1
|
Alves RL, Gonçalves A, Voytyuk I, Harrison DC. Behaviour profile characterization of PS19 and rTg4510 tauopathy mouse models: A systematic review and a meta-analysis. Exp Neurol 2025; 389:115234. [PMID: 40185359 DOI: 10.1016/j.expneurol.2025.115234] [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: 01/17/2025] [Revised: 03/14/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
The rTg4510 and PS19 mouse models are widely used in tauopathy research. Alzheimer's disease (AD) is the most prevalent among tauopathies. Behavioural tests are frequently used to assess emotional, cognitive, and motor behaviours in mouse models of AD. Cognitive deficits begin to manifest in rTg4510 mice around 3 months of age and in PS19 mice around 6 months. However, it's widely recognized that behavioural outcomes can vary due to environmental factors, health status, and husbandry practices, causing phenotypic differences between facilities. This study aims to consolidate current knowledge of the behavioural phenotypes of these two mouse models. We conducted a comprehensive literature review using keyword searches with Boolean operators across databases up to January 2024. Additional studies were included from manual searches. A total of 23 articles were reviewed for rTg4510 mice and 52 for PS19 mice. We extracted methodological details and key findings from each study. Results for rTg4510 mice show consistent findings regarding locomotion, memory and learning, and neurological dysfunction. However, the limited studies on motor and balance behaviour revealed no significant differences, while anxiety-like behaviour showed some inconsistencies. PS19 mice demonstrate more robust results for anxiety-like behaviour, memory and learning, and locomotion, while findings for balance and coordination are more inconsistent. Although there is overall coherence in certain aspects of the behavioural profiles of these tauopathy mouse models, it is crucial to recognize experimental heterogeneity and profile behavioural baselines to optimize the testing of both genetic and pharmacological interventions.
Collapse
Affiliation(s)
- Renata L Alves
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, United Kingdom.
| | | | - Iryna Voytyuk
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, United Kingdom
| | - David C Harrison
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, United Kingdom
| |
Collapse
|
2
|
Attanasio S. Autophagy in cancer and protein conformational disorders. FEBS Lett 2025. [PMID: 40342093 DOI: 10.1002/1873-3468.70061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025]
Abstract
Autophagy is a catabolic process by which cells maintain cellular homeostasis through the degradation of dysfunctional cytoplasmic components, such as toxic misfolded proteins and damaged organelles, within the lysosome. It is a multistep process that is tightly regulated by nutrient, energy, and stress-sensing mechanisms. Autophagy plays a pivotal role in various biological processes, including protein and organelle quality control, defense against pathogen infections, cell metabolism, and immune surveillance. As a result, autophagy dysfunction is linked to a variety of pathological conditions. The role of autophagy in cancer is complex and dynamic. Depending on the context, autophagy can have both tumor-suppressive and pro-tumorigenic effects. In contrast, its role is more clearly defined in protein conformational disorders, where autophagy serves as a mechanism to reduce toxic protein aggregation, thereby improving cellular homeostasis. Because autophagy-based therapies hold promising potential for the treatment of cancer and protein conformational disorders, this review will highlight the latest findings and advancements in these areas.
Collapse
Affiliation(s)
- Sergio Attanasio
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
3
|
Banerjee A, Thekkekkara D, Manjula SN, Nair SP, Lalitha MS. Correlation of autophagy and Alzheimer's disease with special emphasis on the role of phosphodiesterase-4. 3 Biotech 2025; 15:139. [PMID: 40292249 PMCID: PMC12018668 DOI: 10.1007/s13205-025-04306-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Autophagy disruption is important in Alzheimer's disease (AD) as it prevents misfolded proteins from being removed, which leads to the accumulation of amyloid plaques and neurofibrillary tangles (NFTs). Restoring autophagy improves neuronal survival and cognitive function, according to experimental models. In AD models, mTOR inhibition and AMPK activation enhance synaptic plasticity and lessen learning deficits. Inhibitors of phosphodiesterase-4 (PDE4) improve cognition and reduce neuroinflammation via altering cyclic adenosine monophosphate (cAMP) transmission. Furthermore, autophagic-lysosomal clearance is encouraged by upregulating transcription factor EB (TFEB), which lessens the pathogenic damage linked to AD. These results point to autophagy modification as a promising therapeutic approach, with the mTOR, AMPK, cAMP, and TFEB pathways being possible targets for drugs. Though much evidence is based on animal studies, these findings provide valuable insights into autophagy's role in AD pathology, offering promising directions for future research and drug development.
Collapse
Affiliation(s)
- Aniruddha Banerjee
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Dithu Thekkekkara
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - S. N. Manjula
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Salini P. Nair
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Mankala Sree Lalitha
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| |
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
|
Du K, Chen H, Pan Z, Zhao M, Cheng S, Luo Y, Zhang W, Li D. Small-molecule activation of TFEB alleviates Niemann-Pick disease type C via promoting lysosomal exocytosis and biogenesis. eLife 2025; 13:RP103137. [PMID: 40184172 PMCID: PMC11970905 DOI: 10.7554/elife.103137] [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: 04/05/2025] Open
Abstract
Niemann-Pick disease type C (NPC) is a devastating lysosomal storage disease characterized by abnormal cholesterol accumulation in lysosomes. Currently, there is no treatment for NPC. Transcription factor EB (TFEB), a member of the microphthalmia transcription factors (MiTF), has emerged as a master regulator of lysosomal function and promoted the clearance of substrates stored in cells. However, it is not known whether TFEB plays a role in cholesterol clearance in NPC disease. Here, we show that transgenic overexpression of TFEB, but not TFE3 (another member of MiTF family) facilitates cholesterol clearance in various NPC1 cell models. Pharmacological activation of TFEB by sulforaphane (SFN), a previously identified natural small-molecule TFEB agonist by us, can dramatically ameliorate cholesterol accumulation in human and mouse NPC1 cell models. In NPC1 cells, SFN induces TFEB nuclear translocation via a ROS-Ca2+-calcineurin-dependent but MTOR-independent pathway and upregulates the expression of TFEB-downstream genes, promoting lysosomal exocytosis and biogenesis. While genetic inhibition of TFEB abolishes the cholesterol clearance and exocytosis effect by SFN. In the NPC1 mouse model, SFN dephosphorylates/activates TFEB in the brain and exhibits potent efficacy of rescuing the loss of Purkinje cells and body weight. Hence, pharmacological upregulating lysosome machinery via targeting TFEB represents a promising approach to treat NPC and related lysosomal storage diseases, and provides the possibility of TFEB agonists, that is, SFN as potential NPC therapeutic candidates.
Collapse
Affiliation(s)
- Kaili Du
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of TechnologyHangzhouChina
- Department of Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
| | - Hongyu Chen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of TechnologyHangzhouChina
| | - Zhaonan Pan
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of TechnologyHangzhouChina
| | - Mengli Zhao
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of TechnologyHangzhouChina
| | - Shixue Cheng
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of TechnologyHangzhouChina
| | - Yu Luo
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of TechnologyHangzhouChina
| | - Wenhe Zhang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of TechnologyHangzhouChina
| | - Dan Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of TechnologyHangzhouChina
- Department of Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
| |
Collapse
|
6
|
Wu Y, Li T, Jiang X, Ling J, Zhao Z, Zhu J, Chen C, Liu Q, Yang X, Shen X, Ma R, Li G, Liu G. (-)-Epicatechin Rescues Memory Deficits by Activation of Autophagy in a Mouse Model of Tauopathies. MedComm (Beijing) 2025; 6:e70144. [PMID: 40135197 PMCID: PMC11933444 DOI: 10.1002/mco2.70144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 01/23/2025] [Accepted: 02/13/2025] [Indexed: 03/27/2025] Open
Abstract
In tauopathies, defects in autophagy-lysosomal protein degradation are thought to contribute to the abnormal accumulation of aggregated tau. Recent studies have shown that (-)-Epicatechin (Epi), a dietary flavonoid belonging to the flavan-3-ol subgroup, improves blood flow, modulates metabolic profiles, and prevents oxidative damage. However, less research has explored the effects of Epi on tauopathies. Here, we found that Epi rescued cognitive deficits in P301S tau transgenic mice, a model exhibiting characteristics of tauopathies like frontotemporal dementia and Alzheimer's disease, and attenuated tau pathology through autophagy activation. Proteomic and biochemical analyses revealed that P301S mice exhibit deficits in autophagosome formation via modulating mTOR, consequently inhibiting autophagy. Epi inhibited the mTOR signaling pathway to promote autophagosome formation, which is essential for the clearance of tau aggregation. By using chloroquine (CQ) to inhibit autophagy in vivo, we further confirmed that Epi induced tau degradation via the autophagy pathway. Lastly, Epi administration was also found to improve cognition by reversing spine decrease and neuron loss, as well as attenuating neuroinflammation. Our findings suggest that Epi promoted tau clearance by activating autophagy, indicating its potential as a promising therapeutic candidate for tauopathies.
Collapse
Affiliation(s)
- Yanqing Wu
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Health Management CenterRenmin Hospital of Wuhan UniversityWuhanChina
| | - Ting Li
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Ministry of Education of China and Hubei Province for Neurological DisordersTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of PathologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Xingjun Jiang
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jianmin Ling
- Department of Emergency MedicineTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Critical Care MedicineTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zaihua Zhao
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational EnvironmentSchool of Public HealthAir Force Medical UniversityXi'anChina
| | - Jiahui Zhu
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chongyang Chen
- Key Laboratory of Nuclear MedicineMinistry of HealthJiangsu Key Laboratory of Molecular Nuclear MedicineJiangsu Institute of Nuclear MedicineWuxiChina
| | - Qian Liu
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Ministry of Education of China and Hubei Province for Neurological DisordersTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of ShenzhenShenzhen Center for Disease Control and PreventionShenzhenChina
| | - Xuefeng Shen
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational EnvironmentSchool of Public HealthAir Force Medical UniversityXi'anChina
| | - Rong Ma
- Department of PharmacologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Gang Li
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Gongping Liu
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Ministry of Education of China and Hubei Province for Neurological DisordersTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Modern Toxicology of ShenzhenShenzhen Center for Disease Control and PreventionShenzhenChina
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| |
Collapse
|
7
|
Song J, Wang T, Hong JS, Wang Y, Feng J. TFEB-dependent autophagy-lysosomal pathway is required for NRF2-driven antioxidative action in obstructive sleep apnea-induced neuronal injury. Cell Signal 2025; 128:111630. [PMID: 39875050 PMCID: PMC11913475 DOI: 10.1016/j.cellsig.2025.111630] [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: 10/24/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025]
Abstract
Nearly one billion individuals worldwide suffer from obstructive sleep apnea (OSA) and are potentially impacted by related neurodegeneration. TFEB is considered a master regulator of autophagy and lysosomal biogenesis, but little is known about its role in neuronal oxidative stress and resultant injury induced by OSA. This study aimed to investigate these issues. Here, we demonstrated that neuronal TFEB induction is repressed in OSA mouse models. Activation of a TFEB-dependent autophagy-lysosomal pathway (ALP) reduces hippocampal neuronal cell death and mitigates OSA-related cognitive impairment. Neuronal NRF2 induction was also found to be defective in OSA mouse models. A series of staining assays for HO1, SOD3, ROS, GSH, 8-OHdG, MDA and PI revealed that enhancement of NRF2 expression restores neuronal redox balance and protects hippocampal neurons. We then identified a novel interplay between TFEB-dependent ALP and NRF2-mediated relief of oxidative stress. Inhibition of NRF2 hinders TFEB expression and lysosomal biogenesis. Conversely, knockdown of TFEB or blocking autophagy dampens the antioxidative effect of NRF2. Our findings highlight the unexpected and crucial role of TFEB-dependent ALP as a downstream event of NRF2 in NRF2-promoted redox balance. This study provides novel insights into the mechanism behind NRF2-driven antioxidative action and the regulation of TFEB-dependent ALP.
Collapse
Affiliation(s)
- Junxiu Song
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052 Tianjin, China
| | - Tian Wang
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052 Tianjin, China; Respiratory Department, Cangzhou People's Hospital, 061000, Hebei, China
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Dr., Research Triangle Park, Durham, NC 27709, USA
| | - Yubao Wang
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052 Tianjin, China.
| | - Jing Feng
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052 Tianjin, China.
| |
Collapse
|
8
|
Das S, Murumulla L, Ghosh P, Challa S. Heavy metal-induced disruption of the autophagy-lysosomal pathway: implications for aging and neurodegenerative disorders. Biometals 2025; 38:371-417. [PMID: 39960543 DOI: 10.1007/s10534-025-00665-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: 10/01/2024] [Accepted: 01/19/2025] [Indexed: 04/03/2025]
Abstract
Heavy metals such as lead, mercury, cadmium, magnesium, manganese, arsenic, copper pose considerable threats to neuronal health and are increasingly recognized as factors contributing to aging-related neurodegeneration. Exposure to these environmental toxins disrupts cellular homeostasis, resulting in oxidative stress and compromising critical cellular processes, particularly the autophagy-lysosomal pathway. This pathway is vital for preserving cellular integrity by breaking down damaged proteins and organelles; however, toxicity from heavy metals can hinder this function, leading to the buildup of harmful substances, inflammation, and increased neuronal injury. As individuals age, the consequences of neurodegeneration become more significant, raising the likelihood of developing disorders like Alzheimer's and Parkinson's disease. This review explores the intricate relationship between heavy metal exposure, dysfunction of the autophagy-lysosomal pathway, and aging-related neurodegeneration, emphasizing the urgent need for a comprehensive understanding of these mechanisms. The insights gained from this analysis are crucial for creating targeted therapeutic approaches aimed at alleviating the harmful effects of heavy metals on neuronal health and improving cellular resilience in aging populations.
Collapse
Affiliation(s)
- Shrabani Das
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Hyderabad, Telangana, 500007, India
| | - Lokesh Murumulla
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Hyderabad, Telangana, 500007, India
| | - Pritha Ghosh
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Hyderabad, Telangana, 500007, India
| | - Suresh Challa
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Hyderabad, Telangana, 500007, India.
| |
Collapse
|
9
|
Hu W, Chen M, Lin Y, Zhang H, Sun L, Shao W, Ye Y, Cheng Y, Zhou S, Hu P, Wu X, Xu Y, Wang K. Neuronal CD47 induces behavioral alterations and ameliorates microglial synaptic pruning in wild-type and Alzheimer's mouse models. Cell Biosci 2025; 15:38. [PMID: 40140948 PMCID: PMC11948738 DOI: 10.1186/s13578-025-01378-x] [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: 01/15/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Microglia are brain-resident macrophages that play a crucial role in synapse pruning during the development and progression of various neuropsychiatric disorders, including autism spectrum disorder (ASD) and Alzheimer's disease (AD). Mechanistically, CD47 protein acts as a potent 'do not eat me' signal, protecting synapses from phagocytosis by microglia. However, the functional role of the upregulated neuronal CD47 signal under both physiological and pathological conditions remains unclear. RESULTS We utilized an adeno-associated virus gene expression system to induce neuron-specific overexpression of CD47 in wild-type and 5xFAD mice, assessing its effects on microglial synaptic phagocytosis and mouse behaviors. Our results indicate that neuronal CD47 induces ASD-like behaviors and synaptic pruning defects, while promoting behavioral disinhibition and improving memory in wild-type mice. Single-nucleus RNA sequencing was employed to profile gene expression patterns in subpopulations of neurons and microglia. Notably, neuronal CD47 enhances synaptic pathways in neurons and particularly shifts microglial subpopulations from a disease-associated state to a homeostatic state. Additionally, neuronal CD47 reduces excessive microglial synaptic phagocytosis induced by Aβ pathology in 5xFAD mice. CONCLUSION Our study provides evidence that neuronal CD47 overexpression results in synaptic pruning defects and is involved in the pathogenesis of ASD, while also playing a beneficial role in mitigating excessive synaptic loss in Alzheimer's disease.
Collapse
Affiliation(s)
- Wenjie Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Mengting Chen
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Yuxue Lin
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Hui Zhang
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Li Sun
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Wei Shao
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Yuping Ye
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Yujie Cheng
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Shanshan Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
| | - Panpan Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
| | - Xingqi Wu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
| | - Yin Xu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China.
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China.
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China.
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China.
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China.
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China.
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China.
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China.
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China.
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China.
- Anhui Institute of Translational Medicine, Hefei, 230032, China.
| |
Collapse
|
10
|
Zakaria D, Yamashita T, Kosugi Y. Multi-Omics Analysis of Hippocampus in Rats Administered Trimethyltin Chloride. Neurotox Res 2025; 43:13. [PMID: 40095120 PMCID: PMC11914309 DOI: 10.1007/s12640-025-00737-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025]
Abstract
Trimethyltin chloride (TMT) is a neurotoxicant that damages the central nervous system (CNS) and triggers neurodegeneration. This study used multi-omic data, including transcriptomics and proteomics of the rat hippocampus, to identify differentially expressed genes and proteins in TMT-induced neurotoxicity over time, related to neuro-axonal damage marked by plasma Neurofilament Light (NfL) levels. Data were collected at 12, 24, 48, 72, and 168 h post-TMT administration. NfL levels surged at 72 and 168 h, confirming neuro-axonal damage. Transcripts of genes in the chemokine signaling pathway (Cxcl10, Cxcl12, Cxcl14, Cxcl16), apoptosis pathway (Caspase-3, PARP1, CTSD), and TNF signaling pathway (TNFR1, MMP9, ICAM-1, TRAF3) showed significant differential expression starting from 48 h, preceding the NfL increase, suggesting their roles in neuro-axonal damage. Additionally, 11 Alzheimer's disease-related proteins, with significant changes from 72 to 168 h, were detected only in the proteomic dataset, indicating post-translational modifications might be crucial in neurotoxicity. Pathway analysis revealed that neurodegeneration and Alzheimer's disease pathways were among the top 15 affected by TMT-induced gene regulation, aligning with the involvement of TNF signaling, apoptosis, and chemokine signaling in neurodegeneration. This research highlighted the value of longitudinal omics studies, combined with pathway enrichment, gene-disease association, and neuro-axonal damage biomarker analyses, to elucidate neurotoxicant-induced neurodegeneration. Findings from this study could enhance the understanding of TMT-induced neurotoxicity, potentially informing future therapeutic strategies and preventive measures.
Collapse
Affiliation(s)
- Douaa Zakaria
- DMPK&Modeling, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi, 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
- The Graduate University for Advanced Studies, SOKENDAI, Miura, Kanagawa, Japan
| | - Tomoki Yamashita
- DMPK&Modeling, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi, 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Yohei Kosugi
- DMPK&Modeling, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi, 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan.
| |
Collapse
|
11
|
Zeng J, Indajang J, Pitt D, Lo CH. Lysosomal acidification impairment in astrocyte-mediated neuroinflammation. J Neuroinflammation 2025; 22:72. [PMID: 40065324 PMCID: PMC11892208 DOI: 10.1186/s12974-025-03410-w] [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: 05/02/2024] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Astrocytes are a major cell type in the central nervous system (CNS) that play a key role in regulating homeostatic functions, responding to injuries, and maintaining the blood-brain barrier. Astrocytes also regulate neuronal functions and survival by modulating myelination and degradation of pathological toxic protein aggregates. Astrocytes have recently been proposed to possess both autophagic activity and active phagocytic capability which largely depend on sufficiently acidified lysosomes for complete degradation of cellular cargos. Defective lysosomal acidification in astrocytes impairs their autophagic and phagocytic functions, resulting in the accumulation of cellular debris, excessive myelin and lipids, and toxic protein aggregates, which ultimately contributes to the propagation of neuroinflammation and neurodegenerative pathology. Restoration of lysosomal acidification in impaired astrocytes represent new neuroprotective strategy and therapeutic direction. In this review, we summarize pathogenic factors, including neuroinflammatory signaling, metabolic stressors, myelin and lipid mediated toxicity, and toxic protein aggregates, that contribute to lysosomal acidification impairment and associated autophagic and phagocytic dysfunction in astrocytes. We discuss the role of lysosomal acidification dysfunction in astrocyte-mediated neuroinflammation primarily in the context of neurodegenerative diseases along with other brain injuries. We then highlight re-acidification of impaired lysosomes as a therapeutic strategy to restore autophagic and phagocytic functions as well as lysosomal degradative capacity in astrocytes. We conclude by providing future perspectives on the role of astrocytes as phagocytes and their crosstalk with other CNS cells to impart neurodegenerative or neuroprotective effects.
Collapse
Affiliation(s)
- Jialiu Zeng
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA.
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, 13244, USA.
| | - Jonathan Indajang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - David Pitt
- Department of Neurology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Chih Hung Lo
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, 13244, USA.
- Department of Biology, Syracuse University, Syracuse, NY, 13244, USA.
| |
Collapse
|
12
|
Naganishi S, Hagihara H, Miyakawa T. Gene Expression Signatures of Immaturity, Decreased pH, and Neural Hyperexcitation in the Hippocampus of Alzheimer's Disease Model Mice. Neuropsychopharmacol Rep 2025; 45:e70001. [PMID: 39907034 PMCID: PMC11795175 DOI: 10.1002/npr2.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/16/2024] [Accepted: 01/07/2025] [Indexed: 02/06/2025] Open
Abstract
AIMS Alzheimer's disease (AD) is a leading cause of dementia, with increasing prevalence. Mutations in genes like MAPT, PSEN1, and PSEN2 are risk factors, leading to the development of several AD model mice. Recent hypotheses suggest AD brain pathology involves abnormal neurodevelopment, decreased pH, and neural hyperexcitation. However, it remains unclear to what extent these pathologies are reflected in the gene expression changes of AD models. This study aims to compare gene expression patterns in the brains of multiple AD model mice with those related to these three factors, evaluating the extent of overlap. METHODS We conducted a comprehensive search of public databases, collecting 20 gene expression datasets from the hippocampus of AD model mice. These datasets were compared with gene sets related to hippocampal maturation, brain pH, and neural hyperexcitation to statistically assess overlap. Pathway enrichment analysis explored the biological relevance of these gene expression changes. RESULTS The extent of overlap with maturity-, pH-, and hyperexcitation-associated genes varied across AD models, showing significant correlations between lower maturity, lower pH, and increased neural hyperexcitation. In MAPT mutant and APP+PSEN1 homozygous transgenic mice, these signatures became more pronounced with age. Pathway meta-analysis revealed that genes associated with maturity, pH, and hyperexcitation in AD models are involved in synaptic and channel functions, as well as inflammatory responses, consistent with previous studies. CONCLUSION These findings suggest that pathophysiological changes related to maturity, pH, and neural hyperexcitation play varying roles across individual AD model mice. Our recent study found a negative correlation between disease progression and actual pH levels in human AD patients. Considering the results presented in this study, maturity and neural hyperexcitation, which are correlated with pH, may also be linked to disease progression. Thus, gene expression changes in these factors could be useful markers for assessing the pathology in AD models.
Collapse
Affiliation(s)
- Sayaka Naganishi
- Department of Systems Medical ScienceFujita Health University Graduate School of MedicineToyoakeAichiJapan
- Division of Systems Medical Science, Center for Medical ScienceFujita Health UniversityToyoakeAichiJapan
| | - Hideo Hagihara
- Division of Systems Medical Science, Center for Medical ScienceFujita Health UniversityToyoakeAichiJapan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Center for Medical ScienceFujita Health UniversityToyoakeAichiJapan
| |
Collapse
|
13
|
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. RESEARCH SQUARE 2025:rs.3.rs-6038641. [PMID: 40060041 PMCID: PMC11888548 DOI: 10.21203/rs.3.rs-6038641/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/21/2025]
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 hydrolasis by the soluble epoxide hydrolase (sEH). Accordingly, inhibition of sEH has been shown to stabilize the EETs and dampen neuroinflammation in Aβ mouse models of Alzheimer's disease (AD). However, the role of the sEH-EET signaling pathway in other cell types of the CNS and in other neurodegenerative conditions are less understood. Methods Here we examined the mechanisms and the functional role of the sEH-EET axis in tauopathy by treating the PS19 mice with a small molecule sEH inhibitor TPPU and by crossing the PS19 mice with Ephx2 (gene encoding sEH) knockout mice, followed by single-nucleus RNA-sequencing (snRNA-seq), biochemical and immunohistochemical characterization, and behavioral analysis. We also tested the effect of the sEH-EET pathway in primary microglia cultures and human induced pluripotent stem cell (iPSC)-derived neurons that develop seeding-induced Tau inclusions. Results We show that sEH inhibition improved cognitive function, rescued neuronal cell loss, and reduced Tau pathology and microglia reactivity. snRNA-seq revealed that TPPU treatment resulted in the upregulation of actin cytoskeleton and excitatory synaptic pathway genes. Treating the human iPSC-derived neurons with TPPU led to enhanced synaptic density without affecting Tau accumulation, indicating a cell-autonomous effect of sEH blockade in neuroprotection. Further, sEH inhibition reversed disease-associated and interferon-response microglia states in PS19 mice and EET supplementation enhanced Tau phagocytosis and clearance in primary microglia cultures. Conclusion These findings demonstrate that sEH blockade or EET augmentation confer therapeutic benefit against neurodegenerative tauopathies through parallel targeting of neuronal and microglial pathways.
Collapse
Affiliation(s)
- Shuo Wang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
| | - Chuangye Qi
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
| | | | - Baijayanti Ghosh
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
| | - Wen Xiong
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
| | - Baiping Wang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Yanyan Qi
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
| | - Sung Hee Hwang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA
| | - Bruce D. Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA
| | - Hongjie Li
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Li Gan
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine, New York, NY
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| |
Collapse
|
14
|
Wei Y, Zhang Y, Cao W, Cheng N, Xiao Y, Zhu Y, Xu Y, Zhang L, Guo L, Song J, Sha SH, Shao B, Ma F, Yang J, Ying Z, He Z, Chai R, Fang Q, Yang J. RONIN/HCF1-TFEB Axis Protects Against D-Galactose-Induced Cochlear Hair Cell Senescence Through Autophagy Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2407880. [PMID: 39985193 DOI: 10.1002/advs.202407880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 01/17/2025] [Indexed: 02/24/2025]
Abstract
Age-related hearing loss is characterized by senescent inner ear hair cells (HCs) and reduced autophagy. Despite the improved understanding of these processes, detailed molecular mechanisms underlying cochlear HC senescence remain unclear. Transcription Factor EB (TFEB), a key regulator of genes associated with autophagy and lysosomes, crucially affects aging-related illnesses. However, intricate regulatory networks that influence TFEB activity remain to be thoroughly elucidated. The findings revealed that RONIN (THAP11), through its interaction with host cell factor C1 (HCF1/HCFC1), modulated the transcriptional activity of Tfeb, thus contributing to the mitigation (D-galatactose [D-gal]) senescent HC loss. Specifically, RONIN overexpression improved autophagy levels and lysosomal activity and attenuated changes associated with the senescence of HCs triggered by D-gal. These findings highlight the possibility of using RONIN as a viable therapeutic target to ameliorate presbycusis by enhancing the TFEB function.
Collapse
Affiliation(s)
- Yongjie Wei
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yuhua Zhang
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Wei Cao
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Nan Cheng
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yun Xiao
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yongjun Zhu
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yan Xu
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Lei Zhang
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Lingna Guo
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jun Song
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Su-Hua Sha
- Department of Pathology and Laboratory Medicine, The Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Buwei Shao
- School of Medicine, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Fang Ma
- Center for Scientific Research of Anhui Medical University, Hefei, 230032, China
| | - Jingwen Yang
- International Department of Hefei 168 High School, Hefei, 230601, China
| | - Zheng Ying
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Zuhong He
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Co-Innovation Center of Neuroregeneration Nantong University, Nantong, 226001, China
- Department of Neurology, Aerospace Center Hospital, School of Life Science Beijing Institute of Technology, Beijing, 100081, China
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Southeast University Shenzhen Research Institute, Shenzhen, 518063, China
| | - Qiaojun Fang
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jianming Yang
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| |
Collapse
|
15
|
Hao M, Chu J, Zhang T, Yin T, Gu Y, Liang W, Ji W, Zhuang J, Liu Y, Gao J, Yin Y. Nanomaterials-mediated lysosomal regulation: a robust protein-clearance approach for the treatment of Alzheimer's disease. Neural Regen Res 2025; 20:424-439. [PMID: 38819046 PMCID: PMC11317947 DOI: 10.4103/nrr.nrr-d-23-01736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/04/2024] [Accepted: 02/20/2024] [Indexed: 06/01/2024] Open
Abstract
Alzheimer's disease is a debilitating, progressive neurodegenerative disorder characterized by the progressive accumulation of abnormal proteins, including amyloid plaques and intracellular tau tangles, primarily within the brain. Lysosomes, crucial intracellular organelles responsible for protein degradation, play a key role in maintaining cellular homeostasis. Some studies have suggested a link between the dysregulation of the lysosomal system and pathogenesis of neurodegenerative diseases, including Alzheimer's disease. Restoring the normal physiological function of lysosomes hold the potential to reduce the pathological burden and improve the symptoms of Alzheimer's disease. Currently, the efficacy of drugs in treating Alzheimer's disease is limited, with major challenges in drug delivery efficiency and targeting. Recently, nanomaterials have gained widespread use in Alzheimer's disease drug research owing to their favorable physical and chemical properties. This review aims to provide a comprehensive overview of recent advances in using nanomaterials (polymeric nanomaterials, nanoemulsions, and carbon-based nanomaterials) to enhance lysosomal function in treating Alzheimer's disease. This review also explores new concepts and potential therapeutic strategies for Alzheimer's disease through the integration of nanomaterials and modulation of lysosomal function. In conclusion, this review emphasizes the potential of nanomaterials in modulating lysosomal function to improve the pathological features of Alzheimer's disease. The application of nanotechnology to the development of Alzheimer's disease drugs brings new ideas and approaches for future treatment of this disease.
Collapse
Affiliation(s)
- Mengqi Hao
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Jianjian Chu
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Tong Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Yuankai Gu
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Wendanqi Liang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Wenbo Ji
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Jianhua Zhuang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - You Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
16
|
Renganathan A, Minaya MA, Broder M, Alfradique-Dunham I, Moritz M, Bhagat R, Marsh J, Verbeck A, Galasso G, Starr E, Agard DA, Cruchaga C, Karch CM. A novel lncRNA FAM151B-DT regulates autophagy and degradation of aggregation prone proteins. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.22.25320997. [PMID: 39974060 PMCID: PMC11838976 DOI: 10.1101/2025.01.22.25320997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Neurodegenerative diseases share common features of protein aggregation along with other pleiotropic traits, including shifts in transcriptional patterns, neuroinflammation, disruptions in synaptic signaling, mitochondrial dysfunction, oxidative stress, and impaired clearance mechanisms like autophagy. However, key regulators of these pleotropic traits have yet to be identified. Here, we discovered a novel long non-coding RNA (lncRNA), FAM151B-DT, that is reduced in a stem cell model of frontotemporal dementia with tau inclusions (FTLD-tau) and in brains from FTLD-tau, progressive supranuclear palsy, Alzheimer's disease, and Parkinson's disease patients. We show that silencing FAM151B-DT in vitro is sufficient to enhance tau aggregation. To begin to understand the mechanism by which FAM151B-DT mediates tau aggregation and contributes to several neurodegenerative diseases, we deeply characterized this novel lncRNA and found that FAM151B-DT resides in the cytoplasm where it interacts with tau, α-synuclein, HSC70, and other proteins enriched in protein homeostasis. When silenced, FAM151B-DT blocks autophagy, leading to the accumulation of tau and α-synuclein. Importantly, we discovered that increasing FAM151B-DT expression is sufficient to promote autophagic flux, reduce phospho-tau and α-synuclein, and reduce tau aggregation. Overall, these findings pave the way for further exploration of FAM151B-DT as a promising molecular target for several neurodegenerative diseases.
Collapse
Affiliation(s)
- Arun Renganathan
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | - Miguel A. Minaya
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | - Matthew Broder
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | | | - Michelle Moritz
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Reshma Bhagat
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | - Jacob Marsh
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | - Anthony Verbeck
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | - Grant Galasso
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | - Emma Starr
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
| | - David A. Agard
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
- Chan Zuckerberg Imaging Institute, Redwood City, CA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
- Knight Alzheimer Disease Research Center, Washington University in St Louis, St Louis, MO
| | - Celeste M. Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, MO
- Knight Alzheimer Disease Research Center, Washington University in St Louis, St Louis, MO
| |
Collapse
|
17
|
Jia N, Ganesan D, Guan H, Jeong YY, Han S, Rajapaksha G, Nissenbaum M, Kusnecov AW, Cai Q. Mitochondrial bioenergetics stimulates autophagy for pathological MAPT/Tau clearance in tauopathy neurons. Autophagy 2025; 21:54-79. [PMID: 39171695 DOI: 10.1080/15548627.2024.2392408] [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: 01/26/2024] [Revised: 08/04/2024] [Accepted: 08/09/2024] [Indexed: 08/23/2024] Open
Abstract
Hyperphosphorylation and aggregation of MAPT (microtubule-associated protein tau) is a pathogenic hallmark of tauopathies and a defining feature of Alzheimer disease (AD). Pathological MAPT/tau is targeted by macroautophagy/autophagy for clearance after being sequestered within autophagosomes, but autophagy dysfunction is indicated in tauopathy. While mitochondrial bioenergetic deficits have been shown to precede MAPT/tau pathology in tauopathy brains, it is unclear whether energy metabolism deficiency is involved in the pathogenesis of autophagy defects. Here, we reveal that stimulation of anaplerotic metabolism restores defective oxidative phosphorylation (OXPHOS) in tauopathy neurons which, strikingly, leads to pronounced MAPT/tau clearance by boosting autophagy functionality through enhancements of mitochondrial biosynthesis and supply of phosphatidylethanolamine for autophagosome biogenesis. Furthermore, early anaplerotic stimulation of OXPHOS elevates autophagy activity and attenuates MAPT/tau pathology, thereby counteracting memory impairment in tauopathy mice. Taken together, our study sheds light on a pivotal role of mitochondrial bioenergetic deficiency in tauopathy-related autophagy defects and suggests a new therapeutic strategy to prevent the buildup of pathological MAPT/tau in AD and other tauopathy diseases.Abbreviation: AA: antimycin A; AD, Alzheimer disease; ATP, adenosine triphosphate; AV, autophagosome/autophagic vacuole; AZ, active zone; Baf-A1: bafilomycin A1; CHX, cycloheximide; COX, cytochrome c oxidase; DIV, days in vitro; DRG, dorsal root ganglion; ETN, ethanolamine; FRET, Förster/fluorescence resonance energy transfer; FTD, frontotemporal dementia; Gln, glutamine; HA: hydroxylamine; HsMAPT/Tau, human MAPT; IMM, inner mitochondrial membrane; LAMP1, lysosomal-associated membrane protein 1; LIs, lysosomal inhibitors; MDAV, mitochondria-derived autophagic vacuole; MmMAPT/Tau, murine MAPT; NFT, neurofibrillary tangle; OCR, oxygen consumption rate; Omy: oligomycin; OXPHOS, oxidative phosphorylation; PPARGC1A/PGC-1alpha: peroxisome proliferative activated receptor, gamma, coactivator 1 alpha; PE, phosphatidylethanolamine; phospho-MAPT/tau, hyperphosphorylated MAPT; PS, phosphatidylserine; PISD, phosphatidylserine decarboxylase;SQSTM1/p62, sequestosome 1; STX1, syntaxin 1; SYP, synaptophysin; Tg, transgenic; TCA, tricarboxylic acid; TEM, transmission electron microscopy.
Collapse
Affiliation(s)
- Nuo Jia
- Department of Cell Biology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Dhasarathan Ganesan
- Department of Cell Biology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Hongyuan Guan
- Department of Cell Biology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Yu Young Jeong
- Department of Cell Biology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Sinsuk Han
- Department of Cell Biology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Gavesh Rajapaksha
- Department of Cell Biology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Marialaina Nissenbaum
- Department of Psychology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Alexander W Kusnecov
- Department of Psychology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Qian Cai
- Department of Cell Biology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
18
|
Kloske CM, Mahinrad S, Barnum CJ, Batista AF, Bradshaw EM, Butts B, Carrillo MC, Chakrabarty P, Chen X, Craft S, Da Mesquita S, Dabin LC, Devanand D, Duran‐Laforet V, Elyaman W, Evans EE, Fitzgerald‐Bocarsly P, Foley KE, Harms AS, Heneka MT, Hong S, Huang YA, Jackvony S, Lai L, Guen YL, Lemere CA, Liddelow SA, Martin‐Peña A, Orr AG, Quintana FJ, Ramey GD, Rexach JE, Rizzo SJS, Sexton C, Tang AS, Torrellas JG, Tsai AP, van Olst L, Walker KA, Wharton W, Tansey MG, Wilcock DM. Advancements in Immunity and Dementia Research: Highlights from the 2023 AAIC Advancements: Immunity Conference. Alzheimers Dement 2025; 21:e14291. [PMID: 39692624 PMCID: PMC11772715 DOI: 10.1002/alz.14291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/23/2024] [Accepted: 09/07/2024] [Indexed: 12/19/2024]
Abstract
The immune system is a key player in the onset and progression of neurodegenerative disorders. While brain resident immune cell-mediated neuroinflammation and peripheral immune cell (eg, T cell) infiltration into the brain have been shown to significantly contribute to Alzheimer's disease (AD) pathology, the nature and extent of immune responses in the brain in the context of AD and related dementias (ADRD) remain unclear. Furthermore, the roles of the peripheral immune system in driving ADRD pathology remain incompletely elucidated. In March of 2023, the Alzheimer's Association convened the Alzheimer's Association International Conference (AAIC), Advancements: Immunity, to discuss the roles of the immune system in ADRD. A wide range of topics were discussed, such as animal models that replicate human pathology, immune-related biomarkers and clinical trials, and lessons from other fields describing immune responses in neurodegeneration. This manuscript presents highlights from the conference and outlines avenues for future research on the roles of immunity in neurodegenerative disorders. HIGHLIGHTS: The immune system plays a central role in the pathogenesis of Alzheimer's disease. The immune system exerts numerous effects throughout the brain on amyloid-beta, tau, and other pathways. The 2023 AAIC, Advancements: Immunity, encouraged discussions and collaborations on understanding the role of the immune system.
Collapse
|
19
|
Lin Y, Cheng L, Chen Y, Li W, Guo Q, Miao Y. TFEB signaling promotes autophagic degradation of NLRP3 to attenuate neuroinflammation in diabetic encephalopathy. Am J Physiol Cell Physiol 2024; 327:C1481-C1496. [PMID: 39437446 DOI: 10.1152/ajpcell.00322.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/18/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024]
Abstract
Diabetic encephalopathy (DE), a neurological complication of diabetes mellitus, has an unclear etiology. Shreds of evidence show that the nucleotide-binding oligomerization domain-like receptor family protein 3 (NLRP3) inflammasome-induced neuroinflammation and transcription factor EB (TFEB)-mediated autophagy impairment may take part in DE development. The cross talk between these two pathways and their contribution to DE remains to be explored. A mouse model of type 2 diabetes mellitus (T2DM) exhibiting cognitive dysfunction was created, along with high-glucose (HG) cultured BV2 cells. Following, 3-methyladenine (3-MA) and rapamycin were used to modulate autophagy. To evaluate the potential therapeutic benefits of TFEB in DE, we overexpressed and knocked down TFEB in both mice and cells. Autophagy impairment and NLRP3 inflammasome activation were noticed in T2DM mice and HG-cultured BV2 cells. The inflammatory response caused by NLRP3 inflammasome activation was decreased by rapamycin-induced autophagy enhancement, while 3-MA treatment further deteriorated it. Nuclear translocation and expression of TFEB were hampered in HG-cultured BV2 cells and T2DM mice. Exogenous TFEB overexpression boosted NLRP3 degradation via autophagy, which in turn alleviated microglial activation as well as ameliorated cognitive deficits and neuronal damage. In addition, TFEB knockdown exacerbated neuroinflammation by decreasing autophagy-mediated NLRP3 degradation. Our findings have unraveled the pathogenesis of a previously underappreciated disease, implying that the activation of NLRP3 inflammasome and impairment of autophagy in microglia are significant etiological factors in the DE. The TFEB-mediated autophagy pathway can reduce neuroinflammation by enhancing NLRP3 degradation. This could potentially serve as a viable and innovative treatment approach for DE.NEW & NOTEWORTHY This article delves into the intricate connections between inflammation, autophagy, diabetes, and neurodegeneration, with a particular focus on a disease that is not yet fully understood-diabetic encephalopathy (DE). TFEB emerges as a pivotal regulator in balancing autophagy and inflammation in DE. Our findings highlight the crucial function of the TFEB-mediated autophagy pathway in mitigating inflammatory damage in DE, suggesting a new treatment strategy.
Collapse
Affiliation(s)
- Yijia Lin
- Department of Geriatrics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lizhen Cheng
- Department of Geriatrics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yixin Chen
- Department of Geriatrics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Li
- Department of Geriatrics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qihao Guo
- Department of Geriatrics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ya Miao
- Department of Geriatrics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Chen H, Hinz K, Zhang C, Rodriguez Y, Williams SN, Niu M, Ma X, Chao X, Frazier AL, McCarson KE, Wang X, Peng Z, Liu W, Ni HM, Zhang J, Swerdlow RH, Ding WX. Late-Life Alcohol Exposure Does Not Exacerbate Age-Dependent Reductions in Mouse Spatial Memory and Brain TFEB Activity. Biomolecules 2024; 14:1537. [PMID: 39766244 PMCID: PMC11673978 DOI: 10.3390/biom14121537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Alcohol consumption is believed to affect Alzheimer's disease (AD) risk, but the contributing mechanisms are not well understood. A potential mediator of the proposed alcohol-AD connection is autophagy, a degradation pathway that maintains organelle and protein homeostasis. Autophagy is regulated through the activity of Transcription factor EB (TFEB), which promotes lysosome and autophagy-related gene expression. The purpose of this study is to explore whether chronic alcohol consumption worsens the age-related decline in TFEB-mediated lysosomal biogenesis in the brain and exacerbates cognitive decline associated with aging. To explore the effect of alcohol on brain TFEB and autophagy, we exposed young (3-month-old) and aged (23-month-old) mice to two alcohol-feeding paradigms and assessed biochemical, transcriptome, histology, and behavioral endpoints. In young mice, alcohol decreased hippocampal nuclear TFEB staining but increased SQSTM1/p62, LC3-II, ubiquitinated proteins, and phosphorylated Tau. Hippocampal TFEB activity was lower in aged mice than it was in young mice, and Gao-binge alcohol feeding did not worsen the age-related reduction in TFEB activity. Morris Water and Barnes Maze spatial memory tasks were used to characterize the effects of aging and chronic alcohol exposure (mice fed alcohol for 4 weeks). The aged mice showed worse spatial memory acquisition in both tests. Alcohol feeding slightly impaired spatial memory in the young mice, but had little effect or even slightly improved spatial memory acquisition in the aged mice. In conclusion, aging produces greater reductions in brain autophagy flux and impairment of spatial memory than alcohol consumption.
Collapse
Affiliation(s)
- Hao Chen
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Kaitlyn Hinz
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Chen Zhang
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Yssa Rodriguez
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Sha Neisha Williams
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Mengwei Niu
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Xiaowen Ma
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Alexandria L. Frazier
- R.L. Smith IDDRC Rodent Behavior Facility, Disease Model and Assessment Services, The University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Kenneth E. McCarson
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
- R.L. Smith IDDRC Rodent Behavior Facility, Disease Model and Assessment Services, The University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Xiaowan Wang
- Department of Neurology, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (X.W.); (R.H.S.)
| | - Zheyun Peng
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Detroit, MI 48201, USA; (Z.P.); (W.L.)
| | - Wanqing Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Detroit, MI 48201, USA; (Z.P.); (W.L.)
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Jianhua Zhang
- Department of Pathology, Division of Molecular Cellular Pathology, University of Alabama at Birmingham, 901 19th Street South, Birmingham, AL 35294, USA;
| | - Russell H. Swerdlow
- Department of Neurology, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (X.W.); (R.H.S.)
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
- Department of Internal Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
21
|
Song C, Huang W, Zhang P, Shi J, Yu T, Wang J, Hu Y, Zhao L, Zhang R, Wang G, Zhang Y, Chen H, Wang H. Critical role of ROCK1 in AD pathogenesis via controlling lysosomal biogenesis and acidification. Transl Neurodegener 2024; 13:54. [PMID: 39497162 PMCID: PMC11533276 DOI: 10.1186/s40035-024-00442-9] [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: 04/01/2024] [Accepted: 09/11/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Lysosomal homeostasis and functions are essential for the survival of neural cells. Impaired lysosomal biogenesis and acidification in Alzheimer's disease (AD) pathogenesis leads to proteolytic dysfunction and neurodegeneration. However, the key regulatory factors and mechanisms of lysosomal homeostasis in AD remain poorly understood. METHODS ROCK1 expression and its co-localization with LAMP1 and SQSTM1/p62 were detected in post-mortem brains of healthy controls and AD patients. Lysosome-related fluorescence probe staining, transmission electron microscopy and immunoblotting were performed to evaluate the role of ROCK1 in lysosomal biogenesis and acidification in various neural cell types. The interaction between ROCK1 and TFEB was confirmed by surface plasmon resonance and in situ proximity ligation assay (PLA). Moreover, we performed AAV-mediated ROCK1 downregulation followed by immunofluorescence, enzyme-linked immunosorbent assay (ELISA) and behavioral tests to unveil the effects of the ROCK1-TFEB axis on lysosomes in APP/PS1 transgenic mice. RESULTS ROCK1 level was significantly increased in the brains of AD individuals, and was positively correlated with lysosomal markers and Aβ. Lysosomal proteolysis was largely impaired by the high abundance of ROCK1, while ROCK1 knockdown mitigated the lysosomal dysfunction in neurons and microglia. Moreover, we verified ROCK1 as a previously unknown upstream kinase of TFEB independent of m-TOR or GSK-3β. ROCK1 elevation resulted in abundant extracellular Aβ deposition which in turn bound to Aβ receptors and activated RhoA/ROCK1, thus forming a vicious circle of AD pathogenesis. Genetically downregulating ROCK1 lowered its interference with TFEB, promoted TFEB nuclear distribution, lysosomal biogenesis and lysosome-mediated Aβ clearance, and eventually prevented pathological traits and cognitive deficits in APP/PS1 mice. CONCLUSION In summary, our results provide a mechanistic insight into the critical role of ROCK1 in lysosomal regulation and Aβ clearance in AD by acting as a novel upstream serine kinase of TFEB.
Collapse
Affiliation(s)
- Chenghuan Song
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wanying Huang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Pingao Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiyun Shi
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ting Yu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jing Wang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yongbo Hu
- Department of Neurology, Chang-Hai Hospital, The Second Military Medical University, Shanghai, 200433, China
| | - Lanxue Zhao
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Rui Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Gang Wang
- Department of Neurology and Neuroscience Institute, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yongfang Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hongzhuan Chen
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shuguang Lab of Future Health, Shanghai Frontiers Science Center of TCM Chemical Biology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Hao Wang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
22
|
Kam MK, Park JY, Yun GH, Sohn HY, Park JH, Choi J, Koh YH, Jo C. Rottlerin Enhances the Autophagic Degradation of Phosphorylated Tau in Neuronal Cells. Mol Neurobiol 2024; 61:9633-9645. [PMID: 38671330 DOI: 10.1007/s12035-024-04182-9] [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: 11/29/2023] [Accepted: 04/15/2024] [Indexed: 04/28/2024]
Abstract
Intra-neuronal accumulation of hyper-phosphorylated tau as neurofibrillary tangles (NFT) is a hallmark of Alzheimer's disease (AD). To prevent the aggregation of phosphorylated tau in neurons, decreasing the phosphorylated tau protein levels is important. Here, we examined the biological effects of rottlerin, a phytochemical compound extracted from the Kamala tree, Mallotus philippinensis, on phosphorylated tau levels. Notably, rottlerin decreased the levels of intracellular phosphorylated and total tau. A marked increase in the LC3-II, a hallmark of autophagy, was observed in these cells, indicating that rottlerin strongly induced autophagy. Interestingly, rottlerin induced the phosphorylation of Raptor at S792 through the activation of adenosine-monophosphate activated-protein kinase (AMPK), which likely inhibits the mammalian target of rapamycin complex 1 (mTORC1), thus resulting in the activation of transcription factor EB (TFEB), a master regulator of autophagy. In addition, nuclear factor erythroid 2-related factor 2 (Nrf2) activity increased in the presence of rottlerin. The decrease of phosphorylated tau levels in the presence of rottlerin was ameliorated by the knockdown of TFEB and partially attenuated by the knockout of the Nrf2 gene. Taken together, rottlerin likely enhances the degradation of phosphorylated tau through autophagy activated by TFEB and Nrf2. Thus, our results suggest that a natural compound rottlerin could be used as a preventive and therapeutic drug for AD.
Collapse
Affiliation(s)
- Min Kyoung Kam
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea
| | - Jee-Yun Park
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea
| | - Gwang Ho Yun
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea
| | - Hee-Young Sohn
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea
| | - Jung Hyun Park
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea
| | - Jiyoung Choi
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea
| | - Young Ho Koh
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea
| | - Chulman Jo
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-Ro, Osong-Eup, Cheongju-Si, 363-951, Chungcheongbuk-Do, Korea.
| |
Collapse
|
23
|
James A, Hendrixson J, Kadhim I, Marques-Carvalho A, Laster J, Crawford J, Thostenson J, Sato A, Almeida M, Onal M. CRISPR activation of Tfeb , a master regulator of autophagy and lysosomal biogenesis, in osteoblast lineage cells increases bone mass and strength. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615175. [PMID: 39386619 PMCID: PMC11463346 DOI: 10.1101/2024.09.26.615175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Autophagy is a recycling pathway in which damaged or dysfunctional proteins, protein aggregates, and organelles are delivered to lysosomes for degradation. Insufficiency of autophagy is thought to contribute to several age-related diseases including osteoporosis. Consistent with this, elimination of autophagy from the osteoblast lineage reduces bone formation and causes low bone mass. However, whether increasing autophagy would benefit bone health is unknown. Here, we increased expression of the endogenous Transcription Factor EB gene ( Tfeb ) in osteoblast lineage cells in vivo via CRISPR activation. Tfeb overexpression stimulated autophagy and lysosomal biogenesis in osteoblasts. Tfeb overexpressing male mice displayed a robust increase in femoral and vertebral cortical thickness at 4.5 months of age. Histomorphometric analysis revealed that the increase in femoral cortical thickness was due to increased bone formation at the periosteal surface. Tfeb overexpression also increased femoral trabecular bone volume. Consistent with these results, bone strength was increased in Tfeb overexpressing mice. Female Tfeb overexpressing mice also displayed a progressive increase in bone mass over time and at 12 months of age had high cortical thickness and trabecular bone volume. This increase in vertebral trabecular bone volume was due to elevated bone formation. Osteoblastic cultures showed that Tfeb overexpression increased proliferation and osteoblast formation. Overall, these results demonstrate that stimulation of autophagy in osteoblast lineage cells promotes bone formation and strength and may represent an effective approach to combat osteoporosis.
Collapse
|
24
|
Lin X, Chen C, Chen J, Zhu C, Zhang J, Su R, Chen S, Weng S, Chang X, Lin S, Chen Y, Li J, Lin L, Zhou J, Guo Z, Yu G, Shao W, Hu H, Wu S, Zhang Q, Li H, Zheng F. Long Noncoding RNA NR_030777 Alleviates Cobalt Nanoparticles-Induced Neurodegenerative Damage by Promoting Autophagosome-Lysosome Fusion. ACS NANO 2024; 18:24872-24897. [PMID: 39197041 PMCID: PMC11394346 DOI: 10.1021/acsnano.4c05249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 08/30/2024]
Abstract
Potential exposure to cobalt nanoparticles (CoNPs) occurs in various fields, including hard alloy industrial production, the increasing use of new energy lithium-ion batteries, and millions of patients with metal-on-metal joint prostheses. Evidence from human, animal, and in vitro experiments suggests a close relationship between CoNPs and neurotoxicity. However, a systematic assessment of central nervous system (CNS) impairment due to CoNPs exposure and the underlying molecular mechanisms is lacking. In this study, we found that CoNPs induced neurodegenerative damage both in vivo and in vitro, including cognitive impairment, β-amyloid deposition and Tau hyperphosphorylation. CoNPs promoted the formation of autophagosomes and impeding autophagosomal-lysosomal fusion in vivo and in vitro, leading to toxic protein accumulation. Moreover, CoNPs exposure reduced the level of transcription factor EB (TFEB) and the abundance of lysosome, causing a blockage in autophagosomal-lysosomal fusion. Interestingly, overexpression of long noncoding RNA NR_030777 mitigated CoNPs-induced neurodegenerative damage in both in vivo and in vitro models. Fluorescence in situ hybridization assay revealed that NR_030777 directly binds and stabilizes TFEB mRNA, alleviating the blockage of autophagosomal-lysosomal fusion and ultimately restoring neurodegeneration induced by CoNPs in vivo and in vitro. In summary, our study demonstrates that autophagic dysfunction is the main toxic mechanism of neurodegeneration upon CoNPs exposure and NR_030777 plays a crucial role in CoNPs-induced autophagic dysfunction. Additionally, the proposed adverse outcome pathway contributes to a better understanding of CNS toxicity assessment of CoNPs.
Collapse
Affiliation(s)
- Xinpei Lin
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Cheng Chen
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Jinxiang Chen
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Canlin Zhu
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Jiajun Zhang
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Ruiqi Su
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Shujia Chen
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Shucan Weng
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Xiangyu Chang
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Shengsong Lin
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Yilong Chen
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Jiamei Li
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Ling Lin
- Public
Technology Service Center, Fujian Medical
University, Fuzhou, Fujian Province 350122, China
| | - Jinfu Zhou
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- Medical
Genetic Diagnosis and Therapy Center, Fujian Maternity and Child Health
Hospital College of Clinical Medicine for Obstetrics & Gynecology
and Pediatrics, Fujian Medical University, Fuzhou, Fujian Province 350001, China
| | - Zhenkun Guo
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Guangxia Yu
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Wenya Shao
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Hong Hu
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Siying Wu
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- Department
of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Qunwei Zhang
- Department
of Epidemiology and Population Health, School of Public Health and
Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, Kentucky 40292, United States
| | - Huangyuan Li
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Fuli Zheng
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| |
Collapse
|
25
|
Babu S, Nicholson KA, Rothstein JD, Swenson A, Sampognaro PJ, Pant P, Macklin EA, Spruill S, Paganoni S, Gendron TF, Prudencio M, Petrucelli L, Nix D, Landrette S, Nkrumah E, Fandrick K, Edwards J, Young PR. Apilimod dimesylate in C9orf72 amyotrophic lateral sclerosis: a randomized phase 2a clinical trial. Brain 2024; 147:2998-3008. [PMID: 38606777 DOI: 10.1093/brain/awae109] [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: 11/28/2023] [Revised: 02/18/2024] [Accepted: 03/03/2024] [Indexed: 04/13/2024] Open
Abstract
Apilimod dimesylate is a first-in-class phosphoinositide kinase, FYVE-type zinc finger-containing (PIKfyve) inhibitor with a favourable clinical safety profile and has demonstrated activity in preclinical C9orf72 and TDP-43 amyotrophic lateral sclerosis (ALS) models. In this ALS clinical trial, the safety, tolerability, CNS penetrance and modulation of pharmacodynamic target engagement biomarkers were evaluated. This phase 2a, randomized, double-blind, placebo-controlled, biomarker-end-point clinical trial was conducted in four US centres (ClinicalTrials.gov NCT05163886). Participants with C9orf72 repeat expansions were randomly assigned (2:1) to receive twice-daily oral treatment with 125 mg apilimod dimesylate capsules or matching placebo for 12 weeks, followed by a 12-week open-label extension. Safety was measured as the occurrence of treatment-emergent or serious adverse events attributable to the study drug and tolerability at trial completion or treatment over 12 weeks. Changes from baseline in plasma and CSF and concentrations of apilimod dimesylate and its active metabolites and of pharmacodynamic biomarkers of PIKfyve inhibition [soluble glycoprotein nonmetastatic melanoma protein B (sGPNMB) upregulation] and disease-specific CNS target engagement [poly(GP)] were measured. Between 16 December 2021 and 7 July 2022, 15 eligible participants were enrolled. There were no drug-related serious adverse events reported in the trial. Fourteen (93%) participants completed the double-blind period with 99% dose compliance [n = 9 (90%) apilimod dimesylate; n = 5 (100%) placebo]. At Week 12, apilimod dimesylate was measurable in CSF at 1.63 ng/ml [standard deviation (SD): 0.937]. At Week 12, apilimod dimesylate increased plasma sGPNMB by >2.5-fold (P < 0.001), indicating PIKfyve inhibition, and lowered CSF poly(GP) protein levels by 73% (P < 0.001), indicating CNS tissue-level proof of mechanism. Apilimod dimesylate met prespecified key safety and biomarker end-points in this phase 2a trial and demonstrated CNS penetrance and pharmacodynamic target engagement. Apilimod dimesylate was observed to result in the greatest reduction in CSF poly(GP) levels observed to date in C9orf72 clinical trials.
Collapse
Affiliation(s)
- Suma Babu
- Sean M Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Katharine A Nicholson
- Sean M Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Sanofi, Cambridge, MA 02139, USA
| | - Jeffrey D Rothstein
- Department of Neurology, Brain Science Institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Andrea Swenson
- Department of Neurology, University of Iowa, Iowa city, IA 52242, USA
| | - Paul J Sampognaro
- Neuromuscular Division, Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Pravin Pant
- Sean M Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Eric A Macklin
- Biostatistics Center at Massachusetts General Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Susan Spruill
- Applied Statistics and Consulting, Spruce Pine, NC 28777, USA
| | - Sabrina Paganoni
- Sean M Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Liu B, Cui D, Liu J, Shi JS. Transcriptome analysis of the aged SAMP8 mouse model of Alzheimer's disease reveals novel molecular targets of formononetin protection. Front Pharmacol 2024; 15:1440515. [PMID: 39234102 PMCID: PMC11371586 DOI: 10.3389/fphar.2024.1440515] [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: 05/29/2024] [Accepted: 07/26/2024] [Indexed: 09/06/2024] Open
Abstract
Background Senescence-accelerated mouse prone 8 (SAMP8) and age-matched SAMR1 mice are used to study the pathogenesis and therapeutics of Alzheimer's disease (AD); however, the molecular mechanisms are not completely understood. Objective This study aimed to examine the effects of the 5-month administration of formononetin in SAMP8 mice and used RNA-seq to explore the molecular targets. Methods SAMP8 mice were orally administered formononetin (0, 8, and 16 mg/kg) from 4 months of age, and age-matched SAMR1 mice were used as controls. Behavioral tests were performed in 9-month-old mice, followed by histopathologic analysis. Total RNA from the hippocampus was isolated and subjected to RNA-seq, RT-qPCR, and bioinformatics analysis. Results The 9-month-old SAMP8 mice exhibited cognition deficits, evidenced by novel object recognition, open-field test, elevated plus maze, and passive avoidance. Nissl bodies in the cortex and hippocampus were decreased. Formononetin treatments ameliorated behavioral deficits and improved morphological changes, which were evidenced by Nissl and H&E staining. RNA-seq revealed distinct gene expression patterns between SAMP8 and SAMR1 mice. Differentially expressed genes in SAMP8 mice were attenuated or normalized by formononetin. Ingenuity pathway analysis (IPA) of canonical pathway and upstream regulators revealed increases in proinflammatory factors and immune dysfunction and decreases in NRF2 and SIRT-1 signaling pathways, leading to neuroinflammation. Formononetin treatment attenuated or reversed these molecular changes. The transcriptome of SAMP8 mice was correlated with transcriptomic profiles of other AD mouse models in the GEO database. Conclusion Neuroinflammation and decreased antioxidant and SIRT-1 signaling contributed to cognitive deficits in aged SAMP8 mice, which are potential therapeutic targets of formononetin in combination with other therapies.
Collapse
Affiliation(s)
- Bo Liu
- Key Lab for Basic Pharmacology and Joint International Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Di Cui
- Key Lab for Basic Pharmacology and Joint International Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Jie Liu
- Key Lab for Basic Pharmacology and Joint International Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Jing-Shan Shi
- Key Lab for Basic Pharmacology and Joint International Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
27
|
Brunialti E, Rizzi N, Pinto-Costa R, Villa A, Panzeri A, Meda C, Rebecchi M, Di Monte DA, Ciana P. Design and validation of a reporter mouse to study the dynamic regulation of TFEB and TFE3 activity through in vivo imaging techniques. Autophagy 2024; 20:1879-1894. [PMID: 38522425 PMCID: PMC11262230 DOI: 10.1080/15548627.2024.2334111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 03/18/2024] [Indexed: 03/26/2024] Open
Abstract
TFEB and TFE3 belong to the MiT/TFE family of transcription factors that bind identical DNA responsive elements in the regulatory regions of target genes. They are involved in regulating lysosomal biogenesis, function, exocytosis, autophagy, and lipid catabolism. Precise control of TFEB and TFE3 activity is crucial for processes such as senescence, stress response, energy metabolism, and cellular catabolism. Dysregulation of these factors is implicated in various diseases, thus researchers have explored pharmacological approaches to modulate MiT/TFE activity, considering these transcription factors as potential therapeutic targets. However, the physiological complexity of their functions and the lack of suitable in vivo tools have limited the development of selective MiT/TFE modulating agents. Here, we have created a reporter-based biosensor, named CLEARoptimized, facilitating the pharmacological profiling of TFEB- and TFE3-mediated transcription. This innovative tool enables the measurement of TFEB and TFE3 activity in living cells and mice through imaging and biochemical techniques. CLEARoptimized consists of a promoter with six coordinated lysosomal expression and regulation motifs identified through an in-depth bioinformatic analysis of the promoters of 128 TFEB-target genes. The biosensor drives the expression of luciferase and tdTomato reporter genes, allowing the quantification of TFEB and TFE3 activity in cells and in animals through optical imaging and biochemical assays. The biosensor's validity was confirmed by modulating MiT/TFE activity in both cell culture and reporter mice using physiological and pharmacological stimuli. Overall, this study introduces an innovative tool for studying autophagy and lysosomal pathway modulation at various biological levels, from individual cells to the entire organism.Abbreviations: CLEAR: coordinated lysosomal expression and regulation; MAR: matrix attachment regions; MiT: microphthalmia-associated transcription factor; ROI: region of interest; TBS: tris-buffered saline; TF: transcription factor; TFE3: transcription factor binding to IGHM enhancer 3; TFEB: transcription factor EB; TH: tyrosine hydroxylase; TK: thymidine kinase; TSS: transcription start site.
Collapse
Affiliation(s)
| | | | - Rita Pinto-Costa
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Alessandro Villa
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Alessia Panzeri
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Clara Meda
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Monica Rebecchi
- Department of Health Sciences, University of Milan, Milan, Italy
| | | | - Paolo Ciana
- Department of Health Sciences, University of Milan, Milan, Italy
| |
Collapse
|
28
|
Wang X, Wu W, Yang G, Yang XW, Ma X, Zhu DD, Ahmad K, Khan K, Wang YZ, Sui AR, Guo SY, Kong Y, Yuan B, Luo TY, Liu CK, Zhang P, Zhang Y, Li QF, Wang B, Wu Q, Wu XF, Xiao ZC, Ma QH, Li S. Cell-specific Nav1.6 knockdown reduced astrocyte-derived Aβ by reverse Na +-Ca 2+ transporter-mediated autophagy in alzheimer-like mice. J Adv Res 2024:S2090-1232(24)00309-6. [PMID: 39079584 DOI: 10.1016/j.jare.2024.07.024] [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: 03/27/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/16/2024] Open
Abstract
INTRODUCTION Nav1.6 is closely related to the pathology of Alzheimer's Disease (AD), and astrocytes have recently been identified as a significant source of β-amyloid (Aβ). However, little is known about the connection between Nav1.6 and astrocyte-derived Aβ. OBJECTIVE This study explored the crucial role of Nav1.6 in mediated astrocyte-derived Aβ in AD and knockdown astrocytic Nav1.6 alleviates AD progression by promoting autophagy and lysosome-APP fusion. METHODS A mouse model for astrocytic Nav1.6 knockdown was constructed to study the effects of astrocytic Nav1.6 on amyloidosis. The role of astrocytic Nav1.6 on autophagy and lysosome-APP(amyloid precursor protein) fusion was used by transmission electron microscope, immunostaining, western blot and patch clamp. Glial cell activation was detected using immunostaining. Neuroplasticity and neural network were assessed using patch-clamp, Golgi stain and EEG recording. Behavioral experiments were performed to evaluate cognitive defects. RESULTS Astrocytic Nav1.6 knockdown reduces amyloidosis, alleviates glial cell activation and morphological complexity, improves neuroplasticity and abnormal neural networks, as well as promotes learning and memory abilities in APP/PS1 mice. Astrocytic Nav1.6 knockdown reduces itself-derived Aβ by promoting lysosome- APP fusion, which is related to attenuating reverse Na+-Ca2+ exchange current thus reducing intracellular Ca2+ to facilitate autophagic through AKT/mTOR/ULK pathway. CONCLUSION Our findings unveil the crucial role of astrocyte-specific Nav1.6 in reducing astrocyte-derived Aβ, highlighting its potential as a cell-specific target for modulating AD progression.
Collapse
Affiliation(s)
- Xin Wang
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Wei Wu
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Guang Yang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue-Wei Yang
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Xu Ma
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Dan-Dan Zhu
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Kabir Ahmad
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Khizar Khan
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Ying-Zi Wang
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Ao-Ran Sui
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Song-Yu Guo
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Yue Kong
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Bo Yuan
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Tian-Yuan Luo
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Cheng-Kang Liu
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Peng Zhang
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Yue Zhang
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Qi-Fa Li
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Bin Wang
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Qiong Wu
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Xue-Fei Wu
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Zhi-Cheng Xiao
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - Quan-Hong Ma
- Institute of Neuroscience & Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou 215123, China; Department of Neurology and Clinical Research Center of Neurological Disease, the second affilitated Hospital of Soochow University, Suzhou, Jiangsu 215021, China.
| | - Shao Li
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China.
| |
Collapse
|
29
|
Zhao N, Chen X, Chen QG, Liu XT, Geng F, Zhu MM, Yan FL, Zhang ZJ, Ren QG. NLRP3-mediated autophagy dysfunction links gut microbiota dysbiosis to tau pathology in chronic sleep deprivation. Zool Res 2024; 45:857-874. [PMID: 39004863 PMCID: PMC11298670 DOI: 10.24272/j.issn.2095-8137.2024.085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/27/2024] [Indexed: 07/16/2024] Open
Abstract
Emerging evidence indicates that sleep deprivation (SD) can lead to Alzheimer's disease (AD)-related pathological changes and cognitive decline. However, the underlying mechanisms remain obscure. In the present study, we identified the existence of a microbiota-gut-brain axis in cognitive deficits resulting from chronic SD and revealed a potential pathway by which gut microbiota affects cognitive functioning in chronic SD. Our findings demonstrated that chronic SD in mice not only led to cognitive decline but also induced gut microbiota dysbiosis, elevated NLRP3 inflammasome expression, GSK-3β activation, autophagy dysfunction, and tau hyperphosphorylation in the hippocampus. Colonization with the "SD microbiota" replicated the pathological and behavioral abnormalities observed in chronic sleep-deprived mice. Remarkably, both the deletion of NLRP3 in NLRP3 -/- mice and specific knockdown of NLRP3 in the hippocampus restored autophagic flux, suppressed tau hyperphosphorylation, and ameliorated cognitive deficits induced by chronic SD, while GSK-3β activity was not regulated by the NLRP3 inflammasome in chronic SD. Notably, deletion of NLRP3 reversed NLRP3 inflammasome activation, autophagy deficits, and tau hyperphosphorylation induced by GSK-3β activation in primary hippocampal neurons, suggesting that GSK-3β, as a regulator of NLRP3-mediated autophagy dysfunction, plays a significant role in promoting tau hyperphosphorylation. Thus, gut microbiota dysbiosis was identified as a contributor to chronic SD-induced tau pathology via NLRP3-mediated autophagy dysfunction, ultimately leading to cognitive deficits. Overall, these findings highlight GSK-3β as a regulator of NLRP3-mediated autophagy dysfunction, playing a critical role in promoting tau hyperphosphorylation.
Collapse
Affiliation(s)
- Na Zhao
- Department of Neurology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Xiu Chen
- Department of Neurology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Qiu-Gu Chen
- Department of Neurology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Xue-Ting Liu
- Department of Neurology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Fan Geng
- Department of Neurology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Meng-Meng Zhu
- Department of Neurology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Fu-Ling Yan
- Department of Neurology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Zhi-Jun Zhang
- Department of Neurology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Qing-Guo Ren
- Department of Neurology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
- Department of Neurology, Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, China. E-mail:
| |
Collapse
|
30
|
Pollack SJ, Dakkak D, Guo T, Chennell G, Gomez-Suaga P, Noble W, Jimenez-Sanchez M, Hanger DP. Truncated tau interferes with the autophagy and endolysosomal pathway and results in lipid accumulation. Cell Mol Life Sci 2024; 81:304. [PMID: 39009859 PMCID: PMC11335226 DOI: 10.1007/s00018-024-05337-6] [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: 03/16/2024] [Revised: 06/11/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024]
Abstract
The autophagy-lysosomal pathway plays a critical role in the clearance of tau protein aggregates that deposit in the brain in tauopathies, and defects in this system are associated with disease pathogenesis. Here, we report that expression of Tau35, a tauopathy-associated carboxy-terminal fragment of tau, leads to lipid accumulation in cell lines and primary cortical neurons. Our findings suggest that this is likely due to a deleterious block of autophagic clearance and lysosomal degradative capacity by Tau35. Notably, upon induction of autophagy by Torin 1, Tau35 inhibited nuclear translocation of transcription factor EB (TFEB), a key regulator of lysosomal biogenesis. Both cell lines and primary cortical neurons expressing Tau35 also exhibited changes in endosomal protein expression. These findings implicate autophagic and endolysosomal dysfunction as key pathological mechanisms through which disease-associated tau fragments could lead to the development and progression of tauopathy.
Collapse
Affiliation(s)
- Saskia J Pollack
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London, SE5 9RX, UK
| | - Dina Dakkak
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London, SE5 9RX, UK
| | - Tong Guo
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London, SE5 9RX, UK
| | - George Chennell
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London, SE5 9RX, UK
| | - Patricia Gomez-Suaga
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London, SE5 9RX, UK
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas-Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Wendy Noble
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London, SE5 9RX, UK.
- Department of Clinical and Biomedical Sciences, Hatherly Laboratories, University of Exeter, Prince of Wales Road, Exeter, EX4 4PS, UK.
| | - Maria Jimenez-Sanchez
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London, SE5 9RX, UK.
| | - Diane P Hanger
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London, SE5 9RX, UK
| |
Collapse
|
31
|
Wang L, Cao J, Chen H, Ma Y, Zhang Y, Su X, Jing Y, Wang Y. Transcription Factor EB Overexpression through Glial Fibrillary Acidic Protein Promoter Disrupts Neuronal Lamination by Dysregulating Neurogenesis during Embryonic Development. Dev Neurosci 2024; 47:40-54. [PMID: 38583418 PMCID: PMC11709705 DOI: 10.1159/000538656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/26/2024] [Indexed: 04/09/2024] Open
Abstract
INTRODUCTION Transcription factor EB (TFEB), a key regulator of autophagy and lysosomal biogenesis, has diverse roles in various physiological processes. Enhancing lysosomal function by TFEB activation has recently been implicated in restoring neural stem cell (NSC) function. Overexpression of TFEB can inhibit the cell cycle of newborn cortical NSCs. It has also been found that TFEB regulates the pluripotency transcriptional network in mouse embryonic stem cells independent of autophagy and lysosomal biogenesis. This study aims to explore the effects of TFEB activation on neurogenesis in vivo through transgenic mice. METHODS We developed a glial fibrillary acidic protein (GFAP)-driven TFEB overexpression mouse model (TFEB GoE) by crossing the floxed TFEB overexpression mice and hGFAP-Cre mice. We performed immunohistochemical and fluorescence staining on brain tissue from newborn mice to assess neurogenesis changes, employing markers such as GFAP, Nestin, Ki67, doublecortin (DCX), Tbr1, and NeuN to trace different stages of neural development and cell proliferation. RESULTS TFEB GoE mice exhibited premature mortality, dying 10-20 days after birth. Immunohistochemical analysis revealed significant abnormalities, including disrupted hippocampal structure and cortical layering. Compared to control mice, TFEB GoE mice showed a marked increase in radial glial cells (RGCs) in the hippocampus and cortex, with Ki67 staining indicating these cells were predominantly in a quiescent state. This suggests that TFEB overexpression suppresses RGC proliferation. Additionally, abnormal distributions of migrating neurons and mature neurons were observed, highlighted by DCX, Tbr1, and NeuN staining, indicating a disruption in normal neurogenesis. CONCLUSION This study, using transgenic animals in vivo, revealed that GFAP-driven TFEB overexpression leads to abnormal neural layering in the hippocampus and cortex by dysregulating neurogenesis. Our study is the first to discover the detrimental impact of TFEB overexpression on neurogenesis during embryonic development, which has important reference significance for future TFEB overexpression interventions in NSCs for treatment. INTRODUCTION Transcription factor EB (TFEB), a key regulator of autophagy and lysosomal biogenesis, has diverse roles in various physiological processes. Enhancing lysosomal function by TFEB activation has recently been implicated in restoring neural stem cell (NSC) function. Overexpression of TFEB can inhibit the cell cycle of newborn cortical NSCs. It has also been found that TFEB regulates the pluripotency transcriptional network in mouse embryonic stem cells independent of autophagy and lysosomal biogenesis. This study aims to explore the effects of TFEB activation on neurogenesis in vivo through transgenic mice. METHODS We developed a glial fibrillary acidic protein (GFAP)-driven TFEB overexpression mouse model (TFEB GoE) by crossing the floxed TFEB overexpression mice and hGFAP-Cre mice. We performed immunohistochemical and fluorescence staining on brain tissue from newborn mice to assess neurogenesis changes, employing markers such as GFAP, Nestin, Ki67, doublecortin (DCX), Tbr1, and NeuN to trace different stages of neural development and cell proliferation. RESULTS TFEB GoE mice exhibited premature mortality, dying 10-20 days after birth. Immunohistochemical analysis revealed significant abnormalities, including disrupted hippocampal structure and cortical layering. Compared to control mice, TFEB GoE mice showed a marked increase in radial glial cells (RGCs) in the hippocampus and cortex, with Ki67 staining indicating these cells were predominantly in a quiescent state. This suggests that TFEB overexpression suppresses RGC proliferation. Additionally, abnormal distributions of migrating neurons and mature neurons were observed, highlighted by DCX, Tbr1, and NeuN staining, indicating a disruption in normal neurogenesis. CONCLUSION This study, using transgenic animals in vivo, revealed that GFAP-driven TFEB overexpression leads to abnormal neural layering in the hippocampus and cortex by dysregulating neurogenesis. Our study is the first to discover the detrimental impact of TFEB overexpression on neurogenesis during embryonic development, which has important reference significance for future TFEB overexpression interventions in NSCs for treatment.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurology, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Jiaxin Cao
- Institute of Anatomy and Histology and Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Haichao Chen
- Institute of Anatomy and Histology and Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yuezhang Ma
- Institute of Anatomy and Histology and Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yishu Zhang
- Institute of Anatomy and Histology and Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiaomei Su
- Institute of Anatomy and Histology and Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yuhong Jing
- Institute of Anatomy and Histology and Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yonggang Wang
- Department of Neurology, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| |
Collapse
|
32
|
Moors TE, Morella ML, Bertran-Cobo C, Geut H, Udayar V, Timmermans-Huisman E, Ingrassia AMT, Brevé JJP, Bol JGJM, Bonifati V, Jagasia R, van de Berg WDJ. Altered TFEB subcellular localization in nigral neurons of subjects with incidental, sporadic and GBA-related Lewy body diseases. Acta Neuropathol 2024; 147:67. [PMID: 38581586 PMCID: PMC10998821 DOI: 10.1007/s00401-024-02707-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 02/14/2024] [Accepted: 02/14/2024] [Indexed: 04/08/2024]
Abstract
Transcription factor EB (TFEB) is a master regulator of genes involved in the maintenance of autophagic and lysosomal homeostasis, processes which have been implicated in the pathogenesis of GBA-related and sporadic Parkinson's disease (PD), and dementia with Lewy bodies (DLB). TFEB activation results in its translocation from the cytosol to the nucleus. Here, we investigated TFEB subcellular localization and its relation to intracellular alpha-synuclein (aSyn) accumulation in post-mortem human brain of individuals with either incidental Lewy body disease (iLBD), GBA-related PD/DLB (GBA-PD/DLB) or sporadic PD/DLB (sPD/DLB), compared to control subjects. We analyzed nigral dopaminergic neurons using high-resolution confocal and stimulated emission depletion (STED) microscopy and semi-quantitatively scored the TFEB subcellular localization patterns. We observed reduced nuclear TFEB immunoreactivity in PD/DLB patients compared to controls, both in sporadic and GBA-related cases, as well as in iLBD cases. Nuclear depletion of TFEB was more pronounced in neurons with Ser129-phosphorylated (pSer129) aSyn accumulation in all groups. Importantly, we observed previously-unidentified TFEB-immunopositive perinuclear clusters in human dopaminergic neurons, which localized at the Golgi apparatus. These TFEB clusters were more frequently observed and more severe in iLBD, sPD/DLB and GBA-PD/DLB compared to controls, particularly in pSer129 aSyn-positive neurons, but also in neurons lacking detectable aSyn accumulation. In aSyn-negative cells, cytoplasmic TFEB clusters were more frequently observed in GBA-PD/DLB and iLBD patients, and correlated with reduced GBA enzymatic activity as well as increased Braak LB stage. Altered TFEB distribution was accompanied by a reduction in overall mRNA expression levels of selected TFEB-regulated genes, indicating a possible early dysfunction of lysosomal regulation. Overall, we observed cytoplasmic TFEB retention and accumulation at the Golgi in cells without apparent pSer129 aSyn accumulation in iLBD and PD/DLB patients. This suggests potential TFEB impairment at the early stages of cellular disease and underscores TFEB as a promising therapeutic target for synucleinopathies.
Collapse
Affiliation(s)
- Tim E Moors
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Martino L Morella
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Cesc Bertran-Cobo
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Hanneke Geut
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Vinod Udayar
- Roche Pharma Research and Early Development; Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center, Basel, Switzerland
| | - Evelien Timmermans-Huisman
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Angela M T Ingrassia
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - John J P Brevé
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - John G J M Bol
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Vincenzo Bonifati
- Erasmus MC, Department of Clinical Genetics, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ravi Jagasia
- Roche Pharma Research and Early Development; Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center, Basel, Switzerland
| | - Wilma D J van de Berg
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije University, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands.
| |
Collapse
|
33
|
Ojalvo-Pacheco J, Yakhine-Diop SMS, Fuentes JM, Paredes-Barquero M, Niso-Santano M. Role of TFEB in Huntington's Disease. BIOLOGY 2024; 13:238. [PMID: 38666850 PMCID: PMC11048341 DOI: 10.3390/biology13040238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/26/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease caused by an expansion of the CAG trinucleotide repeat in exon 1 of the huntingtin (HTT) gene. This expansion leads to a polyglutamine (polyQ) tract at the N-terminal end of HTT, which reduces the solubility of the protein and promotes its accumulation. Inefficient clearance of mutant HTT (mHTT) by the proteasome or autophagy-lysosomal system leads to accumulation of oligomers and toxic protein aggregates in neurons, resulting in impaired proteolytic systems, transcriptional dysregulation, impaired axonal transport, mitochondrial dysfunction and cellular energy imbalance. Growing evidence suggests that the accumulation of mHTT aggregates and autophagic and/or lysosomal dysfunction are the major pathogenic mechanisms underlying HD. In this context, enhancing autophagy may be an effective therapeutic strategy to remove protein aggregates and improve cell function. Transcription factor EB (TFEB), a master transcriptional regulator of autophagy, controls the expression of genes critical for autophagosome formation, lysosomal biogenesis, lysosomal function and autophagic flux. Consequently, the induction of TFEB activity to promote intracellular clearance may be a therapeutic strategy for HD. However, while some studies have shown that overexpression of TFEB facilitates the clearance of mHTT aggregates and ameliorates the disease phenotype, others indicate such overexpression may lead to mHTT co-aggregation and worsen disease progression. Further studies are necessary to confirm whether TFEB modulation could be an effective therapeutic strategy against mHTT-mediated toxicity in different disease models.
Collapse
Affiliation(s)
- Javier Ojalvo-Pacheco
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Caceres, Spain; (J.O.-P.); (S.M.S.Y.-D.); (J.M.F.)
| | - Sokhna M. S. Yakhine-Diop
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Caceres, Spain; (J.O.-P.); (S.M.S.Y.-D.); (J.M.F.)
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativa, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Caceres, Spain
| | - José M. Fuentes
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Caceres, Spain; (J.O.-P.); (S.M.S.Y.-D.); (J.M.F.)
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativa, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Caceres, Spain
| | - Marta Paredes-Barquero
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativa, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Caceres, Spain
| | - Mireia Niso-Santano
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Caceres, Spain; (J.O.-P.); (S.M.S.Y.-D.); (J.M.F.)
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativa, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Caceres, Spain
| |
Collapse
|
34
|
Lei HY, Pi GL, He T, Xiong R, Lv JR, Liu JL, Wu DQ, Li MZ, Shi K, Li SH, Yu NN, Gao Y, Yu HL, Wei LY, Wang X, Zhou QZ, Zou PL, Zhou JY, Liu YZ, Shen NT, Yang J, Ke D, Wang Q, Liu GP, Yang XF, Wang JZ, Yang Y. Targeting vulnerable microcircuits in the ventral hippocampus of male transgenic mice to rescue Alzheimer-like social memory loss. Mil Med Res 2024; 11:16. [PMID: 38462603 PMCID: PMC10926584 DOI: 10.1186/s40779-024-00512-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 01/11/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Episodic memory loss is a prominent clinical manifestation of Alzheimer's disease (AD), which is closely related to tau pathology and hippocampal impairment. Due to the heterogeneity of brain neurons, the specific roles of different brain neurons in terms of their sensitivity to tau accumulation and their contribution to AD-like social memory loss remain unclear. Therefore, further investigation is necessary. METHODS We investigated the effects of AD-like tau pathology by Tandem mass tag proteomic and phosphoproteomic analysis, social behavioural tests, hippocampal electrophysiology, immunofluorescence staining and in vivo optical fibre recording of GCaMP6f and iGABASnFR. Additionally, we utilized optogenetics and administered ursolic acid (UA) via oral gavage to examine the effects of these agents on social memory in mice. RESULTS The results of proteomic and phosphoproteomic analyses revealed the characteristics of ventral hippocampal CA1 (vCA1) under both physiological conditions and AD-like tau pathology. As tau progressively accumulated, vCA1, especially its excitatory and parvalbumin (PV) neurons, were fully filled with mislocated and phosphorylated tau (p-Tau). This finding was not observed for dorsal hippocampal CA1 (dCA1). The overexpression of human tau (hTau) in excitatory and PV neurons mimicked AD-like tau accumulation, significantly inhibited neuronal excitability and suppressed distinct discrimination-associated firings of these neurons within vCA1. Photoactivating excitatory and PV neurons in vCA1 at specific rhythms and time windows efficiently ameliorated tau-impaired social memory. Notably, 1 month of UA administration efficiently decreased tau accumulation via autophagy in a transcription factor EB (TFEB)-dependent manner and restored the vCA1 microcircuit to ameliorate tau-impaired social memory. CONCLUSION This study elucidated distinct protein and phosphoprotein networks between dCA1 and vCA1 and highlighted the susceptibility of the vCA1 microcircuit to AD-like tau accumulation. Notably, our novel findings regarding the efficacy of UA in reducing tau load and targeting the vCA1 microcircuit may provide a promising strategy for treating AD in the future.
Collapse
Affiliation(s)
- Hui-Yang Lei
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gui-Lin Pi
- Department of Traditional Chinese Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Ting He
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rui Xiong
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing-Ru Lv
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia-Le Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dong-Qin Wu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Meng-Zhu Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Kun Shi
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shi-Hong Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Na-Na Yu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yang Gao
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hui-Ling Yu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lin-Yu Wei
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xin Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qiu-Zhi Zhou
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pei-Lin Zou
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia-Yang Zhou
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ying-Zhou Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Nai-Ting Shen
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jie Yang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dan Ke
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xi-Fei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, Jiangsu, China.
| | - Ying Yang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
35
|
Chen H, Hinz K, Zhang C, Rodriguez Y, Williams SN, Niu M, Ma X, Chao X, Frazier AL, McCarson KE, Wang X, Peng Z, Liu W, Ni HM, Zhang J, Swerdlow RH, Ding WX. Late-Life Alcohol Exposure Does Not Exacerbate Age-Dependent Reductions in Mouse Spatial Memory and Brain TFEB Activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.23.581774. [PMID: 38464149 PMCID: PMC10925107 DOI: 10.1101/2024.02.23.581774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Alcohol consumption is believed to affect Alzheimer's disease (AD) risk, but the contributing mechanisms are not well understood. A potential mediator of the proposed alcohol-AD connection is autophagy, a degradation pathway that maintains organelle and protein homeostasis. Autophagy is in turn regulated through the activity of Transcription factor EB (TFEB), which promotes lysosome and autophagy-related gene expression. To explore the effect of alcohol on brain TFEB and autophagy, we exposed young (3-month old) and aged (23-month old) mice to two alcohol-feeding paradigms and assessed biochemical, transcriptome, histology, and behavioral endpoints. In young mice, alcohol decreased hippocampal nuclear TFEB staining but increased SQSTM1/p62, LC3-II, ubiquitinated proteins, and phosphorylated Tau. Hippocampal TFEB activity was lower in aged mice than it was in young mice, and Gao-binge alcohol feeding did not worsen the age-related reduction in TFEB activity. To better assess the impact of chronic alcohol exposure, we fed young and aged mice alcohol for four weeks before completing Morris Water and Barnes Maze spatial memory testing. The aged mice showed worse spatial memory on both tests. While alcohol feeding slightly impaired spatial memory in the young mice, it had little effect or even slightly improved spatial memory in the aged mice. These findings suggest that aging is a far more important driver of spatial memory impairment and reduced autophagy flux than alcohol consumption.
Collapse
|
36
|
Giamogante F, Barazzuol L, Maiorca F, Poggio E, Esposito A, Masato A, Napolitano G, Vagnoni A, Calì T, Brini M. A SPLICS reporter reveals [Formula: see text]-synuclein regulation of lysosome-mitochondria contacts which affects TFEB nuclear translocation. Nat Commun 2024; 15:1516. [PMID: 38374070 PMCID: PMC10876553 DOI: 10.1038/s41467-024-46007-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 02/07/2024] [Indexed: 02/21/2024] Open
Abstract
Mitochondrial and lysosomal activities are crucial to maintain cellular homeostasis: optimal coordination is achieved at their membrane contact sites where distinct protein machineries regulate organelle network dynamics, ions and metabolites exchange. Here we describe a genetically encoded SPLICS reporter for short- and long- juxtapositions between mitochondria and lysosomes. We report the existence of narrow and wide lysosome-mitochondria contacts differently modulated by mitophagy, autophagy and genetic manipulation of tethering factors. The overexpression of α-synuclein (α-syn) reduces the apposition of mitochondria/lysosomes membranes and affects their privileged Ca2+ transfer, impinging on TFEB nuclear translocation. We observe enhanced TFEB nuclear translocation in α-syn-overexpressing cells. We propose that α-syn, by interfering with mitochondria/lysosomes tethering impacts on local Ca2+ regulated pathways, among which TFEB mediated signaling, and in turn mitochondrial and lysosomal function. Defects in mitochondria and lysosome represent a common hallmark of neurodegenerative diseases: targeting their communication could open therapeutic avenues.
Collapse
Affiliation(s)
- Flavia Giamogante
- Department of Biomedical Sciences (DSB), University of Padova, Padova, Italy
| | - Lucia Barazzuol
- Department of Biomedical Sciences (DSB), University of Padova, Padova, Italy
| | | | - Elena Poggio
- Department of Biology (DIBIO), University of Padova, Padova, Italy
| | - Alessandra Esposito
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Medical and Translational Science, Federico II University, Naples, Italy
| | - Anna Masato
- Department of Biology (DIBIO), University of Padova, Padova, Italy
- UK-Dementia Research Institute at UCL, University College London, London, UK
| | - Gennaro Napolitano
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Medical and Translational Science, Federico II University, Naples, Italy
| | - Alessio Vagnoni
- Department of Basic and Clinical Neurosciences, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Tito Calì
- Department of Biomedical Sciences (DSB), University of Padova, Padova, Italy.
- Padova Neuroscience Center (PNC), University of Padova, Padova, Italy.
- Study Center for Neurodegeneration (CESNE), University of Padova, Padova, Italy.
| | - Marisa Brini
- Department of Biology (DIBIO), University of Padova, Padova, Italy.
- Study Center for Neurodegeneration (CESNE), University of Padova, Padova, Italy.
- Department of Pharmaceutical and Pharmacological Sciences (DSF), University of Padova, Padova, Italy.
| |
Collapse
|
37
|
Jia N, Ganesan D, Guan H, Jeong YY, Han S, Nissenbaum M, Kusnecov AW, Cai Q. Mitochondrial bioenergetics stimulates autophagy for pathological tau clearance in tauopathy neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.12.579959. [PMID: 38405759 PMCID: PMC10888759 DOI: 10.1101/2024.02.12.579959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Hyperphosphorylation and aggregation of microtubule-associated tau is a pathogenic hallmark of tauopathies and a defining feature of Alzheimer's disease (AD). Pathological tau is targeted by autophagy for clearance, but autophagy dysfunction is indicated in tauopathy. While mitochondrial bioenergetic failure has been shown to precede the development of tau pathology, it is unclear whether energy metabolism deficiency is involved in tauopathy-related autophagy defects. Here, we reveal that stimulation of anaplerotic metabolism restores defective oxidative phosphorylation (OXPHOS) in tauopathy which, strikingly, leads to enhanced autophagy and pronounced tau clearance. OXPHOS-induced autophagy is attributed to increased ATP-dependent phosphatidylethanolamine biosynthesis in mitochondria. Excitingly, early bioenergetic stimulation boosts autophagy activity and reduces tau pathology, thereby counteracting memory impairment in tauopathy mice. Taken together, our study sheds light on a pivotal role of bioenergetic dysfunction in tauopathy-linked autophagy defects and suggests a new therapeutic strategy to prevent toxic tau buildup in AD and other tauopathies.
Collapse
|
38
|
Nicoletti T, Bink A, Helmchen B, Briel N, Frontzek K, Vlad B, Gaspert A, Boudriot E, Jung HH, Reuss AM, Weller M, Hortobágyi T. Neurologic involvement in cystinosis: Focus on brain lesions and new evidence of four-repeat (4R-) Tau immunoreactivity. J Neurol Sci 2024; 456:122841. [PMID: 38101161 DOI: 10.1016/j.jns.2023.122841] [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: 08/31/2023] [Revised: 11/10/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
Nephropathic cystinosis is a rare autosomal recessive storage disorder caused by CTNS gene mutations, leading to autophagy-lysosomal pathway impairment and cystine crystals accumulation. Neurologic involvement is highly variable and includes both neurodevelopmental and neurodegenerative disturbances, as well as focal neurologic deficits. By presenting longitudinal data of a 28-year-old patient with a large infratentorial lesion, we summarized the pathology, clinical and imaging features of neurological involvement in cystinosis patients. Brain damage in form of cystinosis-related cerebral lesions occurs in advanced disease phases and is characterized by the accumulation of cystine crystals, subsequent inflammation with vasculitis-like features, necrosis, and calcification. Epilepsy is a frequent comorbidity in affected individuals. Steroids might play a role in the symptomatic treatment of "stroke-like" episodes due to edematous-inflammatory lesions, but probably do not change the overall prognosis. Lifelong compliance to depleting therapy with cysteamine still represents the main therapeutic option. However, consequences of CTNS gene defects are not restricted to cystine accumulation. New evidence of four-repeat (4R-) Tau immunoreactivity suggests concurrent progressive neurodegeneration in cystinosis patients, highlighting the need of innovative therapeutic strategies, and shedding light on the crosstalk between proteinopathies and autophagy-lysosomal system defects. Eventually, emerging easily accessible biomarkers such as serum neurofilament light chains (NfL) might detect subclinical neurologic involvement in cystinosis patients.
Collapse
Affiliation(s)
- Tommaso Nicoletti
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland.
| | - Andrea Bink
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland; Department of Neuroradiology, University Hospital Zurich, Switzerland
| | - Birgit Helmchen
- Department of Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Nils Briel
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland; Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, Munich, Germany
| | - Karl Frontzek
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland; Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - Benjamin Vlad
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Ariana Gaspert
- Department of Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Elisabeth Boudriot
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Hans Heinrich Jung
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Anna Maria Reuss
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Tibor Hortobágyi
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
39
|
Shi H, Zhao Y. Modulation of Tau Pathology in Alzheimer's Disease by Dietary Bioactive Compounds. Int J Mol Sci 2024; 25:831. [PMID: 38255905 PMCID: PMC10815728 DOI: 10.3390/ijms25020831] [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: 10/31/2023] [Revised: 01/02/2024] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Tau is a microtubule-associated protein essential for microtubule assembly and stability in neurons. The abnormal intracellular accumulation of tau aggregates is a major characteristic of brains from patients with Alzheimer's disease (AD) and other tauopathies. In AD, the presence of neurofibrillary tangles (NFTs), which is composed of hyperphosphorylated tau protein, is positively correlated with the severity of the cognitive decline. Evidence suggests that the accumulation and aggregation of tau cause synaptic dysfunction and neuronal degeneration. Thus, the prevention of abnormal tau phosphorylation and elimination of tau aggregates have been proposed as therapeutic strategies for AD. However, currently tau-targeting therapies for AD and other tauopathies are limited. A number of dietary bioactive compounds have been found to modulate the posttranslational modifications of tau, including phosphorylation, small ubiquitin-like modifier (SUMO) mediated modification (SUMOylation) and acetylation, as well as inhibit tau aggregation and/or promote tau degradation. The advantages of using these dietary components over synthetic substances in AD prevention and intervention are their safety and accessibility. This review summarizes the mechanisms leading to tau pathology in AD and highlights the effects of bioactive compounds on the hyperphosphorylation, aggregation and clearance of tau protein. The potential of using these bioactive compounds for AD prevention and intervention is also discussed.
Collapse
Affiliation(s)
- Huahua Shi
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, China;
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Yan Zhao
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, China;
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| |
Collapse
|
40
|
Wang B, Martini-Stoica H, Qi C, Lu TC, Wang S, Xiong W, Qi Y, Xu Y, Sardiello M, Li H, Zheng H. TFEB-vacuolar ATPase signaling regulates lysosomal function and microglial activation in tauopathy. Nat Neurosci 2024; 27:48-62. [PMID: 37985800 DOI: 10.1038/s41593-023-01494-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 10/13/2023] [Indexed: 11/22/2023]
Abstract
Transcription factor EB (TFEB) mediates gene expression through binding to the coordinated lysosome expression and regulation (CLEAR) sequence. TFEB targets include subunits of the vacuolar ATPase (v-ATPase), which are essential for lysosome acidification. Single-nucleus RNA sequencing of wild-type and PS19 (Tau) transgenic mice expressing the P301S mutant tau identified three unique microglia subclusters in Tau mice that were associated with heightened lysosome and immune pathway genes. To explore the lysosome-immune relationship, we specifically disrupted the TFEB-v-ATPase signaling by creating a knock-in mouse line in which the CLEAR sequence of one of the v-ATPase subunits, Atp6v1h, was mutated. CLEAR mutant exhibited a muted response to TFEB, resulting in impaired lysosomal acidification and activity. Crossing the CLEAR mutant with Tau mice led to higher tau pathology but diminished microglia response. These microglia were enriched in a subcluster low in mTOR and HIF-1 pathways and were locked in a homeostatic state. Our studies demonstrate a physiological function of TFEB-v-ATPase signaling in maintaining lysosomal homeostasis and a critical role of the lysosome in mounting a microglia and immune response in tauopathy and Alzheimer's disease.
Collapse
Affiliation(s)
- Baiping Wang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Heidi Martini-Stoica
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
- Department of Otolaryngology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Chuangye Qi
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - Tzu-Chiao Lu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - Shuo Wang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - Wen Xiong
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - Yanyan Qi
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - Yin Xu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- School of Mental Health and Psychological Sciences, Anhui Medical University, Anhui, China
| | - Marco Sardiello
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Dan and Jan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - Hongjie Li
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
41
|
Yan H, Feng L, Li M. The Role of Traditional Chinese Medicine Natural Products in β-Amyloid Deposition and Tau Protein Hyperphosphorylation in Alzheimer's Disease. Drug Des Devel Ther 2023; 17:3295-3323. [PMID: 38024535 PMCID: PMC10655607 DOI: 10.2147/dddt.s380612] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/02/2023] [Indexed: 12/01/2023] Open
Abstract
Alzheimer's disease is a prevalent form of dementia among elderly individuals and is characterized by irreversible neurodegeneration. Despite extensive research, the exact causes of this complex disease remain unclear. Currently available drugs for Alzheimer's disease treatment are limited in their effectiveness, often targeting a single aspect of the disease and causing significant adverse effects. Moreover, these medications are expensive, placing a heavy burden on patients' families and society as a whole. Natural compounds and extracts offer several advantages, including the ability to target multiple pathways and exhibit high efficiency with minimal toxicity. These attributes make them promising candidates for the prevention and treatment of Alzheimer's disease. In this paper, we provide a summary of the common natural products used in Chinese medicine for different pathogeneses of AD. Our aim is to offer new insights and ideas for the further development of natural products in Chinese medicine and the treatment of AD.
Collapse
Affiliation(s)
- Huiying Yan
- Department of Neurology, the Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, Jilin Province, People’s Republic of China
| | - Lina Feng
- Shandong Key Laboratory of TCM Multi-Targets Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, People’s Republic of China
| | - Mingquan Li
- Department of Neurology, the Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, Jilin Province, People’s Republic of China
| |
Collapse
|
42
|
Takla M, Keshri S, Rubinsztein DC. The post-translational regulation of transcription factor EB (TFEB) in health and disease. EMBO Rep 2023; 24:e57574. [PMID: 37728021 PMCID: PMC10626434 DOI: 10.15252/embr.202357574] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/10/2023] [Accepted: 08/25/2023] [Indexed: 09/21/2023] Open
Abstract
Transcription factor EB (TFEB) is a basic helix-loop-helix leucine zipper transcription factor that acts as a master regulator of lysosomal biogenesis, lysosomal exocytosis, and macro-autophagy. TFEB contributes to a wide range of physiological functions, including mitochondrial biogenesis and innate and adaptive immunity. As such, TFEB is an essential component of cellular adaptation to stressors, ranging from nutrient deprivation to pathogenic invasion. The activity of TFEB depends on its subcellular localisation, turnover, and DNA-binding capacity, all of which are regulated at the post-translational level. Pathological states are characterised by a specific set of stressors, which elicit post-translational modifications that promote gain or loss of TFEB function in the affected tissue. In turn, the resulting increase or decrease in survival of the tissue in which TFEB is more or less active, respectively, may either benefit or harm the organism as a whole. In this way, the post-translational modifications of TFEB account for its otherwise paradoxical protective and deleterious effects on organismal fitness in diseases ranging from neurodegeneration to cancer. In this review, we describe how the intracellular environment characteristic of different diseases alters the post-translational modification profile of TFEB, enabling cellular adaptation to a particular pathological state.
Collapse
Affiliation(s)
- Michael Takla
- Department of Medical Genetics, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
- UK Dementia Research Institute, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
| | - Swati Keshri
- Department of Medical Genetics, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
- UK Dementia Research Institute, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
- UK Dementia Research Institute, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
| |
Collapse
|
43
|
Li J, Liu W, Sun W, Rao X, Chen X, Yu L. A Study on Autophagy Related Biomarkers in Alzheimer's Disease Based on Bioinformatics. Cell Mol Neurobiol 2023; 43:3693-3703. [PMID: 37418137 PMCID: PMC11409956 DOI: 10.1007/s10571-023-01379-9] [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: 04/21/2023] [Accepted: 06/20/2023] [Indexed: 07/08/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with an annual incidence increase that poses significant health risks to people. However, the pathogenesis of AD is still unclear. Autophagy, as an intracellular mechanism can degrade damaged cellular components and abnormal proteins, which is closely related to AD pathology. The goal of this work is to uncover the intimate association between autophagy and AD, and to mine potential autophagy-related AD biomarkers by identifying key differentially expressed autophagy genes (DEAGs) and exploring the potential functions of these genes. GSE63061 and GSE140831 gene expression profiles of AD were downloaded from the Gene Expression Omnibus (GEO) database. R language was used to standardize and differentially expressed genes (DEGs) of AD expression profiles. A total of 259 autophagy-related genes were discovered through the autophagy gene databases ATD and HADb. The differential genes of AD and autophagy genes were integrated and analyzed to screen out DEAGs. Then the potential biological functions of DEAGs were predicted, and Cytoscape software was used to detect the key DEAGs. There were ten DEAGs associated with the AD development, including nine up-regulated genes (CAPNS1, GAPDH, IKBKB, LAMP1, LAMP2, MAPK1, PRKCD, RAB24, RAF1) and one down-regulated gene (CASP1). The correlation analysis reveals the potential correlation among 10 core DEAGs. Finally, the significance of the detected DEAGs expression was verified, and the value of DEAGs in AD pathology was detected by the receiver operating characteristic curve. The area under the curve values indicated that ten DEAGs are potentially valuable for the study of the pathological mechanism and may become biomarkers of AD. This pathway analysis and DEAG screening in this study found a strong association between autophagy-related genes and AD, providing new insights into the pathological progression of AD. Exploring the relationship between autophagy and AD: analysis of genes associated with autophagy in pathological mechanisms of AD using bioinformatics. 10 autophagy-related genes play an important role in the pathological mechanisms of AD.
Collapse
Affiliation(s)
- Jian Li
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Wenjia Liu
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Wen Sun
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Xin Rao
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China.
| | - Xiaodong Chen
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China.
- School of Electronic Engineering and Computer Science, Queen Mary University of London, London, E1 4NS, UK.
| | - Liyang Yu
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China.
| |
Collapse
|
44
|
Chae CW, Yoon JH, Lim JR, Park JY, Cho JH, Jung YH, Choi GE, Lee HJ, Han HJ. TRIM16-mediated lysophagy suppresses high-glucose-accumulated neuronal Aβ. Autophagy 2023; 19:2752-2768. [PMID: 37357416 PMCID: PMC10472864 DOI: 10.1080/15548627.2023.2229659] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 06/13/2023] [Accepted: 06/21/2023] [Indexed: 06/27/2023] Open
Abstract
ABBREVIATIONS Aβ: amyloid β; AD: Alzheimer disease; AMPK: 5' adenosine monophosphate-activated protein kinase; CTSB: cathepsin B; CTSD: cathepsin D; DM: diabetes mellitus; ESCRT: endosomal sorting complex required for transport; FBXO27: F-box protein 27; iPSC-NDs: induced pluripotent stem cell-derived neuronal differentiated cells; LAMP1: lysosomal-associated membrane protein 1; LMP: lysosomal membrane permeabilization; LRSAM1: leucine rich repeat and sterile alpha motif containing 1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTORC1: mechanistic target of rapamycin kinase complex 1; p-MAPT/tau: phosphorylated microtubule associated protein tau; ROS: reactive oxygen species; STZ: streptozotocin; TFE3: transcription factor E3; TFEB: transcription factor EB; TRIM16: tripartite motif containing 16; UBE2QL1: ubiquitin conjugating enzyme E2 Q family like 1; VCP: valosin containing protein.
Collapse
Affiliation(s)
- Chang Woo Chae
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Jee Hyeon Yoon
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Jae Ryong Lim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Ji Yong Park
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Ji Hyeon Cho
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Gee Euhn Choi
- Laboratory of Veterinary Biochemistry, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, South Korea
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, South Korea
| | - Hyun Jik Lee
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, South Korea
- Institute for Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| |
Collapse
|
45
|
Zhang L, Li Z, Zhang L, Qin Y, Yu D. Dissecting the multifaced function of transcription factor EB (TFEB) in human diseases: From molecular mechanism to pharmacological modulation. Biochem Pharmacol 2023; 215:115698. [PMID: 37482200 DOI: 10.1016/j.bcp.2023.115698] [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: 05/09/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
The transcription factor EB (TFEB) is a transcription factor of the MiT/TFE family that translocations from the cytoplasm to the nucleus in response to various stimuli, including lysosomal stress and nutrient starvation. By activating genes involved in lysosomal function, autophagy, and lipid metabolism, TFEB plays a crucial role in maintaining cellular homeostasis. Dysregulation of TFEB has been implicated in various diseases, including cancer, neurodegenerative diseases, metabolic diseases, cardiovascular diseases, infectious diseases, and inflammatory diseases. Therefore, modulating TFEB activity with agonists or inhibitors may have therapeutic potential. In this review, we reviewed the recently discovered regulatory mechanisms of TFEB and their impact on human diseases. Additionally, we also summarize the existing TFEB inhibitors and agonists (targeted and non-targeted) and discuss unresolved issues and future research directions in the field. In summary, this review sheds light on the crucial role of TFEB, which may pave the way for its translation from basic research to practical applications, bringing us closer to realizing the full potential of TFEB in various fields.
Collapse
Affiliation(s)
- Lijuan Zhang
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Zhijia Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Yuan Qin
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China.
| | - Dongke Yu
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| |
Collapse
|
46
|
de Wet S, Theart R, Loos B. Cogs in the autophagic machine-equipped to combat dementia-prone neurodegenerative diseases. Front Mol Neurosci 2023; 16:1225227. [PMID: 37720551 PMCID: PMC10500130 DOI: 10.3389/fnmol.2023.1225227] [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: 05/18/2023] [Accepted: 07/31/2023] [Indexed: 09/19/2023] Open
Abstract
Neurodegenerative diseases are often characterized by hydrophobic inclusion bodies, and it may be the case that the aggregate-prone proteins that comprise these inclusion bodies are in fact the cause of neurotoxicity. Indeed, the appearance of protein aggregates leads to a proteostatic imbalance that causes various interruptions in physiological cellular processes, including lysosomal and mitochondrial dysfunction, as well as break down in calcium homeostasis. Oftentimes the approach to counteract proteotoxicity is taken to merely upregulate autophagy, measured by an increase in autophagosomes, without a deeper assessment of contributors toward effective turnover through autophagy. There are various ways in which autophagy is regulated ranging from the mammalian target of rapamycin (mTOR) to acetylation status of proteins. Healthy mitochondria and the intracellular energetic charge they preserve are key for the acidification status of lysosomes and thus ensuring effective clearance of components through the autophagy pathway. Both mitochondria and lysosomes have been shown to bear functional protein complexes that aid in the regulation of autophagy. Indeed, it may be the case that minimizing the proteins associated with the respective neurodegenerative pathology may be of greater importance than addressing molecularly their resulting inclusion bodies. It is in this context that this review will dissect the autophagy signaling pathway, its control and the manner in which it is molecularly and functionally connected with the mitochondrial and lysosomal system, as well as provide a summary of the role of autophagy dysfunction in driving neurodegenerative disease as a means to better position the potential of rapamycin-mediated bioactivities to control autophagy favorably.
Collapse
Affiliation(s)
- Sholto de Wet
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Rensu Theart
- Department of Electric and Electronic Engineering, Stellenbosch University, Stellenbosch, South Africa
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
47
|
Comerota MM, Gedam M, Xiong W, Jin F, Deng L, Wang MC, Wang J, Zheng H. Oleoylethanolamide facilitates PPARα and TFEB signaling and attenuates Aβ pathology in a mouse model of Alzheimer's disease. Mol Neurodegener 2023; 18:56. [PMID: 37580742 PMCID: PMC10426131 DOI: 10.1186/s13024-023-00648-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/08/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Age is the strongest risk factor for the development of Alzheimer's disease (AD). Besides the pathological hallmarks of β-amyloid (Aβ) plaques and neurofibrillary tangles, emerging evidence demonstrates a critical role of microglia and neuroinflammation in AD pathogenesis. Oleoylethanolamide (OEA) is an endogenous lipid amide that has been shown to promote lifespan and healthspan in C. elegans through regulation of lysosome-to-nucleus signaling and cellular metabolism. The goal of our study was to determine the role of OEA in the mediation of microglial activity and AD pathology using its stable analog, KDS-5104. METHODS We used primary microglial cultures and genetic and pharmacological approaches to examine the signaling mechanisms and functional roles of OEA in mediating Aβ phagocytosis and clearance, lipid metabolism and inflammasome formation. Further, we tested the effect of OEA in vivo in acute LPS-induced neuroinflammation and by chronic treatment of 5xFAD mice. RESULTS We found that OEA activates PPARα signaling and its downstream cell-surface receptor CD36 activity. In addition, OEA promotes TFEB lysosomal function in a PPARα-dependent but mTORC1-independent manner, the combination of which leads to enhanced microglial Aβ uptake and clearance. These are associated with the suppression of LPS-induced lipid droplet accumulation and inflammasome activation. Chronic treatment of 5xFAD mice with KDS-5104 restored dysregulated lipid profiles, reduced reactive gliosis and Aβ pathology and rescued cognitive impairments. CONCLUSION Together, our study provides support that augmenting OEA-mediated lipid signaling may offer therapeutic benefit against aging and AD through modulating lipid metabolism and microglia phagocytosis and clearance.
Collapse
Affiliation(s)
- Michele M Comerota
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Manasee Gedam
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Translational Biology and Molecular Medicine Graduate Program, Houston, TX, USA
| | - Wen Xiong
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Feng Jin
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Pharmacology and Chemical Biology, Houston, TX, USA
| | - Lisheng Deng
- Department of Pharmacology and Chemical Biology, Houston, TX, USA
| | - Meng C Wang
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Houston, TX, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
- HHMI Janelia Research Campus, Ashburn, VA, USA
| | - Jin Wang
- Department of Pharmacology and Chemical Biology, Houston, TX, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Translational Biology and Molecular Medicine Graduate Program, Houston, TX, USA.
- Department of Molecular and Human Genetics, Houston, TX, USA.
| |
Collapse
|
48
|
Ning B, Hang S, Zhang W, Mao C, Li D. An update on the bridging factors connecting autophagy and Nrf2 antioxidant pathway. Front Cell Dev Biol 2023; 11:1232241. [PMID: 37621776 PMCID: PMC10445655 DOI: 10.3389/fcell.2023.1232241] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/24/2023] [Indexed: 08/26/2023] Open
Abstract
Macroautophagy/autophagy is a lysosome-dependent catabolic pathway for the degradation of intracellular proteins and organelles. Autophagy dysfunction is related to many diseases, including lysosomal storage diseases, cancer, neurodegenerative diseases, cardiomyopathy, and chronic metabolic diseases, in which increased reactive oxygen species (ROS) levels are also observed. ROS can randomly oxidize proteins, lipids, and DNA, causing oxidative stress and damage. Cells have developed various antioxidant pathways to reduce excessive ROS and maintain redox homeostasis. Treatment targeting only one aspect of diseases with autophagy dysfunction and oxidative stress shows very limited effects. Herein, identifying the bridging factors that can regulate both autophagy and antioxidant pathways is beneficial for dual-target therapies. This review intends to provide insights into the current identified bridging factors that connect autophagy and Nrf2 antioxidant pathway, as well as their tight interconnection with each other. These factors could be potential dual-purpose targets for the treatment of diseases implicated in both autophagy dysfunction and oxidative stress.
Collapse
Affiliation(s)
- Baike Ning
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Shuqi Hang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Wenhe Zhang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Caiwen Mao
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Dan Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
49
|
Zhang C, Chen H, Rodriguez Y, Ma X, Swerdlow RH, Zhang J, Ding WX. A perspective on autophagy and transcription factor EB in Alcohol-Associated Alzheimer's disease. Biochem Pharmacol 2023; 213:115576. [PMID: 37127251 PMCID: PMC11009931 DOI: 10.1016/j.bcp.2023.115576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/03/2023]
Abstract
Alzheimer's disease (AD) is the most common form of progressive dementia and there is no truly efficacious treatment. Accumulating evidence indicates that impaired autophagic function for removal of damaged mitochondria and protein aggregates such as amyloid and tau protein aggregates may contribute to the pathogenesis of AD. Epidemiologic studies have implicated alcohol abuse in promoting AD, yet the underlying mechanisms are poorly understood. In this review, we discuss mechanisms of selective autophagy for mitochondria and protein aggregates and how these mechanisms are impaired by aging and alcohol consumption. We also discuss potential genetic and pharmacological approaches for targeting autophagy/mitophagy, as well as lysosomal and mitochondrial biogenesis, for the potential prevention and treatment of AD.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hao Chen
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yssa Rodriguez
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xiaowen Ma
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Russell H Swerdlow
- Department of Neurology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jianhua Zhang
- Department of Pathology, Division of Molecular Cellular Pathology, University of Alabama at Birmingham, 901 19th street South, Birmingham, AL 35294, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Internal Medicine, Division of Gastroenterology, Hepatology & Motility, The University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
50
|
Zhou X, Chen X, Cheng X, Lin L, Quan S, Li S, Zhan R, Wu Q, Liu S. Paeoniflorin, ferulic acid, and atractylenolide III improved LPS-induced neuroinflammation of BV2 microglia cells by enhancing autophagy. J Pharmacol Sci 2023; 152:151-161. [PMID: 37169480 DOI: 10.1016/j.jphs.2023.04.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/02/2023] [Accepted: 04/13/2023] [Indexed: 05/13/2023] Open
Abstract
Microglia hyperactivation is an important cause of neuroinflammation in Alzheimer's disease (AD). Paeoniflorin (PF), ferulic acid (FA), and atractylenolide III (ATL) are potent in anti-inflammation and neuroprotection. Multiple components can act on different targets simultaneously to exert synergistic therapeutic effects and exploring the synergistic potential between compounds is an important area of research. We investigated the effects of PF, FA, and ATL, alone or in combination, on LPS-induced neuroinflammation and autophagy in BV2 microglia cells. We found that PF, FA, and ATL, alone or in combination, significantly reduced the production of inflammatory factors such as IL-6, IL-1β, and TNF-α, especially in the PF + FA + ATL group, which performed the best. In addition, the combination of PF, FA, and ATL significantly increased the expression of autophagy-related proteins p-AMPK, p-ULK1, Beclin1, LC3, and TFEB and decreased the expression of p62. Moreover, the restoration of autophagic flux by the combination of PF, FA, and ATL was abrogated by the addition of the autophagy inhibitor Wortmannin. In conclusion, PF, FA, and ATL have a synergistic effect in reducing LPS-induced inflammatory factor release from BV2 microglia cells, and its protective effect may be through activation of the AMPK/ULK1/TFEB autophagic signaling pathway.
Collapse
Affiliation(s)
- Xiaolan Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xingru Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoqing Cheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liting Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shijian Quan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shijun Li
- Department of Radiology, Chinese PLA General Hospital, Beijing, China; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, MA, USA
| | - Ruoting Zhan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China; Key Laboratory of Chinese Medicinal Resource from Lingnan, Guangzhou University of Chinese Medicine, Ministry of Education, Guangzhou, China
| | - Qingguang Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Sijun Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|