective effects in the aging heart. Evidence suggests they may play a role in cellular senescence by regulating mitochondria, thus reducing adverse effects of aging in the heart. In this review, we discuss how mitochondria induce cellular senescence and how epoxylipids affect the senescence process in the aged heart.
Collapse
Affiliation(s)
- Ala Yousef
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Liye Fang
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Mobina Heidari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Joshua Kranrod
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - John M. Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
19
|
Luo Y, Yuan L, Liu Z, Dong W, Huang L, Liao A, Xie Y, Liu R, Lan W, Cai Y, Zhu W. Inhibition of PCSK9 Protects against Cerebral Ischemia‒Reperfusion Injury via Attenuating Microcirculatory Dysfunction. Neurochem Res 2024; 50:10. [PMID: 39548030 DOI: 10.1007/s11064-024-04272-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: 11/05/2023] [Revised: 08/16/2024] [Accepted: 09/18/2024] [Indexed: 11/17/2024]
Abstract
Proprotein convertase substilin/kexin type 9 (PCSK9), a pivotal protein regulating lipid metabolism, has been implicated in promoting microthrombotic formation and inflammatory cascades, thereby contributing to cardiovascular ischemia/reperfusion (I/R) injury. However, its involvement in cerebral I/R injury and its potential role in microcirculation protection remain unexplored. In this investigation, we utilized a middle cerebral artery occlusion/reperfusion (MCAO/R) mouse model to simulate ischemic stroke. Different concentrations of evolocumab (1, 5, 10 mg/kg, i.v.), a PCSK9 inhibitor, were administered to assess its impact. Immunofluorescence staining was employed to analyze changes in the expression of occludin, claudin-5, thrombocyte, ICAM-1, VCAM-1, and CD45, providing insights into blood-brain barrier integrity, platelet adhesion, and immune cell infiltration. Moreover, the Morris water maze and elevated plus maze were utilized to evaluate neurological and behavioral functions in MCAO/R mice, shedding light on the effects of PCSK9 inhibition. Our findings revealed a surge in plasma PCSK9 levels post-MCAO/R, peaking at 24 h post-reperfusion. Evolocumab (10 mg/kg) treatment significantly mitigated brain infarction and neurological deficits, evidenced by enhanced locomotor function and reduced post-stroke anxiety. However, it did not ameliorate cognitive impairment following MCAO/R. Additionally, evolocumab administration led to diminished leakage of evans blue solution and upregulated expression of occludin and claudin-5. Thrombocyte, ICAM-1, VCAM-1, and CD45 levels were notably reduced in the penumbral area post-evolocumab treatment. These protective effects are speculated to be mediated through the inhibition of the ERK/NF-κB pathway. The PCSK9 inhibitor evolocumab holds promise as a therapeutic agent during the acute phase of stroke, exerting its beneficial effects by modulating the ERK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yuanfei Luo
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Linying Yuan
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhihui Liu
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Weichen Dong
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Li Huang
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Anyu Liao
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yi Xie
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Rui Liu
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wenya Lan
- Department of Cerebrovascular Disease Treatment Center, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Yulong Cai
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Wusheng Zhu
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
20
|
Swer PB, Kharbuli B, Syiem D, Sharma R. Age-related decline in the expression of BRG1, ATM and ATR are partially reversed by dietary restriction in the livers of female mice. Biogerontology 2024; 25:1025-1037. [PMID: 38970714 DOI: 10.1007/s10522-024-10117-7] [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: 05/24/2024] [Accepted: 06/26/2024] [Indexed: 07/08/2024]
Abstract
BRG1 (Brahma-related gene 1) is a member of the SWI/SNF (switch/sucrose nonfermentable) chromatin remodeling complex which utilizes the energy from ATP hydrolysis for its activity. In addition to its role of regulating the expression of a vast array of genes, BRG1 mediates DNA repair upon genotoxic stress and regulates senescence. During organismal ageing, there is accumulation of unrepaired/unrepairable DNA damage due to progressive breakdown of the DNA repair machinery. The present study investigates the expression level of BRG1 as a function of age in the liver of 5- and 21-month-old female mice. It also explores the impact of dietary restriction on BRG1 expression in the old (21-month) mice. Salient findings of the study are: Real-time PCR and Western blot analyses reveal that BRG1 levels are higher in 5-month-old mice but decrease significantly with age. Dietary restriction increases BRG1 expression in the 21-month-old mice, nearly restoring it to the level observed in the younger group. Similar expression patterns are observed for DNA damage response genes ATM (Ataxia Telangiectasia Mutated) and ATR (Ataxia Telangiectasia and Rad3-related) with the advancement in age and which appears to be modulated by dietary restriction. BRG1 transcriptionally regulates ATM as a function of age and dietary restriction. These results suggest that BRG1, ATM and ATR are downregulated as mice age, and dietary restriction can restore their expression. This implies that dietary restriction may play a crucial role in regulating BRG1 and related gene expression, potentially maintaining liver repair and metabolic processes as mice age.
Collapse
Affiliation(s)
- Pynskhem Bok Swer
- Department of Biochemistry, North-Eastern Hill University, Shillong, 793022, India
| | | | - Donkupar Syiem
- Department of Biochemistry, North-Eastern Hill University, Shillong, 793022, India
| | - Ramesh Sharma
- Department of Biochemistry, North-Eastern Hill University, Shillong, 793022, India.
| |
Collapse
|
21
|
Zhao MM, Yang JJ, Hashimoto K. Soluble epoxide hydrolase: Mechanisms and therapeutic potential in psychiatric and neurological disorders. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 102:237-266. [PMID: 39929581 DOI: 10.1016/bs.apha.2024.10.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Soluble epoxide hydrolase (sEH), encoded by the EPHX2 gene, is a critical enzyme involved in the metabolism of polyunsaturated fatty acids, specifically anti-inflammatory epoxy fatty acids (EpFAs). By converting EpFAs into less active forms, sEH promotes inflammation. Preclinical data using knock-out and overexpression of the Ephx2 gene have demonstrated its key role in the development and progression of symptoms in various disease models. Inhibition of sEH increases EpFAs, thereby enhancing their anti-inflammatory effects and reducing the levels of pro-inflammatory mediators. Numerous preclinical studies suggest that sEH inhibitors show promise in reducing inflammation and its related symptoms across various diseases, highlighting their therapeutic potential. This chapter reviews the role of sEH in the development and progression of various disorders including psychiatric disorders (depression, schizophrenia, autism spectrum disorder), neurological disorders (Alzheimer's disease, Parkinson's disease, brain injury), and pain.
Collapse
Affiliation(s)
- Ming-Ming Zhao
- Chiba University Center for Forensic Mental Health, Chiba, Japan; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China; Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou University, Zhengzhou, P.R. China
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China; Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou University, Zhengzhou, P.R. China
| | - Kenji Hashimoto
- Chiba University Center for Forensic Mental Health, Chiba, Japan; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China.
| |
Collapse
|
22
|
Li P, Zhao J, Wei X, Luo L, Chu Y, Zhang T, Zhu A, Yan J. Acupuncture may play a key role in anti-depression through various mechanisms in depression. Chin Med 2024; 19:135. [PMID: 39367470 PMCID: PMC11451062 DOI: 10.1186/s13020-024-00990-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/28/2024] [Indexed: 10/06/2024] Open
Abstract
Depression has emerged as a significant global health concern, exerting a profound impact on individuals, as evidenced by its high prevalence and associated suicide rates. Considering its pervasive nature, the absence of optimal treatment modalities remains a challenge. Acupuncture has garnered substantial clinical and experimental validation for its efficacy in addressing diverse forms of depression, including postpartum, post-stroke, and adolescent depression. This article endeavors to elucidate the distinctive attributes and underlying mechanisms of acupuncture in the contemporary treatment of depression. Research has demonstrated that acupuncture exerts diverse physiological effects in animal models of depression, encompassing modulation of the brain, serum, and brain-gut axis. These effects are attributed to various mechanisms, including anti-inflammatory and anti-oxidative actions, promotion of neuronal plasticity, neuroprotection, neurotrophic effects, modulation of neurotransmitters, regulation of endocrine and immune functions, and modulation of cell signal pathways. Currently, the therapeutic mechanism of acupuncture involves the engagement of multiple targets, pathways, and bidirectional regulation. Hence, acupuncture emerges as a promising alternative medical modality, exhibiting substantial research prospects and meriting comprehensive worth further study and dissemination.
Collapse
Affiliation(s)
- Peng Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Department of clinical medicine, Xiamen medical college, xiamen, China
| | - Jiangna Zhao
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Xiuxiang Wei
- Rehabilitation Medicine Department, Shenzhen Hospital of Traditional Chinese and Western Medicine , Shenzhen, China
| | - Longfei Luo
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Yuzhou Chu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Tao Zhang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Anning Zhu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| | - Juntao Yan
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| |
Collapse
|
23
|
Liu H, Yu C, Qin C. The changes of peripheral blood hub genes in 24-week-old APP/PS1/Tau triple transgenic mouse model based on weighted gene co-expression network analysis. AN ACAD BRAS CIENC 2024; 96:e20240120. [PMID: 39383430 DOI: 10.1590/0001-3765202420240120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/06/2024] [Indexed: 10/11/2024] Open
Abstract
Peripheral regulation emerges as a promising intervention in the early stages of Alzheimer's disease (AD). The hub genes in the peripheral blood of MCI patients from GEO database (GSE63060, GSE63061) were screened using weighted gene co-expression analysis (WGCNA). Meanwhile, behavioral tests, HE staining and Nissl staining were used to detect the memory impairment and histopathological changes in 24-week-old male 3×Tg-AD mice. Thioflavin-S and immunohistochemical staining were used to determine the Aβ deposition in both intracellular and extracellular neurons. Subsequently, the MCI-hub genes were verified by quantitative real-time PCR (qRT-PCR) in the peripheral blood of 3×Tg-AD mice. The research revealed ten hub genes associated with MCI were identified WGCNA. Short-term memory loss, intracellular Aβ deposition and limited of extracellular amyloid plaques in 3×Tg-AD mice. The qRT-PCR analysis of peripheral blood from these mice revealed significantly down-regulation in the expression levels of ATP5C1, ITGB2, EFTUD2 and RPS27A genes; whereas the expression level of VCP gene was significantly up-regulated. These findings confirmed that 24-week-old male 3×Tg-AD mice were a valuable animal model for simulating the early symptomatic stages of AD. Additionally, the peripheral blood MCI-hub genes related to immune response, energy metabolism and ribosomal coding efficiency provide potential biomarkers for this stage.
Collapse
Affiliation(s)
- Hexu Liu
- First Affiliated Hospital of Guangxi Medical University, Department of Neurology, No. 6 ShuangYong Road, Nanning 530021, Guangxi Autonomous Region, China
| | - Changyin Yu
- Affiliated Hospital of Zunyi Medical University, Department of Neurology, No. 149 Dalian Road, Zunyi 563000, Guizhou Province, China
| | - Chao Qin
- First Affiliated Hospital of Guangxi Medical University, Department of Neurology, No. 6 ShuangYong Road, Nanning 530021, Guangxi Autonomous Region, China
| |
Collapse
|
24
|
Guo Z, Yin M, Sun C, Xu G, Wang T, Jia Z, Zhang Z, Zhu C, Zheng D, Wang L, Huang S, Liu D, Zhang Y, Xie R, Gao N, Zhan L, He S, Zhu Y, Li Y, Nashan B, Andrea S, Xu J, Zhao Q, He X. Liver protects neuron viability and electrocortical activity in post-cardiac arrest brain injury. EMBO Mol Med 2024; 16:2322-2348. [PMID: 39300235 PMCID: PMC11479250 DOI: 10.1038/s44321-024-00140-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: 02/24/2024] [Revised: 07/18/2024] [Accepted: 08/14/2024] [Indexed: 09/22/2024] Open
Abstract
Brain injury is the leading cause of mortality among patients who survive cardiac arrest (CA). Clinical studies have shown that the presence of post-CA hypoxic hepatitis or pre-CA liver disease is associated with increased mortality and inferior neurological recovery. In our in vivo global cerebral ischemia model, we observed a larger infarct area, elevated tissue injury scores, and increased intravascular CD45+ cell adhesion in reperfused brains with simultaneous hepatic ischemia than in those without it. In the ex vivo brain normothermic machine perfusion (NMP) model, we demonstrated that addition of a functioning liver to the brain NMP circuit significantly reduced post-CA brain injury, increased neuronal viability, and improved electrocortical activity. Furthermore, significant alterations were observed in both the transcriptome and metabolome in the presence or absence of hepatic ischemia. Our study highlights the crucial role of the liver in the pathogenesis of post-CA brain injury.
Collapse
Affiliation(s)
- Zhiyong Guo
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China.
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China.
| | - Meixian Yin
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chengjun Sun
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
- Department of Organ Transplantation, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Guixing Xu
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tielong Wang
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Zehua Jia
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Zhiheng Zhang
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Caihui Zhu
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Donghua Zheng
- Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Linhe Wang
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Shanzhou Huang
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Di Liu
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Yixi Zhang
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Rongxing Xie
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Ningxin Gao
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Liqiang Zhan
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Shujiao He
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Yifan Zhu
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Yuexin Li
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Björn Nashan
- Organ Transplant Center, The First Affiliated Hospital of the University of Science and Technology of China, Hefei, China
| | - Schlegel Andrea
- General and Liver Transplant Surgery Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Jin Xu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Qiang Zhao
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China.
| | - Xiaoshun He
- Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China.
| |
Collapse
|
25
|
Liu N, Haziyihan A, Zhao W, Chen Y, Chao H. Trajectory of brain-derived amyloid beta in Alzheimer's disease: where is it coming from and where is it going? Transl Neurodegener 2024; 13:42. [PMID: 39160618 PMCID: PMC11331646 DOI: 10.1186/s40035-024-00434-9] [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: 01/30/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurological disorder that primarily impacts cognitive function. Currently there are no disease-modifying treatments to stop or slow its progression. Recent studies have found that several peripheral and systemic abnormalities are associated with AD, and our understanding of how these alterations contribute to AD is becoming more apparent. In this review, we focuse on amyloid‑beta (Aβ), a major hallmark of AD, summarizing recent findings on the source of brain-derived Aβ and discussing where and how the brain-derived Aβ is cleared in vivo. Based on these findings, we propose future strategies for AD prevention and treatment, from a novel perspective on Aβ metabolism.
Collapse
Affiliation(s)
- Ni Liu
- Zhengzhou University, Zhengzhou, 450001, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China
| | | | - Wei Zhao
- Zhengzhou University, Zhengzhou, 450001, China
| | - Yu Chen
- Zhengzhou University, Zhengzhou, 450001, China
| | - Hongbo Chao
- Zhengzhou University, Zhengzhou, 450001, China.
- Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
26
|
Wei C, Li X, Jin Y, Zhang Y, Yuan Q. Does the liver facilitate aging-related cognitive impairment: Conversation between liver and brain during exercise? J Cell Physiol 2024; 239:e31287. [PMID: 38704693 DOI: 10.1002/jcp.31287] [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: 01/13/2024] [Revised: 03/24/2024] [Accepted: 04/16/2024] [Indexed: 05/07/2024]
Abstract
Liver, an important regulator of metabolic homeostasis, is critical for healthy brain function. In particular, age-related neurodegenerative diseases seriously reduce the quality of life for the elderly. As population aging progresses rapidly, unraveling the mechanisms that effectively delay aging has become critical. Appropriate exercise is reported to improve aging-related cognitive impairment. Whereas current studies focused on exploring the effect of exercise on the aging brain itself, ignoring the persistent effects of peripheral organs on the brain through the blood circulation. The aim of this paper is to summarize the communication and aging processes of the liver and brain and to emphasize the metabolic mechanisms of the liver-brain axis about exercise ameliorating aging-related neurodegenerative diseases. A comprehensive understanding of the potential mechanisms about exercise ameliorating aging is critical for improving adaptation to age-related brain changes and formulating effective interventions against age-related cognitive decline.
Collapse
Affiliation(s)
- Changling Wei
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan, China
| | - Xue Li
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan, China
| | - Yu Jin
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan, China
| | - Yuanting Zhang
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan, China
| | - Qiongjia Yuan
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan, China
| |
Collapse
|
27
|
Li S, Song H, Sun Y, Sun Y, Zhang H, Gao Z. Inhibition of soluble epoxide hydrolase as a therapeutic approach for blood-brain barrier dysfunction. Biochimie 2024; 223:13-22. [PMID: 38531484 DOI: 10.1016/j.biochi.2024.03.015] [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: 01/12/2024] [Revised: 03/20/2024] [Accepted: 03/23/2024] [Indexed: 03/28/2024]
Abstract
The blood-brain barrier (BBB) is a protective semi-permeable structure that regulates the exchange of biomolecules between the peripheral blood and the central nervous system (CNS). Due to its specialized tight junctions and low vesicle trafficking, the BBB strictly limits the paracellular passage and transcellular transport of molecules to maintain the physiological condition of brain tissues. BBB breakdown is associated with many CNS disorders. Soluble epoxide hydrolase (sEH) is a hydrolase enzyme that converts epoxy-fatty acids (EpFAs) to their corresponding diols and is involved in the onset and progression of multiple diseases. EpFAs play a protective role in the central nervous system via preventing neuroinflammation, making sEH a potential therapeutic target for CNS diseases. Recent studies showed that sEH inhibition prevented BBB impairment caused by stroke, hemorrhage, traumatic brain injury, hyperglycemia and sepsis via regulating the expression of tight junctions. In this review, the protective actions of sEH inhibition on BBB and potential mechanisms are summarized, and some important questions that remain to be resolved are also addressed.
Collapse
Affiliation(s)
- Shuo Li
- Hebei Province Key Laboratory of Molecular Chemistry for Drug, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, China
| | - Huijia Song
- Hebei Province Key Laboratory of Molecular Chemistry for Drug, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, China
| | - Yanping Sun
- Hebei Province Key Laboratory of Molecular Chemistry for Drug, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, China
| | - Yongjun Sun
- Hebei Province Key Laboratory of Molecular Chemistry for Drug, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, China
| | - Huimin Zhang
- Hebei Province Key Laboratory of Molecular Chemistry for Drug, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, China
| | - Zibin Gao
- Hebei Province Key Laboratory of Molecular Chemistry for Drug, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, China.
| |
Collapse
|
28
|
Yu X, Wang S, Ji Z, Meng J, Mou Y, Wu X, Yang X, Xiong P, Li M, Guo Y. Ferroptosis: An important mechanism of disease mediated by the gut-liver-brain axis. Life Sci 2024; 347:122650. [PMID: 38631669 DOI: 10.1016/j.lfs.2024.122650] [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: 02/22/2024] [Revised: 03/27/2024] [Accepted: 04/13/2024] [Indexed: 04/19/2024]
Abstract
AIMS As a unique iron-dependent non-apoptotic cell death, Ferroptosis is involved in the pathogenesis and development of many human diseases and has become a research hotspot in recent years. However, the regulatory role of ferroptosis in the gut-liver-brain axis has not been elucidated. This paper summarizes the regulatory role of ferroptosis and provides theoretical basis for related research. MATERIALS AND METHODS We searched PubMed, CNKI and Wed of Science databases on ferroptosis mediated gut-liver-brain axis diseases, summarized the regulatory role of ferroptosis on organ axis, and explained the adverse effects of related regulatory effects on various diseases. KEY FINDINGS According to our summary, the main way in which ferroptosis mediates the gut-liver-brain axis is oxidative stress, and the key cross-talk of ferroptosis affecting signaling pathway network is Nrf2/HO-1. However, there were no specific marker between different organ axes mediate by ferroptosis. SIGNIFICANCE Our study illustrates the main ways and key cross-talk of ferroptosis mediating the gut-liver-brain axis, providing a basis for future research.
Collapse
Affiliation(s)
- Xinxin Yu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Shihao Wang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Zhongjie Ji
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Jiaqi Meng
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Yunying Mou
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Xinyi Wu
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Xu Yang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Panyang Xiong
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Mingxia Li
- Nursing School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Yinghui Guo
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China.
| |
Collapse
|
29
|
Li Q, Li B, Liu L, Wang KJ, Liu MY, Deng Y, Li Z, Zhao WD, Wu LY, Chen YH, Zhang K. Monocytes release cystatin F dimer to associate with Aβ and aggravate amyloid pathology and cognitive deficits in Alzheimer's disease. J Neuroinflammation 2024; 21:125. [PMID: 38730470 PMCID: PMC11088181 DOI: 10.1186/s12974-024-03119-2] [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: 03/04/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Understanding the molecular mechanisms of Alzheimer's disease (AD) has important clinical implications for guiding therapy. Impaired amyloid beta (Aβ) clearance is critical in the pathogenesis of sporadic AD, and blood monocytes play an important role in Aβ clearance in the periphery. However, the mechanism underlying the defective phagocytosis of Aβ by monocytes in AD remains unclear. METHODS Initially, we collected whole blood samples from sporadic AD patients and isolated the monocytes for RNA sequencing analysis. By establishing APP/PS1 transgenic model mice with monocyte-specific cystatin F overexpression, we assessed the influence of monocyte-derived cystatin F on AD development. We further used a nondenaturing gel to identify the structure of the secreted cystatin F in plasma. Flow cytometry, enzyme-linked immunosorbent assays and laser scanning confocal microscopy were used to analyse the internalization of Aβ by monocytes. Pull down assays, bimolecular fluorescence complementation assays and total internal reflection fluorescence microscopy were used to determine the interactions and potential interactional amino acids between the cystatin F protein and Aβ. Finally, the cystatin F protein was purified and injected via the tail vein into 5XFAD mice to assess AD pathology. RESULTS Our results demonstrated that the expression of the cystatin F protein was specifically increased in the monocytes of AD patients. Monocyte-derived cystatin F increased Aβ deposition and exacerbated cognitive deficits in APP/PS1 mice. Furthermore, secreted cystatin F in the plasma of AD patients has a dimeric structure that is closely related to clinical signs of AD. Moreover, we noted that the cystatin F dimer blocks the phagocytosis of Aβ by monocytes. Mechanistically, the cystatin F dimer physically interacts with Aβ to inhibit its recognition and internalization by monocytes through certain amino acid interactions between the cystatin F dimer and Aβ. We found that high levels of the cystatin F dimer protein in blood contributed to amyloid pathology and cognitive deficits as a risk factor in 5XFAD mice. CONCLUSIONS Our findings highlight that the cystatin F dimer plays a crucial role in regulating Aβ metabolism via its peripheral clearance pathway, providing us with a potential biomarker for diagnosis and potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Qiang Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Bing Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China
| | - Li Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Department of Neurology, Shenyang Fifth People Hospital, Shenyang, 110023, China
| | - Kang-Ji Wang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China
| | - Ming-Yue Liu
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Ze Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China
| | - Wei-Dong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China.
| | - Li-Yong Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Yu-Hua Chen
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China.
| | - Ke Zhang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology,, Ministry of Public Health, China Medical University, 77 Puhe Road, Shenyang, 110122, China.
| |
Collapse
|
30
|
Wu Q, Jiang N, Wang Y, Song G, Li P, Fang Y, Xu L, Wang W, Xie M. Soluble epoxide hydrolase inhibitor (TPPU) alleviates ferroptosis by regulating CCL5 after intracerebral hemorrhage in mice. Biomed Pharmacother 2024; 172:116301. [PMID: 38377737 DOI: 10.1016/j.biopha.2024.116301] [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: 12/06/2023] [Revised: 02/16/2024] [Accepted: 02/17/2024] [Indexed: 02/22/2024] Open
Abstract
Soluble epoxide hydrolase (sEH) inhibition has been shown multiple beneficial effects against brain injuries of Intracerebral hemorrhage (ICH). However, the underlying mechanism of its neuroprotective effects after ICH has not been explained fully. Ferroptosis, a new form of iron-dependent programmed cell death, has been shown to be implicated in the secondary injuries after ICH. In this study, We examined whether sEH inhibition can alleviate brain injuries of ICH through inhibiting ferroptosis. Expression of several markers for ferroptosis was observed in the peri-hematomal brain tissues in mice after ICH. lip-1, a ferroptosis inhibitor, alleviated iron accumulation, lipid peroxidation and the secondary damages post-ICH in mice model. Intraperitoneal injection of 1-Trifluoromethoxyphenyl-3- (1-propionylpiperidin-4-yl)urea (TPPU), a highly selective sEH inhibitor, could inhibit ferroptosis and alleviate brain damages in ICH mice. Furthermore, RNA-sequencing was applied to explore the potential regulatory mechanism underlying the effects of TPPU in ferroptosis after ICH. C-C chemokine ligand 5 (CCL5) may be the key factor by which TPPU regulated ferroptosis after ICH since CCL5 antagonist could mimic the effects of TPPU and CCL5 reversed the inhibitive effect of TPPU on ferroptosis and the neuroprotective effects of TPPU on secondary damage after ICH. Taken together, these data indicate that ferroptosis is a key pathological feature of ICH and Soluble epoxide hydrolase inhibitor can exert neuroprotective effect by preventing ferroptosis after ICH.
Collapse
Affiliation(s)
- Qiao Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Na Jiang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Yao Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Guini Song
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Ping Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Yongkang Fang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Li Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| |
Collapse
|
31
|
Watanabe S, Souza FDC, Kusumoto I, Shen Q, Nitin N, Lein PJ, Taha AY. Intraperitoneally injected d11-11(12)-epoxyeicosatrienoic acid is rapidly incorporated and esterified within rat plasma and peripheral tissues but not the brain. Prostaglandins Leukot Essent Fatty Acids 2024; 202:102622. [PMID: 38954932 PMCID: PMC11613899 DOI: 10.1016/j.plefa.2024.102622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 07/04/2024]
Abstract
Epoxyeicosatrienoic acids (EpETrEs) are bioactive lipid mediators of arachidonic acid cytochrome P450 oxidation. In vivo, the free (unbound) form of EpETrEs regulate multiple processes including blood flow, angiogenesis and inflammation resolution. Free EpETrEs are thought to rapidly degrade via soluble epoxide hydrolase (sEH); yet, in many tissues, the majority of EpETrEs are esterified to complex lipids (e.g. phospholipids) suggesting that esterification may play a major role in regulating free, bioactive EpETrE levels. This hypothesis was tested by quantifying the metabolism of intraperitoneally injected free d11-11(12)-Epoxyeicosatrienoic acid (d11-11(12)-EpETrE) in male and female rats. Plasma and tissues (liver, adipose and brain) were obtained 3 to 4 min later and assayed for d11-11(12)-EpETrE and its sEH metabolite, d11-11,12-dihydroxyeicosatrienoic acid (d11-11,12-diHETrE) in both the free and esterified lipid fractions. In both males and females, the majority of injected tracer was recovered in liver followed by plasma and adipose. No tracer was detected in the brain, indicating that brain levels are maintained by endogenous synthesis from precursor fatty acids. In plasma, liver, and adipose, the majority (>54 %) of d11-11(12)-EpETrE was found esterified to phospholipids or neutral lipids (triglycerides and cholesteryl esters). sEH-derived d11-11,12-diHETrE was not detected in plasma or tissues, suggesting negligible conversion within the 3-4 min period post tracer injection. This study shows that esterification is the main pathway regulating free 11(12)-EpETrE levels in vivo.
Collapse
Affiliation(s)
- Sho Watanabe
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California, Davis, CA 95616, USA; Food Function Analysis Laboratory, Graduate School of Agricultural Science, Tohoku University, Miyagi, 9800845, Japan
| | - Felipe Da Costa Souza
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California, Davis, CA 95616, USA
| | - Ibuki Kusumoto
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California, Davis, CA 95616, USA; Food Function Analysis Laboratory, Graduate School of Agricultural Science, Tohoku University, Miyagi, 9800845, Japan
| | - Qing Shen
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California, Davis, CA 95616, USA
| | - Nitin Nitin
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California, Davis, CA 95616, USA; Department of Biological and Agricultural Engineering, University of California, Davis, One Shields Ave, Davis, CA 95616, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California, Davis, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA; MIND Institute, University of California-Davis, 2825 50th Street, Sacramento, CA 95817
| | - Ameer Y Taha
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California, Davis, CA 95616, USA; Center for Neuroscience, University of California-Davis, One Shields Avenue, Davis, CA 95616, USA; West Coast Metabolomics Center, Genome Center, University of California-Davis, Davis, CA 95616, USA.
| |
Collapse
|
32
|
Aging Biomarker Consortium, Jiang M, Zheng Z, Wang X, Chen Y, Qu J, Ding Q, Zhang W, Liu YS, Yang J, Tang W, Hou Y, He J, Wang L, Huang P, Li LC, He Z, Gao Q, Lu Q, Wei L, Wang YJ, Ju Z, Fan JG, Ruan XZ, Guan Y, Liu GH, Pei G, Li J, Wang Y. A biomarker framework for liver aging: the Aging Biomarker Consortium consensus statement. LIFE MEDICINE 2024; 3:lnae004. [PMID: 39872390 PMCID: PMC11749002 DOI: 10.1093/lifemedi/lnae004] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/29/2024] [Indexed: 01/11/2025]
Abstract
In human aging, liver aging per se not only increases susceptibility to liver diseases but also increases vulnerability of other organs given its central role in regulating metabolism. Total liver function tends to be well maintained in the healthy elderly, so liver aging is generally difficult to identify early. In response to this critical challenge, the Aging Biomarker Consortium of China has formulated an expert consensus on biomarkers of liver aging by synthesizing the latest scientific literature, comprising insights from both scientists and clinicians. This consensus provides a comprehensive assessment of biomarkers associated with liver aging and presents a systematic framework to characterize these into three dimensions: functional, imaging, and humoral. For the functional domain, we highlight biomarkers associated with cholesterol metabolism and liver-related coagulation function. For the imaging domain, we note that hepatic steatosis and liver blood flow can serve as measurable biomarkers for liver aging. Finally, in the humoral domain, we pinpoint hepatokines and enzymatic alterations worthy of attention. The aim of this expert consensus is to establish a foundation for assessing the extent of liver aging and identify early signs of liver aging-related diseases, thereby improving liver health and the healthy life expectancy of the elderly population.
Collapse
Affiliation(s)
| | - Mengmeng Jiang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhuozhao Zheng
- Department of Radiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Xuan Wang
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Yanhao Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qiurong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - You-Shuo Liu
- Department of Geriatrics, the Second Xiangya Hospital, and the Institute of Aging and Geriatrics, Central South University, Changsha 410011, China
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Weiqing Tang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Yunlong Hou
- Yiling Pharmaceutical Academician Workstation, Shijiazhuang 050035, China
| | - Jinhan He
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Pengyu Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Engineering Research Center of Pulmonary and Critical Care Medicine Technology and Device (Ministry of Education), Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Lin-Chen Li
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Zhiying He
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200092, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qian Lu
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education), School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Lai Wei
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Yan-Jiang Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou 510632, China
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xiong Zhong Ruan
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gang Pei
- Collaborative Innovation Center for Brain Science, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Yunfang Wang
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education), School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 102218, China
| |
Collapse
|
33
|
Liu XL, Yeerlan J, Liu Z, Bai Y, Wang Q, Yan Y, Xu L, Jia C, Zhang L. Past, Present, and Future of Liver-Brain Axis in Alzheimer's Disease: A Bibliometric Review. J Alzheimers Dis 2024; 101:1267-1280. [PMID: 39302376 DOI: 10.3233/jad-240688] [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: 09/22/2024]
Abstract
Background No effective drugs currently exist to cure Alzheimer's disease (AD) due to its complexity and the lack of understanding of the involved molecular signaling and pathways. The relationship between liver health and AD is now widely recognized. Still, molecular links and shared pathways between the liver and brain remain unclear, making the liver-brain axis in AD therapies a new area for exploration. However, bibliometric studies on this topic are lacking. Objective This study aims to review the liver-brain axis in AD and identify future research hotspots and trends through bibliometric analysis. Methods Articles and reviews related to AD and liver and its related diseases were searched in the Web of Science Core Collection (WoSCC) database up to 2024. Data were processed and visually analyzed using VOSviewer, CiteSpace, and Pajek. Results We collected 1,777 articles on AD and liver and its related diseases from 2,517 institutions across 80 countries. Keyword cluster analysis identified 11 clusters, with 'insulin resistance,' 'amyloid-beta,' 'apolipoprotein-E,' 'oxidative stress,' and 'inflammation' appearing most frequently, and exhibiting strong total link strength. These results indicate that these topics have been the primary focus of research on the liver-brain axis in AD. Conclusions This study is the first to comprehensively analyze the liver-brain axis in AD using bibliometric methods. The research results identify recent research frontiers and hotspots, aiding scholars in gaining a deeper understanding of the correlation between AD and the liver.
Collapse
Affiliation(s)
- Xin Lian Liu
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Pathology and Pathophysiology, Institute of Neuroscience, Chengdu Medical College, Chengdu, China
| | | | - Zhirong Liu
- Department of General Surgery, Chengdu Second People's Hospital, Chengdu, China
| | - Yang Bai
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Qin Wang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - YiRui Yan
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - LuKe Xu
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Cui Jia
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Pathology and Pathophysiology, Institute of Neuroscience, Chengdu Medical College, Chengdu, China
| | - LuShun Zhang
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Pathology and Pathophysiology, Institute of Neuroscience, Chengdu Medical College, Chengdu, China
| |
Collapse
|
34
|
Oh YT, Yang J, Stefanovski D, Hammock B, Youn JH. Determinants of Meal-Induced Changes in Circulating FFA Epoxides, Diols, and Diol-to-Epoxide Ratios as Indices of Soluble Epoxide Hydrolase Activity. Int J Mol Sci 2023; 24:17351. [PMID: 38139180 PMCID: PMC10743433 DOI: 10.3390/ijms242417351] [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: 10/24/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Soluble epoxide hydrolase (sEH) is an important enzyme for metabolic and cardiovascular health. sEH converts FFA epoxides (EpFAs), many of which are regulators of various cellular processes, to biologically less active diols. In human studies, diol (sEH product) to EpFA (sEH substrate) ratios in plasma or serum have been used as indices of sEH activity. We previously showed these ratios profoundly decreased in rats during acute feeding, possibly reflecting decreases in tissue sEH activities. The present study was designed to test which tissue(s) these measurements in the blood represent and if factors other than sEH activity, such as renal excretion or dietary intake of EpFAs and diols, significantly alter plasma EpFAs, diols, and/or their ratios. The results show that postprandial changes in EpFAs and diols and their ratios in plasma were very similar to those observed in the liver but not in other tissues, suggesting that the liver is largely responsible for these changes in plasma levels. EpFAs and diols were excreted into the urine, but their levels were not significantly altered by feeding, suggesting that renal excretion of EpFAs and diols may not play a major role in postprandial changes in circulating EpFAs, diols, or their ratios. Diet intake had significant impacts on circulating EpFA and diol levels but not on diol-to-EpFA (D-to-E) ratios, suggesting that these ratios, reflecting sEH activities, may not be significantly affected by the availability of sEH substrates (i.e., EpFAs). In conclusion, changes in FFA D-to-E ratios in plasma may reflect those in the liver, which may in turn represent sEH activities in the liver, and they may not be significantly affected by renal excretion or the dietary intake of EpFAs and diols.
Collapse
Affiliation(s)
- Young Taek Oh
- Department of Physiology and Neuroscience, University of Southern California Keck School of Medicine, Los Angeles, CA 90089, USA;
| | - Jun Yang
- Department of Entomology and Nematology, Comprehensive Cancer Center UCDMC, University of California, Davis, CA 95616, USA; (J.Y.); (B.H.)
| | - Darko Stefanovski
- Department of Biostatistics, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19146, USA;
| | - Bruce Hammock
- Department of Entomology and Nematology, Comprehensive Cancer Center UCDMC, University of California, Davis, CA 95616, USA; (J.Y.); (B.H.)
| | - Jang H. Youn
- Department of Physiology and Neuroscience, University of Southern California Keck School of Medicine, Los Angeles, CA 90089, USA;
| |
Collapse
|
35
|
Xu FR, Wei ZH, Xu XX, Zhang XG, Wei CJ, Qi XM, Li YH, Gao XL, Wu Y. The hypothalamic steroidogenic pathway mediates susceptibility to inflammation-evoked depression in female mice. J Neuroinflammation 2023; 20:293. [PMID: 38062440 PMCID: PMC10704691 DOI: 10.1186/s12974-023-02976-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Depression is two-to-three times more frequent among women. The hypothalamus, a sexually dimorphic area, has been implicated in the pathophysiology of depression. Neuroinflammation-induced hypothalamic dysfunction underlies behaviors associated with depression. The lipopolysaccharide (LPS)-induced mouse model of depression has been well-validated in numerous laboratories, including our own, and is widely used to investigate the relationship between neuroinflammation and depression. However, the sex-specific differences in metabolic alterations underlying depression-associated hypothalamic neuroinflammation remain unknown. METHODS Here, we employed the LPS-induced mouse model of depression to investigate hypothalamic metabolic changes in both male and female mice using a metabolomics approach. Through bioinformatics analysis, we confirmed the molecular pathways and biological processes associated with the identified metabolites. Furthermore, we employed quantitative real-time PCR, enzyme-linked immunosorbent assay, western blotting, and pharmacological interventions to further elucidate the underlying mechanisms. RESULTS A total of 124 and 61 differential metabolites (DMs) were detected in male and female mice with depressive-like behavior, respectively, compared to their respective sex-matched control groups. Moreover, a comparison between female and male model mice identified 37 DMs. We capitalized on biochemical clustering and functional enrichment analyses to define the major metabolic changes in these DMs. More than 55% of the DMs clustered into lipids and lipid-like molecules, and an imbalance in lipids metabolism was presented in the hypothalamus. Furthermore, steroidogenic pathway was confirmed as a potential sex-specific pathway in the hypothalamus of female mice with depression. Pregnenolone, an upstream component of the steroid hormone biosynthesis pathway, was downregulated in female mice with depressive-like phenotypes but not in males and had considerable relevance to depressive-like behaviors in females. Moreover, exogenous pregnenolone infusion reversed depressive-like behaviors in female mice with depression. The 5α-reductase type I (SRD5A1), a steroidogenic hub enzyme involved in pregnenolone metabolism, was increased in the hypothalamus of female mice with depression. Its inhibition increased hypothalamic pregnenolone levels and ameliorated depressive-like behaviors in female mice with depression. CONCLUSIONS Our study findings demonstrate a marked sexual dimorphism at the metabolic level in depression, particularly in hypothalamic steroidogenic metabolism, identifying a potential sex-specific pathway in female mice with depressive-like behaviors.
Collapse
Affiliation(s)
- Fu-Rong Xu
- Department of Nursing, The Second People's Hospital of Wuwei, Wuwei, 733000, China
| | - Zhen-Hong Wei
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, 730000, China
- Institute of Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Xiao-Xia Xu
- Department of Nursing, People's Hospital of Wuwei, Wuwei, 733000, China
| | - Xiao-Gang Zhang
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Chao-Jun Wei
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, 730000, China
- Institute of Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Xiao-Ming Qi
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, 730000, China
- Institute of Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Yong-Hong Li
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, 730000, China.
- Institute of Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, 730000, China.
| | - Xiao-Ling Gao
- The Clinical Laboratory Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570100, China.
| | - Yu Wu
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, 730000, China.
- Institute of Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, 730000, China.
- School of Psychology, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
36
|
Yan M, Man S, Sun B, Ma L, Guo L, Huang L, Gao W. Gut liver brain axis in diseases: the implications for therapeutic interventions. Signal Transduct Target Ther 2023; 8:443. [PMID: 38057297 PMCID: PMC10700720 DOI: 10.1038/s41392-023-01673-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/10/2023] [Accepted: 09/28/2023] [Indexed: 12/08/2023] Open
Abstract
Gut-liver-brain axis is a three-way highway of information interaction system among the gastrointestinal tract, liver, and nervous systems. In the past few decades, breakthrough progress has been made in the gut liver brain axis, mainly through understanding its formation mechanism and increasing treatment strategies. In this review, we discuss various complex networks including barrier permeability, gut hormones, gut microbial metabolites, vagus nerve, neurotransmitters, immunity, brain toxic metabolites, β-amyloid (Aβ) metabolism, and epigenetic regulation in the gut-liver-brain axis. Some therapies containing antibiotics, probiotics, prebiotics, synbiotics, fecal microbiota transplantation (FMT), polyphenols, low FODMAP diet and nanotechnology application regulate the gut liver brain axis. Besides, some special treatments targeting gut-liver axis include farnesoid X receptor (FXR) agonists, takeda G protein-coupled receptor 5 (TGR5) agonists, glucagon-like peptide-1 (GLP-1) receptor antagonists and fibroblast growth factor 19 (FGF19) analogs. Targeting gut-brain axis embraces cognitive behavioral therapy (CBT), antidepressants and tryptophan metabolism-related therapies. Targeting liver-brain axis contains epigenetic regulation and Aβ metabolism-related therapies. In the future, a better understanding of gut-liver-brain axis interactions will promote the development of novel preventative strategies and the discovery of precise therapeutic targets in multiple diseases.
Collapse
Affiliation(s)
- Mengyao Yan
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Shuli Man
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China.
| | - Benyue Sun
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Lanping Guo
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700, Beijing, China.
| | - Luqi Huang
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, 300072, Tianjin, China.
| |
Collapse
|
37
|
Zhang Y, Fang XM. The pan-liver network theory: From traditional chinese medicine to western medicine. CHINESE J PHYSIOL 2023; 66:401-436. [PMID: 38149555 DOI: 10.4103/cjop.cjop-d-22-00131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023] Open
Abstract
In traditional Chinese medicine (TCM), the liver is the "general organ" that is responsible for governing/maintaining the free flow of qi over the entire body and storing blood. According to the classic five elements theory, zang-xiang theory, yin-yang theory, meridians and collaterals theory, and the five-viscera correlation theory, the liver has essential relationships with many extrahepatic organs or tissues, such as the mother-child relationships between the liver and the heart, and the yin-yang and exterior-interior relationships between the liver and the gallbladder. The influences of the liver to the extrahepatic organs or tissues have been well-established when treating the extrahepatic diseases from the perspective of modulating the liver by using the ancient classic prescriptions of TCM and the acupuncture and moxibustion. In modern medicine, as the largest solid organ in the human body, the liver has the typical functions of filtration and storage of blood; metabolism of carbohydrates, fats, proteins, hormones, and foreign chemicals; formation of bile; storage of vitamins and iron; and formation of coagulation factors. The liver also has essential endocrine function, and acts as an immunological organ due to containing the resident immune cells. In the perspective of modern human anatomy, physiology, and pathophysiology, the liver has the organ interactions with the extrahepatic organs or tissues, for example, the gut, pancreas, adipose, skeletal muscle, heart, lung, kidney, brain, spleen, eyes, skin, bone, and sexual organs, through the circulation (including hemodynamics, redox signals, hepatokines, metabolites, and the translocation of microbiota or its products, such as endotoxins), the neural signals, or other forms of pathogenic factors, under normal or diseases status. The organ interactions centered on the liver not only influence the homeostasis of these indicated organs or tissues, but also contribute to the pathogenesis of cardiometabolic diseases (including obesity, type 2 diabetes mellitus, metabolic [dysfunction]-associated fatty liver diseases, and cardio-cerebrovascular diseases), pulmonary diseases, hyperuricemia and gout, chronic kidney disease, and male and female sexual dysfunction. Therefore, based on TCM and modern medicine, the liver has the bidirectional interaction with the extrahepatic organ or tissue, and this established bidirectional interaction system may further interact with another one or more extrahepatic organs/tissues, thus depicting a complex "pan-hepatic network" model. The pan-hepatic network acts as one of the essential mechanisms of homeostasis and the pathogenesis of diseases.
Collapse
Affiliation(s)
- Yaxing Zhang
- Department of Physiology; Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong; Issue 12th of Guangxi Apprenticeship Education of Traditional Chinese Medicine (Shi-Cheng Class of Guangxi University of Chinese Medicine), College of Continuing Education, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xian-Ming Fang
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine (Guangxi Hospital of Integrated Chinese Medicine and Western Medicine, Ruikang Clinical Faculty of Guangxi University of Chinese Medicine), Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
38
|
Inoue Y, Bamkole M, Kanekiyo T. Hepatic soluble epoxide hydrolase: A promising target for unveiling the liver-brain axis in Alzheimer's disease. Neuron 2023; 111:2775-2777. [PMID: 37734320 DOI: 10.1016/j.neuron.2023.08.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 09/23/2023]
Abstract
Wu and Dong et al.1 report that hepatic soluble epoxide hydrolase (sEH) manipulation impacts amyloid-β (Aβ) deposits and cognitive impairment in mouse models for Alzheimer's disease (AD), suggesting that hepatic sEH activity is a promising therapeutic target to treat AD.
Collapse
Affiliation(s)
- Yasuteru Inoue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Michael Bamkole
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|