1
|
Braak H, Mayer B, Feldengut S, Schön M, Del Tredici K. Sequence and trajectory of early Alzheimer's disease-related tau inclusions in the hippocampal formation of cases without amyloid-β deposits. Acta Neuropathol 2025; 149:50. [PMID: 40407905 PMCID: PMC12102137 DOI: 10.1007/s00401-025-02862-x] [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: 07/19/2024] [Revised: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 05/26/2025]
Abstract
Sporadic Alzheimer's disease (AD) involves specific neuronal types and progresses in a systematic manner, permitting subdivision into six neuropathological stages. Neurofibrillary tangle (NFT) stages I-III display abnormal tau inclusions confined to subcortical nuclei and temporal allocortical regions, frequently without amyloid β (Aβ) deposition. We previously suggested a sequence of neuronal involvement in AD that could proceed from entorhinal pre-α cells to hippocampal prosubicular pyramidal cells and the CA1/CA2 sectors, from there to the thorny excrescences on mossy cells in CA3/CA4, and, finally, from the mossy cells to dentate fascia (Fd) granular cells. Here, we aimed to see if associations existed between the early NFT stages I-III, when Aβ deposits are frequently absent, and the following four categories: (1) anatomical regions and abnormal morphological tau changes in region-specific layers, (2) nerve cell loss, (3) APOE genotype, and (4) the trajectory (directionality) of tau progression in the hippocampal formation. To do so, we examined the transentorhinal/entorhinal regions and hippocampal formation using AT8-immunohistochemistry in 100 µm sections from N = 308 brains with tau inclusions lacking Aβ deposits between NFT stages I and III (average age at death 66.7 years for females, 66.4 years for males). Our results indicated a significantly (p < 0.001) ordered progression of abnormal tau in a direction opposite to currently known unidirectional intrahippocampal connections, thereby indirectly supporting the idea of transneuronal abnormal tau spreading, i.e., anterogradely, through the hippocampal formation. Tau-related neuronal loss was also significant (p < 0.001 for the transentorhinal/entorhinal regions and for sectors CA1/CA2 and p = 0.003 for CA3/CA4/Fd). These findings challenge the amyloid cascade and the PART hypotheses, corroborating the concept that early AD-related tau inclusions and tau-related neuronal loss occur independently of Aβ deposition.
Collapse
Affiliation(s)
- Heiko Braak
- Department of Neurology, Center for Biomedical Research, Clinical Neuroanatomy, University of Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany.
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, University of Ulm, 89075, Ulm, Germany
| | - Simone Feldengut
- Department of Neurology, Center for Biomedical Research, Clinical Neuroanatomy, University of Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany
| | - Michael Schön
- Institute for Anatomy and Cell Biology, University of Ulm, 89081, Ulm, Germany
| | - Kelly Del Tredici
- Department of Neurology, Center for Biomedical Research, Clinical Neuroanatomy, University of Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany
| |
Collapse
|
2
|
Sugandhi VV, Gadhave DG, Ugale AR, Kulkarni N, Nangare SN, Patil HP, Rath S, Saxena R, Lavate A, Patel AT, Jadhav A, Paudel KR. Advances in Alzheimer's Therapy: Exploring Neuropathological Mechanisms to Revolutionize the Future Therapeutic Landscape. Ageing Res Rev 2025; 109:102775. [PMID: 40403980 DOI: 10.1016/j.arr.2025.102775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 05/07/2025] [Accepted: 05/16/2025] [Indexed: 05/24/2025]
Abstract
Alzheimer's disease (AD) is still an excessively complicated neurological disorder that impacts millions of individuals globally. The ideal defensive feature of the central nervous system (CNS) is the intimate junction of endothelial cells, which functions as a biological barrier to safely control molecular transport throughout the brain. The blood-brain barrier (BBB) comprises tightly locked astrocyte cell junctions on CNS blood capillaries. This biological barrier shields the brain from hazardous toxins by preventing the entry of polar medications, cells, and ions. However, it is very challenging to provide any treatment to the brain for neurodegenerative illnesses like Alzheimer's. Different causative mechanisms, such as amyloid-β (Aβ) plaques, tubulin-associated unit (Tau) tangles, and neuroinflammation, cause neuronal dysfunction, leading to dementia and memory loss in the subject. Several treatments are approved for AD therapy, whereas most only help treat related symptoms. Disappointingly, current remedies have not been able to control the progression of AD due to associated side effects. Specific pathogenic mechanisms are involved in the initiation and development of this disease. Therefore, the expected survival of a patient with AD is limited and is approximately ten years. Hence, the pathogenic mechanism behind AD progression must be understood to better comprehend and improve the overall survival rate. This review highlighted the recent insights into AD pathogenesis, molecular mechanisms, advancements in theragnostic techniques, the existing updates of clinical trials, and emerging innovations for AD medicinal development. That has helped researchers develop other strategies to address the shortcomings of traditional medications.
Collapse
Affiliation(s)
- Vrashabh V Sugandhi
- College of Pharmacy & Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Dnyandev G Gadhave
- College of Pharmacy & Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA; Department of Pharmaceutics, Dattakala Shikshan Sanstha's, Dattakala College of Pharmacy (Affiliated to Savitribai Phule Pune University), Swami Chincholi, Daund, Pune, Maharashtra 413130, India.
| | - Akanksha R Ugale
- College of Pharmacy & Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Nilesh Kulkarni
- Department of Pharmaceutics, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra 425405, India
| | - Sopan N Nangare
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra 425405, India
| | - Harshal P Patil
- Department of Pharmaceutics, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra 425405, India
| | - Seepra Rath
- Center for Dermal Research, Rutgers, the State University of New Jersey, 145 Bevier Road, Piscataway 08854, USA
| | - Rahul Saxena
- Graduate Programs in Molecular Biosciences, Rutgers, the State University of New Jersey, Piscataway 08854, USA
| | - Amol Lavate
- Department of Pharmaceutics, Dattakala Shikshan Sanstha's, Dattakala College of Pharmacy (Affiliated to Savitribai Phule Pune University), Swami Chincholi, Daund, Pune, Maharashtra 413130, India
| | - Apeksha T Patel
- Department of Quality Assurance, Navinta III INC, Boca Raton, 33487, Florida, USA
| | - Ashish Jadhav
- Department of Pharmaceutics, Dattakala Shikshan Sanstha's, Dattakala College of Pharmacy (Affiliated to Savitribai Phule Pune University), Swami Chincholi, Daund, Pune, Maharashtra 413130, India
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life Science, Centenary Institute and University of Technology Sydney, Sydney 2007, Australia.
| |
Collapse
|
3
|
Wu X, Yang Z, Zou J, Gao H, Shao Z, Li C, Lei P. Protein kinases in neurodegenerative diseases: current understandings and implications for drug discovery. Signal Transduct Target Ther 2025; 10:146. [PMID: 40328798 PMCID: PMC12056177 DOI: 10.1038/s41392-025-02179-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: 11/01/2024] [Revised: 01/03/2025] [Accepted: 02/12/2025] [Indexed: 05/08/2025] Open
Abstract
Neurodegenerative diseases (e.g., Alzheimer's, Parkinson's, Huntington's disease, and Amyotrophic Lateral Sclerosis) are major health threats for the aging population and their prevalences continue to rise with the increasing of life expectancy. Although progress has been made, there is still a lack of effective cures to date, and an in-depth understanding of the molecular and cellular mechanisms of these neurodegenerative diseases is imperative for drug development. Protein phosphorylation, regulated by protein kinases and protein phosphatases, participates in most cellular events, whereas aberrant phosphorylation manifests as a main cause of diseases. As evidenced by pharmacological and pathological studies, protein kinases are proven to be promising therapeutic targets for various diseases, such as cancers, central nervous system disorders, and cardiovascular diseases. The mechanisms of protein phosphatases in pathophysiology have been extensively reviewed, but a systematic summary of the role of protein kinases in the nervous system is lacking. Here, we focus on the involvement of protein kinases in neurodegenerative diseases, by summarizing the current knowledge on the major kinases and related regulatory signal transduction pathways implicated in diseases. We further discuss the role and complexity of kinase-kinase networks in the pathogenesis of neurodegenerative diseases, illustrate the advances of clinical applications of protein kinase inhibitors or novel kinase-targeted therapeutic strategies (such as antisense oligonucleotides and gene therapy) for effective prevention and early intervention.
Collapse
Affiliation(s)
- Xiaolei Wu
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhangzhong Yang
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinjun Zou
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chuanzhou Li
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
4
|
Suzuki T, Fujii T, Kametani K, Li W, Tabuchi K. Tubulin and GTP Are Crucial Elements for Postsynaptic Density Construction and Aggregation. J Neurochem 2025; 169:e70085. [PMID: 40396438 DOI: 10.1111/jnc.70085] [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: 07/13/2024] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/22/2025]
Abstract
In our previous experiments on the postsynaptic density lattice (PSDL), which is thought to serve as the backbone structure for the PSD, we suggested that tubulin plays a fundamental role in the PSD structure at excitatory synapses. In this study, we further reveal an unrecognized characteristic of tubulin within the PSD. First, using electron microscopy, we identified an interaction between postsynaptic structures (PSDL and PSD) and polymerizing microtubules, which led to the binding of polymerizing microtubules to PSDL and PSD. In turn, this interaction induced changes in the microtubule morphology. These results support earlier findings suggesting that microtubules transiently intruding into the spine head can associate with PSDs, inducing structural changes in the PSD. Next, we observed that the structural integrity of both PSD and PSDL was compromised upon exposure to GTP and microtubule-affecting reagents. These findings reinforce the idea that tubulin is a crucial building block of the PSD architecture. Moreover, we found that PSD aggregation was enhanced following interactions with polymerizing tubulin and was disintegrated upon treatment with GTP and microtubule-affecting reagents. These results indicate that microtubules also play a key role in PSD aggregation in vitro. Collectively, our study highlights the involvement of tubulin in the construction, function (specifically its interaction with polymerizing microtubules), and aggregation of the PSD, which may impact both physiological and pathological conditions. Furthermore, our in vitro findings suggest that GTP can either destroy or induce the enlargement and reorganization of PSD structures, depending on its interaction with growing microtubules.
Collapse
Affiliation(s)
- Tatsuo Suzuki
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Toshihiro Fujii
- Shinshu University, Textile Science and Technology, Ueda, Japan
| | - Kiyokazu Kametani
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, Rakuno Gakuen University 582 Midori Cho, Ebetsu, Hokkaido, Japan
| | - Weidong Li
- Center for Brain Health and Brain Technology, Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research Shinshu University, Matsumoto, Japan
| | - Katsuhiko Tabuchi
- Department of Biological Sciences for Intractable Neurological Diseases, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research Shinshu University, Matsumoto, Japan
- Department of Molecular and Cellular Physiology, Shinshu University Academic Assembly, Institute of Medicine, Shinshu University Academic Assembly, Matsumoto, Japan
| |
Collapse
|
5
|
Buchholz S, Kabbani MAA, Bell‐Simons M, Kluge L, Cagmak C, Klimek J, Haag N, Iohan LC, Coulon A, Costa MR, Kilinc D, Zempel H. The tau isoform 1N4R confers vulnerability of MAPT knockout human iPSC-derived neurons to amyloid beta and phosphorylated tau-induced neuronal dysfunction. Alzheimers Dement 2025; 21:e14403. [PMID: 40019008 PMCID: PMC12089071 DOI: 10.1002/alz.14403] [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: 06/01/2024] [Revised: 10/01/2024] [Accepted: 10/21/2024] [Indexed: 03/01/2025]
Abstract
INTRODUCTION Human tau protein, composed of six brain-specific isoforms, is a major driver of Alzheimer's disease (AD). The role of its isoforms however remains unclear and human AD models are scarce. METHODS We generated human MAPT- (tau-) knockout (KO) induced pluripotent stem cells (iPSC) using CRISPR/Cas9, differentiated these into glutamatergic neurons, and assessed isoform-specific functions of tau in these neurons. We used omic- approaches, live-cell imaging, subcompartmental analysis, and lentivirus-based reintroduction of specific tau isoforms to investigate isoform-mediated neuronal dysfunction in an AD model. RESULTS Tau KO human iPSC-derived neurons showed decreased neurite outgrowth and axon initial segment length and, notably, resisted amyloid beta oligomer (AβO)-induced neuronal activity reduction. Introducing the 1N4R-tau isoform, but not other isoforms, confers AβO vulnerability and increases KxGS phosphorylation of tau, without altering neuronal activity or microtubule modifications. DISCUSSION While tau KO impacts neuronal development and activity, tau-KO also confers resistance against AβO insult. 1N4R-tau likely mediates AβO-induced and phosphorylated tau toxicity, representing a novel prime therapeutic target for AD. HIGHLIGHTS Tau knockout alters neurite growth and axon initial segment formation in human neurons. Tau isoforms show differential axonal localization in human neurons. Tau depletion protects against amyloid beta oligomer (AβO)-mediated neurotoxicity. 1N4R tau mediates AβO-induced toxicity in human neurons.
Collapse
Affiliation(s)
- Sarah Buchholz
- Institute of Human Genetics, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Mohamed Aghyad Al Kabbani
- Institute of Human Genetics, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Michael Bell‐Simons
- Institute of Human Genetics, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Lena Kluge
- Institute of Human Genetics, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Cagla Cagmak
- Institute of Human Genetics, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Jennifer Klimek
- Institute of Human Genetics, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Natja Haag
- Institute for Human Genetics and Genomic Medicine, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Lukas C. Iohan
- Université de LilleInserm, CHU Lille, Institut Pasteur de Lille, U1167‐RID‐AGE‐ Risk factors and molecular determinants of aging‐related diseasesLilleFrance
- Bioinformatics Multidisciplinary Environment (BIoME)Federal University of Rio Grande do Norte, Campus Universitário, Lagoa NovaNatalBrazil
| | - Audrey Coulon
- Université de LilleInserm, CHU Lille, Institut Pasteur de Lille, U1167‐RID‐AGE‐ Risk factors and molecular determinants of aging‐related diseasesLilleFrance
| | - Marcos R. Costa
- Université de LilleInserm, CHU Lille, Institut Pasteur de Lille, U1167‐RID‐AGE‐ Risk factors and molecular determinants of aging‐related diseasesLilleFrance
- Brain InstituteFederal University of Rio Grande do Norte, Campus Universitário, Lagoa NovaNatalBrazil
| | - Devrim Kilinc
- Université de LilleInserm, CHU Lille, Institut Pasteur de Lille, U1167‐RID‐AGE‐ Risk factors and molecular determinants of aging‐related diseasesLilleFrance
| | - Hans Zempel
- Institute of Human Genetics, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| |
Collapse
|
6
|
Folarin OR, Olopade FE, Gilbert TT, Ladagu AD, Pires Dos Santos PI, Mustapha OA, Kpasham LZ, Olopade JO, Outeiro TF. Chronic Vanadium Exposure Promotes Aggregation of Alpha-Synuclein, Tau and Amyloid Beta in Mouse Brain. J Neurochem 2025; 169:e70082. [PMID: 40377064 DOI: 10.1111/jnc.70082] [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: 11/01/2024] [Revised: 04/01/2025] [Accepted: 04/21/2025] [Indexed: 05/18/2025]
Abstract
The interaction of toxic environmental metals and metalloids with brain proteins and polypeptides can result in pathological aggregations and formation of toxic oligomers, which are key features of many neurodegenerative diseases. Occupational and environmental exposure to vanadium is connected to a neurological syndrome that includes psychiatric symptoms, cognitive decline, and neurodegeneration. In this study, we hypothesized that prolonged vanadium exposure may be a potential risk factor for Alzheimer's and Parkinson's diseases. A total of 72 male BALB/c mice, each 4 weeks' old, were used. Vanadium-treated groups received intraperitoneal injections of 3 mg/kg body weight of vanadium three times a week for 6, 12, or 18 months. The control group received sterile water while the withdrawal group were given vanadium injection for 3 months, followed by withdrawal of the metal, but treatment with sterile water only, and were sacrificed at 3-, 9-, or 15-months post vanadium exposure. Sagittal sections of brain paraffin-embedded tissue were prepared and analyzed using immunofluorescence to study the immunoreactivity and cellular localization of α-synuclein (α-syn), amyloid-β (Aβ), and tau proteins. Our findings revealed pathological aggregation of these proteins in the frontoparietal cortices and the dorsal CA1 and CA3 regions. Double immunolabeling with glial cells and neurons showed neuronal degeneration, functional gliosis, and activation of astrocytes and microglia at sites of α-synuclein immunoreactivity. We observed increased immunoreactivity of phosphorylated nuclei tau both in the parietal cortices and corpus callosum white matter while we observed intraneuronal accumulation of Aβ deposits in the cortex and dorsal hippocampal regions in vanadium treated brains. These cellular changes and proteinopathies, although persisting, were significantly attenuated after vanadium withdrawal. Our findings show that prolonged vanadium exposure promotes abnormal accumulation of neurodegeneration-associated proteins (α-syn, Tau, and Aβ) in the brain, which is further exacerbated by aging in the context of extended exposure to the metal.
Collapse
Affiliation(s)
- O R Folarin
- Department of Biomedical Laboratory Science, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - F E Olopade
- Department of Anatomy, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - T T Gilbert
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - A D Ladagu
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - P I Pires Dos Santos
- Department of Experimental Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - O A Mustapha
- Department of Veterinary Anatomy, College of Veterinary Medicine, Federal University of Abeokuta, Abeokuta, Nigeria
| | - L Z Kpasham
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - J O Olopade
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - T F Outeiro
- Department of Experimental Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
7
|
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
|
8
|
Ariawan D, van der Hoven J, Morey N, Pushpitha K, Genoud S, Stefen H, Veltman S, Przybyla M, Deng Y, Fath T, Tietz O, van Eersel J, Ittner LM. Engineered Cyclotide Blocks Neuronal Excitotoxicity. J Med Chem 2025; 68:5211-5221. [PMID: 39630561 DOI: 10.1021/acs.jmedchem.4c01710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Cyclotides are naturally occurring cyclic peptides with three disulfide bonds, offering remarkable stability. In neurological disorders, the formation of a complex between postsynaptic density protein 95 and NMDA receptors (NMDARs) can lead to neuronal cell death. In this study, we modified the MCoTI-II cyclotide backbone with polyarginines for enhanced intracellular delivery and grafted a 9-amino acid PSD-95-NMDAR inhibitor sequence, NR2B9c, into loop 6. We found that incorporating polyarginines into the cyclotide backbone significantly improved uptake into neuronal cells. Primary neurons treated with the NR2B9c cyclotide (c5R-NR2B9c) prevented cell death in response to high concentrations of N-methyl-d-aspartate (NMDA), demonstrating protection from excitotoxicity. Administration of c5R-NR2B9c in a chemically induced seizure model in mice resulted in increased survival and reduced seizure severity. Overall, we show that modifying cyclotides with a polyarginine backbone can enhance the delivery of therapeutic peptides into neuronal cells, which can be utilized to administer therapeutic peptides for the protection of neuronal cells from excitotoxicity.
Collapse
Affiliation(s)
- Daryl Ariawan
- Dementia Research Centre, Macquarie Medical School, Macquarie University 2109 Sydney, Australia
| | - Julia van der Hoven
- Dementia Research Centre, Macquarie Medical School, Macquarie University 2109 Sydney, Australia
| | - Nicolle Morey
- Dementia Research Centre, Macquarie Medical School, Macquarie University 2109 Sydney, Australia
| | - Kanishka Pushpitha
- Dementia Research Centre, Macquarie Medical School, Macquarie University 2109 Sydney, Australia
| | - Sian Genoud
- Dementia Research Centre, Macquarie Medical School, Macquarie University 2109 Sydney, Australia
| | - Holly Stefen
- Dementia Research Centre, Macquarie Medical School, Macquarie University 2109 Sydney, Australia
| | - Sanne Veltman
- Dementia Research Centre, Macquarie Medical School, Macquarie University 2109 Sydney, Australia
| | - Magdalena Przybyla
- Dementia Research Centre, Macquarie Medical School, Macquarie University 2109 Sydney, Australia
| | - Yuanyuan Deng
- Dementia Research Centre, Macquarie Medical School, Macquarie University 2109 Sydney, Australia
| | - Thomas Fath
- Dementia Research Centre, Macquarie Medical School, Macquarie University 2109 Sydney, Australia
| | - Ole Tietz
- Dementia Research Centre, Macquarie Medical School, Macquarie University 2109 Sydney, Australia
| | - Janet van Eersel
- Dementia Research Centre, Macquarie Medical School, Macquarie University 2109 Sydney, Australia
| | - Lars Matthias Ittner
- Dementia Research Centre, Macquarie Medical School, Macquarie University 2109 Sydney, Australia
| |
Collapse
|
9
|
Theodorou A, Tsantzali I, Stefanou MI, Sacco S, Katsanos AH, Shoamanesh A, Karapanayiotides T, Koutroulou I, Stamati P, Werring DJ, Cordonnier C, Palaiodimou L, Zompola C, Boviatsis E, Stavrinou L, Frantzeskaki F, Steiner T, Alexandrov AV, Paraskevas GP, Tsivgoulis G. CSF and plasma biomarkers in cerebral amyloid angiopathy: A single-center study and a systematic review/meta-analysis. Eur Stroke J 2025; 10:278-288. [PMID: 38869035 PMCID: PMC11569450 DOI: 10.1177/23969873241260538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/14/2024] [Indexed: 06/14/2024] Open
Abstract
INTRODUCTION There are limited data regarding cerebrospinal fluid (CSF) and plasma biomarkers among patients with Cerebral Amyloid Angiopathy (CAA). We sought to investigate the levels of four biomarkers [β-amyloids (Aβ42 and Aβ40), total tau (tau) and phosphorylated tau (p-tau)] in CAA patients compared to healthy controls (HC) and patients with Alzheimer Disease (AD). PATIENTS AND METHODS A systematic review and meta-analysis of published studies, including also a 5 year single-center cohort study, with available data on CSF and plasma biomarkers in symptomatic sporadic CAA versus HC and AD was conducted. Biomarkers' comparisons were investigated using random-effects models based on the ratio of mean (RoM) biomarker concentrations. RoM < 1 and RoM > 1 indicate lower and higher biomarker concentration in CAA compared to another population, respectively. RESULTS We identified nine cohorts, comprising 327 CAA patients (mean age: 71 ± 5 years; women: 45%) versus 336 HC (mean age: 65 ± 5 years; women: 45%) and 384 AD patients (mean age: 68 ± 3 years; women: 53%) with available data on CSF biomarkers. CSF Aβ42 levels [RoM: 0.47; 95% CI: 0.36-0.62; p < 0.0001], Aβ40 levels [RoM: 0.70; 95% CI: 0.63-0.79; p < 0.0001] and the ratio Aβ42/Aβ40 [RoM: 0.62; 95% CI: 0.39-0.98; p = 0.0438] differentiated CAA from HC. CSF Aβ40 levels [RoM: 0.73; 95% CI: 0.64-0.83; p = 0.0003] differentiated CAA from AD. CSF tau and p-tau levels differentiated CAA from HC [RoM: 1.71; 95% CI: 1.41-2.09; p = 0.0002 and RoM: 1.44; 95% CI: 1.20-1.73; p = 0.0014, respectively] and from AD [RoM: 0.65; 95% CI: 0.58-0.72; p < 0.0001 and RoM: 0.64; 95% CI: 0.57-0.71; p < 0.0001, respectively]. Plasma Aβ42 [RoM: 1.14; 95% CI: 0.89-1.45; p = 0.2079] and Aβ40 [RoM: 1.07; 95% CI: 0.91-1.25; p = 0.3306] levels were comparable between CAA and HC. CONCLUSIONS CAA is characterized by a distinct CSF biomarker pattern compared to HC and AD. CSF Aβ40 levels are lower in CAA compared to HC and AD, while tau and p-tau levels are higher in CAA compared to HC, but lower in comparison to AD patients.
Collapse
Affiliation(s)
- Aikaterini Theodorou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Tsantzali
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria-Ioanna Stefanou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Simona Sacco
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, Via Vetoio, Italy
| | - Aristeidis H Katsanos
- Division of Neurology, McMaster University/Population Health Research Institute, Hamilton, Canada
| | - Ashkan Shoamanesh
- Division of Neurology, McMaster University/Population Health Research Institute, Hamilton, Canada
| | - Theodoros Karapanayiotides
- Second Department of Neurology, Aristotle University of Thessaloniki, School of Medicine, AHEPA University Hospital, Thessaloniki, Greece
| | - Ioanna Koutroulou
- Second Department of Neurology, Aristotle University of Thessaloniki, School of Medicine, AHEPA University Hospital, Thessaloniki, Greece
| | - Polyxeni Stamati
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Biopolis, Mezourlo Hill, Larissa, Greece
| | - David J Werring
- Stroke Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Charlotte Cordonnier
- University Lille, Inserm, CHU Lille, U1172, LilNCog, Lille Neuroscience and Cognition, France
| | - Lina Palaiodimou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Zompola
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Boviatsis
- Second Department of Neurosurgery, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Lampis Stavrinou
- Second Department of Neurosurgery, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Frantzeska Frantzeskaki
- Second Critical Care Department, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Thorsten Steiner
- Departments of Neurology, Klinikum Frankfurt Höchst, Frankfurt and Heidelberg University Hospital, Heidelberg, Germany
| | - Andrei V Alexandrov
- Department of Neurology, University of Arizona, Banner University Medical Center, Phoenix
| | - Georgios P Paraskevas
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios Tsivgoulis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Neurology, University of Tennessee Health Science Center, Memphis
| |
Collapse
|
10
|
Li T, Liu R, He Y, Zhang B, Rui X, Yang X, Wang J, Zeng J, Li G, Li X, Liu G. Overexpression of TECPR1 improved cognitive function of P301S-tau mice via activation of autophagy in the early and late process. Aging Cell 2025; 24:e14404. [PMID: 39511758 PMCID: PMC11896361 DOI: 10.1111/acel.14404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/17/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024] Open
Abstract
Autophagy disorders in AD patients and animal models were well known, however, the effect of P301S-tau on autophagy is not clear. Here, we found that autophagy related protein Tectonin Beta-Propeller Repeat-Containing Protein 1 (TECPR1) decreased in the hippocampus of P301S-tau transgenic mice by proteomics, which was proved in vivo and in vitro, and P301S-tau induced autophagic deficits in early and late process. TECPR1 overexpression attenuated P301S-tau induced autophagy defects via promoting autophagosome generation and autophagosome and lysosomes fusion. We also found that TECPR1 overexpression ameliorated the behavior disorders of P301S-tau mice with promoting tau degradation, improving synaptic plasticity and neuron loss. Lastly, CQ or 3-MA treatment reversed TECPR1 induced improvement effects on autophagic and cognitive disorders, further proved that, TECPR1 activated the early and late process of autophagy to ameliorate the cognition of P301S-tau mice. Our data suggest that TECPR1 is a potential therapy target for AD.
Collapse
Affiliation(s)
- Ting Li
- Department of PathologyRenmin Hospital of Wuhan UniversityWuhanChina
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ruijuan Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ye He
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Bingge Zhang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xuexiang Rui
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Subject of Modern ToxicologyShenzhen Center for Disease Control and PreventionShenzhenChina
| | - Jian‐Zhi Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Juan Zeng
- Department of PathologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Gang Li
- Department of Neurology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiao Li
- Department of PathologyWuhan No. 1 HospitalWuhanChina
| | - Gong‐Ping Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Subject of Modern ToxicologyShenzhen Center for Disease Control and PreventionShenzhenChina
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantongJiangsuChina
| |
Collapse
|
11
|
Cheng Q, Fan Y, Zhang P, Liu H, Han J, Yu Q, Wang X, Wu S, Lu Z. Biomarkers of synaptic degeneration in Alzheimer's disease. Ageing Res Rev 2025; 104:102642. [PMID: 39701184 DOI: 10.1016/j.arr.2024.102642] [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: 03/27/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 12/21/2024]
Abstract
Synapse has been considered a critical neuronal structure in the procession of Alzheimer's disease (AD), attacked by two pathological molecule aggregates (amyloid-β and phosphorylated tau) in the brain, disturbing synaptic homeostasis before disease manifestation and subsequently causing synaptic degeneration. Recently, evidence has emerged indicating that soluble oligomeric amyloid-β (AβO) and tau exert direct toxicity on synapses, causing synaptic damage. Synaptic degeneration is closely linked to cognitive decline in AD, even in the asymptomatic stages of AD. Therefore, the identification of novel, specific, and sensitive biomarkers involved in synaptic degeneration holds significant promise for early diagnosis of AD, reducing synaptic degeneration and loss, and controlling the progression of AD. Currently, a range of biomarkers in cerebrospinal fluid (CSF), such as synaptosome-associated protein 25 (SNAP-25), synaptotagmin-1, growth-associated protein-43 (GAP-43), and neurogranin (Ng), along with functional brain imaging techniques, can detect variations in synaptic density, offering high sensitivity and specificity for AD diagnosis. However, these methods face challenges, including invasiveness, high cost, and limited accessibility. In contrast, biomarkers found in blood or urine provide a minimally invasive, cost-effective, and more accessible alternative to traditional diagnostic methods. Notably, neuron-derived exosomes in blood, which contain synaptic proteins, show variations in concentration that can serve as indicators of synaptic injury, providing an additional, less invasive approach to AD diagnosis and monitoring.
Collapse
Affiliation(s)
- Qian Cheng
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Yiou Fan
- Laboratory and Quality Management Department, Centers for Disease Control and Prevention of Shandong, Jinan, Shandong, China
| | - Pengfei Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Huan Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Jialin Han
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Qian Yu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xueying Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Shuang Wu
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Zhiming Lu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
12
|
Eisenbaum M, Bachmeier C. Contribution of astrocytes to the neurovascular elimination of tau. Neural Regen Res 2024; 19:2559-2560. [PMID: 38808980 PMCID: PMC11168527 DOI: 10.4103/nrr.nrr-d-23-01705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/12/2024] [Accepted: 01/25/2024] [Indexed: 05/30/2024] Open
Affiliation(s)
| | - Corbin Bachmeier
- The Roskamp Institute, Sarasota, FL, USA
- Bay Pines VA Healthcare System, Bay Pines, FL, USA
| |
Collapse
|
13
|
Ferjančič Benetik S, Knez D, Obreza A, Košak U, Gobec S. Dual inhibition of butyrylcholinesterase and p38α mitogen-activated protein kinase: A new approach for the treatment of Alzheimer's disease. Pharmacol Ther 2024; 264:108748. [PMID: 39521443 DOI: 10.1016/j.pharmthera.2024.108748] [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: 07/02/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
The simultaneous targeting of neuroinflammation and cholinergic hypofunction, the key pathological changes in Alzheimer's disease (AD), is not addressed by drugs currently in clinical trials, highlighting a critical therapeutic gap. We propose that dual-acting small molecules that inhibit butyrylcholinesterase (BChE) and mitogen-activated protein kinase p38α (p38α MAPK) represent a novel strategy to combat AD. This hypothesis is supported by cellular and animal studies as well as in silico modelling showing that it is possible to act simultaneously on both enzymes. Amyloid beta (Aβ) plaques trigger a pro-inflammatory microglial response that overactivates p38α MAPK, leading to increased Aβ synthesis, tau hyperphosphorylation, and altered synaptic plasticity. Overactivated microglia exacerbate neuroinflammation and cholinergic degeneration, ultimately leading to cognitive impairment. Structural similarities between the binding sites of BChE and p38α MAPK provide a promising basis for the development of dual inhibitors that could alleviate AD symptoms and address the underlying pathology.
Collapse
Affiliation(s)
| | - Damijan Knez
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Aleš Obreza
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Urban Košak
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Stanislav Gobec
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia.
| |
Collapse
|
14
|
Paidlewar M, Kumari S, Dhapola R, Sharma P, HariKrishnaReddy D. Unveiling the role of astrogliosis in Alzheimer's disease Pathology: Insights into mechanisms and therapeutic approaches. Int Immunopharmacol 2024; 141:112940. [PMID: 39154532 DOI: 10.1016/j.intimp.2024.112940] [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: 05/09/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024]
Abstract
Alzheimer's disease (AD) is one of the most debilitating age-related disorders that affect people globally. It impacts social and cognitive behavior of the individual and is characterized by phosphorylated tau and Aβ accumulation. Astrocytesmaintain a quiescent, anti-inflammatory state on anatomical level, expressing few cytokines and exhibit phagocytic activity to remove misfolded proteins. But in AD, in response to specific stimuli, astrocytes overstimulate their phagocytic character with overexpressing cytokine gene modules. Upon interaction with generated Aβ and neurofibrillary tangle, astrocytes that are continuously activated release a large number of inflammatory cytokines. This cytokine storm leads to neuroinflammation which is also one of the recognizable features of AD. Astrogliosis eventually promotes cholinergic dysfunction, calcium imbalance, oxidative stress and excitotoxicity. Furthermore, C5aR1, Lcn2/, BDNF/TrkB and PPARα/TFEB signaling dysregulation has a major impact on the disease progression. This review clarifies numerous ways that lead to astrogliosis, which is stimulated by a variety of processes that exacerbate AD pathology and make it a suitable target for AD treatment. Drugs under clinical and preclinical investigations that target several pathways managing astrogliosis and are efficacious in ameliorating the pathology of the disease are also included in this study. D-ALA2GIP, TRAM-34, Genistein, L-serine, MW150 and XPro1595 are examples of few drugs targeting astrogliosis. Therefore, this study may aid in the development of a potent therapeutic agent for ameliorating astrogliosis mediated AD progression.
Collapse
Affiliation(s)
- Mohit Paidlewar
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda-151401, Punjab, India.
| |
Collapse
|
15
|
Wlodarczyk J, Bhattacharyya R, Dore K, Ho GPH, Martin DDO, Mejias R, Hochrainer K. Altered Protein Palmitoylation as Disease Mechanism in Neurodegenerative Disorders. J Neurosci 2024; 44:e1225242024. [PMID: 39358031 PMCID: PMC11450541 DOI: 10.1523/jneurosci.1225-24.2024] [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: 06/20/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 10/04/2024] Open
Abstract
Palmitoylation, a lipid-based posttranslational protein modification, plays a crucial role in regulating various aspects of neuronal function through altering protein membrane-targeting, stabilities, and protein-protein interaction profiles. Disruption of palmitoylation has recently garnered attention as disease mechanism in neurodegeneration. Many proteins implicated in neurodegenerative diseases and associated neuronal dysfunction, including but not limited to amyloid precursor protein, β-secretase (BACE1), postsynaptic density protein 95, Fyn, synaptotagmin-11, mutant huntingtin, and mutant superoxide dismutase 1, undergo palmitoylation, and recent evidence suggests that altered palmitoylation contributes to the pathological characteristics of these proteins and associated disruption of cellular processes. In addition, dysfunction of enzymes that catalyze palmitoylation and depalmitoylation has been connected to the development of neurological disorders. This review highlights some of the latest advances in our understanding of palmitoylation regulation in neurodegenerative diseases and explores potential therapeutic implications.
Collapse
Affiliation(s)
- Jakub Wlodarczyk
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw 02-093, Poland
| | - Raja Bhattacharyya
- Genetics and Aging Research Unit, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Kim Dore
- Department of Neurosciences, Center for Neural Circuits and Behavior, UCSD, La Jolla, California 92093
| | - Gary P H Ho
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Dale D O Martin
- Department of Biology, Faculty of Science, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Rebeca Mejias
- Department of Physiology, School of Biology, Universidad de Sevilla, Seville, 41012 Spain
- Instituto de Investigaciones Biomédicas de Sevilla, IBIS/Universidad de Sevilla/Hospital Universitario Virgen del Rocío/Junta de Andalucía/CSIC, Seville 41013, Spain
| | - Karin Hochrainer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| |
Collapse
|
16
|
Zhang X, Wang J, Zhang Z, Ye K. Tau in neurodegenerative diseases: molecular mechanisms, biomarkers, and therapeutic strategies. Transl Neurodegener 2024; 13:40. [PMID: 39107835 PMCID: PMC11302116 DOI: 10.1186/s40035-024-00429-6] [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: 11/22/2023] [Accepted: 07/05/2024] [Indexed: 09/14/2024] Open
Abstract
The deposition of abnormal tau protein is characteristic of Alzheimer's disease (AD) and a class of neurodegenerative diseases called tauopathies. Physiologically, tau maintains an intrinsically disordered structure and plays diverse roles in neurons. Pathologically, tau undergoes abnormal post-translational modifications and forms oligomers or fibrous aggregates in tauopathies. In this review, we briefly introduce several tauopathies and discuss the mechanisms mediating tau aggregation and propagation. We also describe the toxicity of tau pathology. Finally, we explore the early diagnostic biomarkers and treatments targeting tau. Although some encouraging results have been achieved in animal experiments and preclinical studies, there is still no cure for tauopathies. More in-depth basic and clinical research on the pathogenesis of tauopathies is necessary.
Collapse
Affiliation(s)
- Xingyu Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jiangyu Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430000, China.
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
17
|
Asada-Utsugi M, Urushitani M. Tau beyond Tangles: DNA Damage Response and Cytoskeletal Protein Crosstalk on Neurodegeneration. Int J Mol Sci 2024; 25:7906. [PMID: 39063148 PMCID: PMC11277103 DOI: 10.3390/ijms25147906] [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: 06/05/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Neurons in the brain are continuously exposed to various sources of DNA damage. Although the mechanisms of DNA damage repair in mitotic cells have been extensively characterized, the repair pathways in post-mitotic neurons are still largely elusive. Moreover, inaccurate repair can result in deleterious mutations, including deletions, insertions, and chromosomal translocations, ultimately compromising genomic stability. Since neurons are terminally differentiated cells, they cannot employ homologous recombination (HR) for double-strand break (DSB) repair, suggesting the existence of neuron-specific repair mechanisms. Our research has centered on the microtubule-associated protein tau (MAPT), a crucial pathological protein implicated in neurodegenerative diseases, and its interplay with neurons' DNA damage response (DDR). This review aims to provide an updated synthesis of the current understanding of the complex interplay between DDR and cytoskeletal proteins in neurons, with a particular focus on the role of tau in neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Makoto Urushitani
- Department of Neurology, Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan;
| |
Collapse
|
18
|
Harvey J. Novel Leptin-Based Therapeutic Strategies to Limit Synaptic Dysfunction in Alzheimer's Disease. Int J Mol Sci 2024; 25:7352. [PMID: 39000459 PMCID: PMC11242278 DOI: 10.3390/ijms25137352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Accumulation of hyper-phosphorylated tau and amyloid beta (Aβ) are key pathological hallmarks of Alzheimer's disease (AD). Increasing evidence indicates that in the early pre-clinical stages of AD, phosphorylation and build-up of tau drives impairments in hippocampal excitatory synaptic function, which ultimately leads to cognitive deficits. Consequently, limiting tau-related synaptic abnormalities may have beneficial effects in AD. There is now significant evidence that the hippocampus is an important brain target for the endocrine hormone leptin and that leptin has pro-cognitive properties, as activation of synaptic leptin receptors markedly influences higher cognitive processes including learning and memory. Clinical studies have identified a link between the circulating leptin levels and the risk of AD, such that AD risk is elevated when leptin levels fall outwith the physiological range. This has fuelled interest in targeting the leptin system therapeutically. Accumulating evidence supports this possibility, as numerous studies have shown that leptin has protective effects in a variety of models of AD. Recent findings have demonstrated that leptin has beneficial effects in the preclinical stages of AD, as leptin prevents the early synaptic impairments driven by tau protein and amyloid β. Here we review recent findings that implicate the leptin system as a potential novel therapeutic target in AD.
Collapse
Affiliation(s)
- Jenni Harvey
- Department of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| |
Collapse
|
19
|
Palihati N, Tang Y, Yin Y, Yu D, Liu G, Quan Z, Ni J, Yan Y, Qing H. Clusterin is a Potential Therapeutic Target in Alzheimer's Disease. Mol Neurobiol 2024; 61:3836-3850. [PMID: 38017342 DOI: 10.1007/s12035-023-03801-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/10/2023] [Indexed: 11/30/2023]
Abstract
In recent years, Clusterin, a glycosylated protein with multiple biological functions, has attracted extensive research attention. It is closely associated with the physiological and pathological states within the organism. Particularly in Alzheimer's disease (AD) research, Clusterin plays a significant role in the disease's occurrence and progression. Numerous studies have demonstrated a close association between Clusterin and AD. Firstly, the expression level of Clusterin in the brain tissue of AD patients is closely related to pathological progression. Secondly, Clusterin is involved in the deposition and formation of β-amyloid, which is a crucial process in AD development. Furthermore, Clusterin may affect the pathogenesis of AD through mechanisms such as regulating inflammation, controlling cell apoptosis, and clearing pathological proteins. Therefore, further research on the relationship between Clusterin and AD will contribute to a deeper understanding of the etiology of this neurodegenerative disease and provide a theoretical basis for developing early diagnostic and therapeutic strategies for AD. This also makes Clusterin one of the research focuses as a potential biomarker for AD diagnosis and treatment monitoring.
Collapse
Affiliation(s)
- Nazhakaiti Palihati
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Yuanhong Tang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Yajuan Yin
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Ding Yu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Yan Yan
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
- Department of Biology, Shenzhen MSU-BIT University, Shenzhen, 518172, China.
| |
Collapse
|
20
|
Iram F, Shahid M, Ansari J, Ashraf GM, Hassan MI, Islam A. Navigating the Maze of Alzheimer's disease by exploring BACE1: Discovery, current scenario, and future prospects. Ageing Res Rev 2024; 98:102342. [PMID: 38762102 DOI: 10.1016/j.arr.2024.102342] [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: 03/07/2024] [Revised: 05/04/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
Alzheimer's disease (AD) is a chronic neurological condition that has become a leading cause of cognitive decline in elder individuals. Hardly any effective medication has been developed to halt the progression of AD due to the disease's complexity. Several theories have been put forward to clarify the mechanisms underlying AD etiology. The identification of amyloid plaques as a hallmark of AD has sparked the development of numerous drugs targeting the players involved in the amyloidogenic pathway, such as the β-site of amyloid precursor protein cleavage enzyme 1 (BACE1) blockers. Over the last ten years, preclinical and early experimental research has led several pharmaceutical companies to prioritize producing BACE1 inhibitors. Despite all these efforts, earlier discovered inhibitors were discontinued in consideration of another second-generation small molecules and recent BACE1 antagonists failed in the final stages of clinical trials because of the complications associated either with toxicity or effectiveness. In addition to discussing the difficulties associated with development of BACE1 inhibitors, this review aims to provide an overview of BACE1 and offer perspectives on the causes behind the failure of five recent BACE1 inhibitors, that would be beneficial for choosing effective treatment approaches in the future.
Collapse
Affiliation(s)
- Faiza Iram
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Mohammad Shahid
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Jaoud Ansari
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ghulam Md Ashraf
- University of Sharjah, College of Health Sciences, and Research Institute for Medical and Health Sciences, Department of Medical Laboratory Sciences, Sharjah 27272, United Arab Emirates
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
21
|
Wang S, Yu X, Cheng L, Ren W, Wen G, Wu X, Lou H, Ren X, Lu L, Hermenean A, Yao J, Li B, Lu Y, Wu X. Dexmedetomidine improves the circulatory dysfunction of the glymphatic system induced by sevoflurane through the PI3K/AKT/ΔFosB/AQP4 pathway in young mice. Cell Death Dis 2024; 15:448. [PMID: 38918408 PMCID: PMC11199640 DOI: 10.1038/s41419-024-06845-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
Multiple sevoflurane exposures may damage the developing brain. The neuroprotective function of dexmedetomidine has been widely confirmed in animal experiments and human studies. However, the effect of dexmedetomidine on the glymphatic system has not been clearly studied. We hypothesized that dexmedetomidine could alleviate sevoflurane-induced circulatory dysfunction of the glymphatic system in young mice. Six-day-old C57BL/6 mice were exposed to 3% sevoflurane for 2 h daily, continuously for 3 days. Intraperitoneal injection of either normal saline or dexmedetomidine was administered before every anaesthesia. Meanwhile the circulatory function of glymphatic system was detected by tracer injection at P8 and P32. On P30-P32, behavior tests including open field test, novel object recognition test, and Y-maze test were conducted. Primary astrocyte cultures were established and treated with the PI3K activator 740Y-P, dexmedetomidine, and small interfering RNA (siRNA) to silence ΔFosB. We propose for the first time that multiple exposure to sevoflurane induces circulatory dysfunction of the glymphatic system in young mice. Dexmedetomidine improves the circulatory capacity of the glymphatic system in young mice following repeated exposure to sevoflurane through the PI3K/AKT/ΔFosB/AQP4 signaling pathway, and enhances their long-term learning and working memory abilities.
Collapse
Affiliation(s)
- Shuying Wang
- School of Forensic Medicine, China Medical University, Shenyang, China
- Department of Anaesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Xiaojin Yu
- School of Forensic Medicine, China Medical University, Shenyang, China
- Department of Anaesthesiology, Affiliated Shengjing Hospital of China Medical University, Shenyang, China
| | - Lili Cheng
- School of Forensic Medicine, China Medical University, Shenyang, China
- Department of Anaesthesiology, Affiliated Shengjing Hospital of China Medical University, Shenyang, China
| | - Weishu Ren
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Gehua Wen
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xue Wu
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Haoyang Lou
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xinghua Ren
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Lei Lu
- Department of pediatrics Neonatology, University of Chicago, Chicago, IL, 60615, USA
| | - Anca Hermenean
- Faculty of Medicine, Vasile Goldis Western University of Arad, Arad, Romania
| | - Jun Yao
- School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Center of Forensic Investigation, Shenyang, China
| | - Baoman Li
- School of Forensic Medicine, China Medical University, Shenyang, China.
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China.
- China Medical University Center of Forensic Investigation, Shenyang, China.
| | - Yan Lu
- Key Laboratory of Health Ministry in Congenital Malformation, Affiliated Shengjing Hospital of China Medical University, Shenyang, China.
| | - Xu Wu
- School of Forensic Medicine, China Medical University, Shenyang, China.
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China.
- China Medical University Center of Forensic Investigation, Shenyang, China.
| |
Collapse
|
22
|
Hole KL, Zhu B, Huggon L, Brown JT, Mason JM, Williams RJ. Tau P301L disengages from the proteosome core complex and neurogranin coincident with enhanced neuronal network excitability. Cell Death Dis 2024; 15:429. [PMID: 38890273 PMCID: PMC11189525 DOI: 10.1038/s41419-024-06815-2] [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: 10/30/2023] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024]
Abstract
Tauopathies are characterised by the pathological accumulation of misfolded tau. The emerging view is that toxic tau species drive synaptic dysfunction and potentially tau propagation before measurable neurodegeneration is evident, but the underlying molecular events are not well defined. Human non-mutated 0N4R tau (tauWT) and P301L mutant 0N4R tau (tauP301L) were expressed in mouse primary cortical neurons using adeno-associated viruses to monitor early molecular changes and synaptic function before the onset of neuronal loss. In this model tauP301L was differentially phosphorylated relative to tauwt with a notable increase in phosphorylation at ser262. Affinity purification - mass spectrometry combined with tandem mass tagging was used to quantitatively compare the tauWT and tauP301L interactomes. This revealed an enrichment of tauP301L with ribosomal proteins but a decreased interaction with the proteasome core complex and reduced tauP301L degradation. Differences in the interaction of tauP301L with members of a key synaptic calcium-calmodulin signalling pathway were also identified, most notably, increased association with CaMKII but reduced association with calcineurin and the candidate AD biomarker neurogranin. Decreased association of neurogranin to tauP301L corresponded with the appearance of enhanced levels of extracellular neurogranin suggestive of potential release or leakage from synapses. Finally, analysis of neuronal network activity using micro-electrode arrays showed that overexpression of tauP301L promoted basal hyperexcitability coincident with these changes in the tau interactome and implicating tau in specific early alterations in synaptic function.
Collapse
Affiliation(s)
- Katriona L Hole
- Department of Life Sciences, University of Bath, Bath, UK
- The Francis Crick Institute, London, UK
| | - Bangfu Zhu
- Department of Life Sciences, University of Bath, Bath, UK
| | - Laura Huggon
- Department of Life Sciences, University of Bath, Bath, UK
- UK Dementia Research Institute at King's College London, London, UK
| | - Jon T Brown
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| | - Jody M Mason
- Department of Life Sciences, University of Bath, Bath, UK
| | | |
Collapse
|
23
|
Yang J, Zhi W, Wang L. Role of Tau Protein in Neurodegenerative Diseases and Development of Its Targeted Drugs: A Literature Review. Molecules 2024; 29:2812. [PMID: 38930877 PMCID: PMC11206543 DOI: 10.3390/molecules29122812] [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: 04/26/2024] [Revised: 06/07/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Tau protein is a microtubule-associated protein that is widely distributed in the central nervous system and maintains and regulates neuronal morphology and function. Tau protein aggregates abnormally and forms neurofibrillary tangles in neurodegenerative diseases, disrupting the structure and function of neurons and leading to neuronal death, which triggers the initiation and progression of neurological disorders. The aggregation of tau protein in neurodegenerative diseases is associated with post-translational modifications, which may affect the hydrophilicity, spatial conformation, and stability of tau protein, promoting tau protein aggregation and the formation of neurofibrillary tangles. Therefore, studying the role of tau protein in neurodegenerative diseases and the mechanism of aberrant aggregation is important for understanding the mechanism of neurodegenerative diseases and finding therapeutic approaches. This review describes the possible mechanisms by which tau protein promotes neurodegenerative diseases, the post-translational modifications of tau protein and associated influencing factors, and the current status of drug discovery and development related to tau protein, which may contribute to the development of new therapeutic approaches to alleviate or treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Jiakai Yang
- Graduate Collaborative Training Base of Academy of Military Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China;
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Weijia Zhi
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Lifeng Wang
- Graduate Collaborative Training Base of Academy of Military Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China;
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| |
Collapse
|
24
|
Martin SC, Joyce KK, Lord JS, Harper KM, Nikolova VD, Cohen TJ, Moy SS, Diering GH. Sleep Disruption Precedes Forebrain Synaptic Tau Burden and Contributes to Cognitive Decline in a Sex-Dependent Manner in the P301S Tau Transgenic Mouse Model. eNeuro 2024; 11:ENEURO.0004-24.2024. [PMID: 38858068 PMCID: PMC11209651 DOI: 10.1523/eneuro.0004-24.2024] [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: 01/02/2024] [Revised: 05/02/2024] [Accepted: 05/14/2024] [Indexed: 06/12/2024] Open
Abstract
Sleep disruption and impaired synaptic processes are common features in neurodegenerative diseases, including Alzheimer's disease (AD). Hyperphosphorylated Tau is known to accumulate at neuronal synapses in AD, contributing to synapse dysfunction. However, it remains unclear how sleep disruption and synapse pathology interact to contribute to cognitive decline. Here, we examined sex-specific onset and consequences of sleep loss in AD/tauopathy model PS19 mice. Using a piezoelectric home-cage monitoring system, we showed PS19 mice exhibited early-onset and progressive hyperarousal, a selective dark-phase sleep disruption, apparent at 3 months in females and 6 months in males. Using the Morris water maze test, we report that chronic sleep disruption (CSD) accelerated the onset of decline of hippocampal spatial memory in PS19 males only. Hyperarousal occurs well in advance of robust forebrain synaptic Tau burden that becomes apparent at 6-9 months. To determine whether a causal link exists between sleep disruption and synaptic Tau hyperphosphorylation, we examined the correlation between sleep behavior and synaptic Tau, or exposed mice to acute or chronic sleep disruption at 6 months. While we confirm that sleep disruption is a driver of Tau hyperphosphorylation in neurons of the locus ceruleus, we were unable to show any causal link between sleep loss and Tau burden in forebrain synapses. Despite the finding that hyperarousal appears earlier in females, female cognition was resilient to the effects of sleep disruption. We conclude sleep disruption interacts with the synaptic Tau burden to accelerate the onset of cognitive decline with greater vulnerability in males.
Collapse
Affiliation(s)
- Shenée C Martin
- Departments of Cell Biology and Physiology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Kathryn K Joyce
- Departments of Cell Biology and Physiology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Julia S Lord
- Departments of Cell Biology and Physiology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Kathryn M Harper
- Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Viktoriya D Nikolova
- Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Todd J Cohen
- Neurology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Sheryl S Moy
- Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
- Carolina Institute for Developmental Disabilities, Carrboro, North Carolina 27510
| | - Graham H Diering
- Departments of Cell Biology and Physiology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
- Carolina Institute for Developmental Disabilities, Carrboro, North Carolina 27510
| |
Collapse
|
25
|
Chu D, Yang X, Wang J, Zhou Y, Gu JH, Miao J, Wu F, Liu F. Tau truncation in the pathogenesis of Alzheimer's disease: a narrative review. Neural Regen Res 2024; 19:1221-1232. [PMID: 37905868 PMCID: PMC11467920 DOI: 10.4103/1673-5374.385853] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/07/2023] [Accepted: 07/25/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Alzheimer's disease is characterized by two major neuropathological hallmarks-the extracellular β-amyloid plaques and intracellular neurofibrillary tangles consisting of aggregated and hyperphosphorylated Tau protein. Recent studies suggest that dysregulation of the microtubule-associated protein Tau, especially specific proteolysis, could be a driving force for Alzheimer's disease neurodegeneration. Tau physiologically promotes the assembly and stabilization of microtubules, whereas specific truncated fragments are sufficient to induce abnormal hyperphosphorylation and aggregate into toxic oligomers, resulting in them gaining prion-like characteristics. In addition, Tau truncations cause extensive impairments to neural and glial cell functions and animal cognition and behavior in a fragment-dependent manner. This review summarizes over 60 proteolytic cleavage sites and their corresponding truncated fragments, investigates the role of specific truncations in physiological and pathological states of Alzheimer's disease, and summarizes the latest applications of strategies targeting Tau fragments in the diagnosis and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xingyue Yang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Jing Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Yan Zhou
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong, Jiangsu Province, China
| | - Jin-Hua Gu
- Department of Clinical Pharmacy, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province, China
| | - Jin Miao
- Laboratory of Animal Center, Nantong University, Nantong, Jiangsu Province, China
| | - Feng Wu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
26
|
Zheng H, Sun H, Cai Q, Tai HC. The Enigma of Tau Protein Aggregation: Mechanistic Insights and Future Challenges. Int J Mol Sci 2024; 25:4969. [PMID: 38732197 PMCID: PMC11084794 DOI: 10.3390/ijms25094969] [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: 03/30/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Tau protein misfolding and aggregation are pathological hallmarks of Alzheimer's disease and over twenty neurodegenerative disorders. However, the molecular mechanisms of tau aggregation in vivo remain incompletely understood. There are two types of tau aggregates in the brain: soluble aggregates (oligomers and protofibrils) and insoluble filaments (fibrils). Compared to filamentous aggregates, soluble aggregates are more toxic and exhibit prion-like transmission, providing seeds for templated misfolding. Curiously, in its native state, tau is a highly soluble, heat-stable protein that does not form fibrils by itself, not even when hyperphosphorylated. In vitro studies have found that negatively charged molecules such as heparin, RNA, or arachidonic acid are generally required to induce tau aggregation. Two recent breakthroughs have provided new insights into tau aggregation mechanisms. First, as an intrinsically disordered protein, tau is found to undergo liquid-liquid phase separation (LLPS) both in vitro and inside cells. Second, cryo-electron microscopy has revealed diverse fibrillar tau conformations associated with different neurodegenerative disorders. Nonetheless, only the fibrillar core is structurally resolved, and the remainder of the protein appears as a "fuzzy coat". From this review, it appears that further studies are required (1) to clarify the role of LLPS in tau aggregation; (2) to unveil the structural features of soluble tau aggregates; (3) to understand the involvement of fuzzy coat regions in oligomer and fibril formation.
Collapse
Affiliation(s)
| | | | | | - Hwan-Ching Tai
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
27
|
Buchholz S, Zempel H. The six brain-specific TAU isoforms and their role in Alzheimer's disease and related neurodegenerative dementia syndromes. Alzheimers Dement 2024; 20:3606-3628. [PMID: 38556838 PMCID: PMC11095451 DOI: 10.1002/alz.13784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 04/02/2024]
Abstract
INTRODUCTION Alternative splicing of the human MAPT gene generates six brain-specific TAU isoforms. Imbalances in the TAU isoform ratio can lead to neurodegenerative diseases, underscoring the need for precise control over TAU isoform balance. Tauopathies, characterized by intracellular aggregates of hyperphosphorylated TAU, exhibit extensive neurodegeneration and can be classified by the TAU isoforms present in pathological accumulations. METHODS A comprehensive review of TAU and related dementia syndromes literature was conducted using PubMed, Google Scholar, and preprint server. RESULTS While TAU is recognized as key driver of neurodegeneration in specific tauopathies, the contribution of the isoforms to neuronal function and disease development remains largely elusive. DISCUSSION In this review we describe the role of TAU isoforms in health and disease, and stress the importance of comprehending and studying TAU isoforms in both, physiological and pathological context, in order to develop targeted therapeutic interventions for TAU-associated diseases. HIGHLIGHTS MAPT splicing is tightly regulated during neuronal maturation and throughout life. TAU isoform expression is development-, cell-type and brain region specific. The contribution of TAU to neurodegeneration might be isoform-specific. Ineffective TAU-based therapies highlight the need for specific targeting strategies.
Collapse
Affiliation(s)
- Sarah Buchholz
- Institute of Human GeneticsFaculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
- Present address:
Department Schaefer, Neurobiology of AgeingMax Planck Institute for Biology of AgeingCologneGermany
| | - Hans Zempel
- Institute of Human GeneticsFaculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| |
Collapse
|
28
|
Yakout DW, Shroff A, Wei W, Thaker V, Allen ZD, Sajish M, Nazarko TY, Mabb AM. Tau regulates Arc stability in neuronal dendrites via a proteasome-sensitive but ubiquitin-independent pathway. J Biol Chem 2024; 300:107237. [PMID: 38552740 PMCID: PMC11061231 DOI: 10.1016/j.jbc.2024.107237] [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: 05/01/2023] [Revised: 02/23/2024] [Accepted: 03/19/2024] [Indexed: 04/26/2024] Open
Abstract
Tauopathies are neurodegenerative disorders characterized by the deposition of aggregates of the microtubule-associated protein tau, a main component of neurofibrillary tangles. Alzheimer's disease (AD) is the most common type of tauopathy and dementia, with amyloid-beta pathology as an additional hallmark feature of the disease. Besides its role in stabilizing microtubules, tau is localized at postsynaptic sites and can regulate synaptic plasticity. The activity-regulated cytoskeleton-associated protein (Arc) is an immediate early gene that plays a key role in synaptic plasticity, learning, and memory. Arc has been implicated in AD pathogenesis and regulates the release of amyloid-beta. We found that decreased Arc levels correlate with AD status and disease severity. Importantly, Arc protein was upregulated in the hippocampus of Tau KO mice and dendrites of Tau KO primary hippocampal neurons. Overexpression of tau decreased Arc stability in an activity-dependent manner, exclusively in neuronal dendrites, which was coupled to an increase in the expression of dendritic and somatic surface GluA1-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors. The tau-dependent decrease in Arc was found to be proteasome-sensitive, yet independent of Arc ubiquitination and required the endophilin-binding domain of Arc. Importantly, these effects on Arc stability and GluA1 localization were not observed in the commonly studied tau mutant, P301L. These observations provide a potential molecular basis for synaptic dysfunction mediated through the accumulation of tau in dendrites. Our findings confirm that Arc is misregulated in AD and further show a physiological role for tau in regulating Arc stability and AMPA receptor targeting.
Collapse
Affiliation(s)
- Dina W Yakout
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Ankit Shroff
- Department of Biology, Georgia State University, Atlanta, Georgia, USA
| | - Wei Wei
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Vishrut Thaker
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Zachary D Allen
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Mathew Sajish
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA
| | - Taras Y Nazarko
- Department of Biology, Georgia State University, Atlanta, Georgia, USA
| | - Angela M Mabb
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia, USA.
| |
Collapse
|
29
|
Facal CL, Fernández Bessone I, Muñiz JA, Pereyra AE, Pedroncini O, Páez-Paz I, Clerici-Delville R, Arnaiz C, Urrutia L, Falasco G, Argañaraz CV, Saez T, Marin-Burgin A, Soiza-Reilly M, Falzone T, Avale ME. Tau reduction with artificial microRNAs modulates neuronal physiology and improves tauopathy phenotypes in mice. Mol Ther 2024; 32:1080-1095. [PMID: 38310353 PMCID: PMC11163272 DOI: 10.1016/j.ymthe.2024.01.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/19/2023] [Accepted: 01/30/2024] [Indexed: 02/05/2024] Open
Abstract
Abnormal tau accumulation is the hallmark of several neurodegenerative diseases, named tauopathies. Strategies aimed at reducing tau in the brain are promising therapeutic interventions, yet more precise therapies would require targeting specific nuclei and neuronal subpopulations affected by disease while avoiding global reduction of physiological tau. Here, we developed artificial microRNAs directed against the human MAPT mRNA to dwindle tau protein by engaging the endogenous RNA interference pathway. In human differentiated neurons in culture, microRNA-mediated tau reduction diminished neuronal firing without affecting neuronal morphology or impairing axonal transport. In the htau mouse model of tauopathy, we locally expressed artificial microRNAs in the prefrontal cortex (PFC), an area particularly vulnerable to initiating tau pathology in this model. Tau knockdown prevented the accumulation of insoluble and hyperphosphorylated tau, modulated firing activity of putative pyramidal neurons, and improved glucose uptake in the PFC. Moreover, such tau reduction prevented cognitive decline in aged htau mice. Our results suggest target engagement of designed tau-microRNAs to effectively reduce tau pathology, providing a proof of concept for a potential therapeutic approach based on local tau knockdown to rescue tauopathy-related phenotypes.
Collapse
Affiliation(s)
- Carolina Lucía Facal
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), CONICET, Buenos Aires, Argentina
| | - Iván Fernández Bessone
- Instituto de Biología Celular y Neurociencias (IBCN), Universidad de Buenos Aires, CONICET-UBA, Buenos Aires, Argentina
| | - Javier Andrés Muñiz
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), CONICET, Buenos Aires, Argentina
| | - A Ezequiel Pereyra
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), CONICET, Buenos Aires, Argentina
| | - Olivia Pedroncini
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), Partner Institute of the Max Planck Society, CONICET-MPSP, Buenos Aires, Argentina
| | - Indiana Páez-Paz
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), CONICET, Buenos Aires, Argentina
| | - Ramiro Clerici-Delville
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), CONICET, Buenos Aires, Argentina
| | - Cayetana Arnaiz
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), Partner Institute of the Max Planck Society, CONICET-MPSP, Buenos Aires, Argentina
| | - Leandro Urrutia
- Centro de imágenes Moleculares, FLENI, Buenos Aires, Argentina
| | - Germán Falasco
- Centro de imágenes Moleculares, FLENI, Buenos Aires, Argentina
| | - Carla Verónica Argañaraz
- Instituto de Fisiología Biología Molecular y Neurociencias (IFIBYNE), Universidad de Buenos Aires, CONICET-UBA, Buenos Aires, Argentina
| | - Trinidad Saez
- Instituto de Biología Celular y Neurociencias (IBCN), Universidad de Buenos Aires, CONICET-UBA, Buenos Aires, Argentina
| | - Antonia Marin-Burgin
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), Partner Institute of the Max Planck Society, CONICET-MPSP, Buenos Aires, Argentina
| | - Mariano Soiza-Reilly
- Instituto de Fisiología Biología Molecular y Neurociencias (IFIBYNE), Universidad de Buenos Aires, CONICET-UBA, Buenos Aires, Argentina
| | - Tomás Falzone
- Instituto de Biología Celular y Neurociencias (IBCN), Universidad de Buenos Aires, CONICET-UBA, Buenos Aires, Argentina; Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), Partner Institute of the Max Planck Society, CONICET-MPSP, Buenos Aires, Argentina
| | - María Elena Avale
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), CONICET, Buenos Aires, Argentina.
| |
Collapse
|
30
|
Jarek DJ, Mizerka H, Nuszkiewicz J, Szewczyk-Golec K. Evaluating p-tau217 and p-tau231 as Biomarkers for Early Diagnosis and Differentiation of Alzheimer's Disease: A Narrative Review. Biomedicines 2024; 12:786. [PMID: 38672142 PMCID: PMC11048667 DOI: 10.3390/biomedicines12040786] [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: 02/15/2024] [Revised: 03/26/2024] [Accepted: 03/30/2024] [Indexed: 04/28/2024] Open
Abstract
The escalating prevalence of Alzheimer's disease (AD) highlights the urgent need to develop reliable biomarkers for early diagnosis and intervention. AD is characterized by the pathological accumulation of amyloid-beta plaques and tau neurofibrillary tangles. Phosphorylated tau (p-tau) proteins, particularly p-tau217 and p-tau231, have been identified as promising biomarker candidates to differentiate the disease progression from preclinical stages. This narrative review is devoted to a critical evaluation of the diagnostic accuracy, sensitivity, and specificity of p-tau217 and p-tau231 levels in the detection of AD, measured in plasma, serum, and cerebrospinal fluid, compared to established biomarkers. Additionally, the efficacy of these markers in distinguishing AD from other neurodegenerative disorders is examined. The significant advances offered by p-tau217 and p-tau231 in AD diagnostics are highlighted, demonstrating their unique utility in early detection and differential diagnosis. This comprehensive analysis not only confirms the excellent diagnostic capabilities of these markers, but also deepens the understanding of the molecular dynamics of AD, contributing to the broader scientific discourse on neurodegenerative diseases. This review is aimed to provide key information for researchers and clinicians across disciplines, filling interdisciplinary gaps and highlighting the role of p-tau proteins in revolutionizing AD research and clinical practice.
Collapse
Affiliation(s)
- Dorian Julian Jarek
- Student Research Club of Medical Biology and Biochemistry, Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland;
| | - Hubert Mizerka
- Student Research Club of Medical Biology and Biochemistry, Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland;
| | - Jarosław Nuszkiewicz
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland;
| | - Karolina Szewczyk-Golec
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland;
| |
Collapse
|
31
|
Han M, Saxton A, Currey H, Waldherr SM, Liachko NF, Kraemer BC. Transgenic Dendra2::tau expression allows in vivo monitoring of tau proteostasis in Caenorhabditis elegans. Dis Model Mech 2024; 17:dmm050473. [PMID: 38469687 PMCID: PMC10985736 DOI: 10.1242/dmm.050473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 03/04/2024] [Indexed: 03/13/2024] Open
Abstract
Protein homeostasis is perturbed in aging-related neurodegenerative diseases called tauopathies, which are pathologically characterized by aggregation of the microtubule-associated protein tau (encoded by the human MAPT gene). Transgenic Caenorhabditis elegans serve as a powerful model organism to study tauopathy disease mechanisms, but moderating transgenic expression level has proven problematic. To study neuronal tau proteostasis, we generated a suite of transgenic strains expressing low, medium or high levels of Dendra2::tau fusion proteins by comparing integrated multicopy transgene arrays with single-copy safe-harbor locus strains generated by recombinase-mediated cassette exchange. Multicopy Dendra2::tau strains exhibited expression level-dependent neuronal dysfunction that was modifiable by known genetic suppressors or an enhancer of tauopathy. Single-copy Dendra2::tau strains lacked distinguishable phenotypes on their own but enabled detection of enhancer-driven neuronal dysfunction. We used multicopy Dendra2::tau strains in optical pulse-chase experiments measuring tau turnover in vivo and found that Dendra2::tau turned over faster than the relatively stable Dendra2. Furthermore, Dendra2::tau turnover was dependent on the protein expression level and independent of co-expression with human TDP-43 (officially known as TARDBP), an aggregating protein interacting with pathological tau. We present Dendra2::tau transgenic C. elegans as a novel tool for investigating molecular mechanisms of tau proteostasis.
Collapse
Affiliation(s)
- Marina Han
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - Aleen Saxton
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Heather Currey
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Sarah M Waldherr
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Nicole F Liachko
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Brian C Kraemer
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
32
|
Jos S, Poulose R, Kambaru A, Gogoi H, Dalavaikodihalli Nanjaiah N, Padmanabhan B, Mehta B, Padavattan S. Tau-S214 Phosphorylation Inhibits Fyn Kinase Interaction and Increases the Decay Time of NMDAR-mediated Current. J Mol Biol 2024; 436:168445. [PMID: 38218365 DOI: 10.1016/j.jmb.2024.168445] [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: 08/17/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
Fyn kinase SH3 domain interaction with PXXP motif in the Tau protein is implicated in AD pathology and is central to NMDAR function. Among seven PXXP motifs localized in proline-rich domain of Tau protein, tandem 5th and 6th PXXP motifs are critical to Fyn-SH3 domain interaction. Here, we report the crystal structure of Fyn-SH3 -Tau (207-221) peptide consisting of 5th and 6th PXXP motif complex to 1.01 Å resolution. Among five AD-specific phosphorylation sites encompassing the 5th and 6th PXXP motifs, only S214 residue showed interaction with SH3 domain. Biophysical studies showed that Tau (207-221) with S214-phosphorylation (pS214) inhibits its interaction with Fyn-SH3 domain. The individual administration of Tau (207-221) with/without pS214 peptides to a single neuron increased the decay time of evoked NMDA current response. Recordings of spontaneous NMDA EPSCs at +40 mV indicate an increase in frequency and amplitude of events for the Tau (207-221) peptide. Conversely, the Tau (207-221) with pS214 peptide exhibited a noteworthy amplitude increase alongside a prolonged decay time. These outcomes underscore the distinctive modalities of action associated with each peptide in the study. Overall, this study provides insights into how Tau (207-221) with/without pS214 affects the molecular framework of NMDAR signaling, indicating its involvement in Tau-related pathogenesis.
Collapse
Affiliation(s)
- Sneha Jos
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Roshni Poulose
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Archanalakshmi Kambaru
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Hemanga Gogoi
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | | | - Balasundaram Padmanabhan
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Bhupesh Mehta
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India.
| | - Sivaraman Padavattan
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India.
| |
Collapse
|
33
|
Alonso ADC, El Idrissi A, Candia R, Morozova V, Kleiman FE. Tau: More than a microtubule-binding protein in neurons. Cytoskeleton (Hoboken) 2024; 81:71-77. [PMID: 37819542 DOI: 10.1002/cm.21795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023]
Abstract
Tau protein was discovered as a microtubule-associated protein nearly 50 years ago, and our understanding of tau has revolved around that role. Even with tau's rise to stardom as a central player in neurodegenerative disease, therapeutic efforts have largely been targeted toward cytoskeletal changes. While some studies hinted toward non-cytoskeletal roles for tau, it is only fairly recently that these ideas have begun to receive considerable attention. Many new binding partners for tau have been identified, including DNA, RNA, RNA-binding proteins, some receptors, and other tau molecules. The diversity of tau binding partners coupled with the discovery of tau other than axonal compartments such as nucleus, dendrites, and synapses have led to the proposal of novel functions for tau in roles such as nuclear stability, cell signaling, transcriptional processing, and protein synthesis. Tau self-assembly in particular has made an impact, leading to the hypothesis that a prion-like function of hyperphosphorylated tau is central to tauopathies. With tau emerging as a multifaceted protein that operates in many parts of the cell and with many molecular partners, the field of tau biology is primed for discoveries that can provide new perspectives on both the unique biochemistry of tau and the nature of devastating neurological diseases.
Collapse
Affiliation(s)
- Alejandra Del Carmen Alonso
- Biology Department and Center for Developmental Neuroscience, College of Staten Island, Staten Island, New York, USA
- Biology Program, The Graduate Center, The City University of New York, New York, New York, USA
| | - Abdeslem El Idrissi
- Biology Department and Center for Developmental Neuroscience, College of Staten Island, Staten Island, New York, USA
- Biology Program, The Graduate Center, The City University of New York, New York, New York, USA
| | - Robert Candia
- Biology Department and Center for Developmental Neuroscience, College of Staten Island, Staten Island, New York, USA
- Biology Program, The Graduate Center, The City University of New York, New York, New York, USA
| | - Viktoriya Morozova
- Biology Department and Center for Developmental Neuroscience, College of Staten Island, Staten Island, New York, USA
- Biology Program, The Graduate Center, The City University of New York, New York, New York, USA
- Helene Fuld College of Nursing, New York, New York, USA
| | - Frida Esther Kleiman
- Biology Program, The Graduate Center, The City University of New York, New York, New York, USA
- Chemistry Department, Hunter College, The City University of New York, New York, New York, USA
| |
Collapse
|
34
|
Cai Q, Tai HC. Super-Resolution Imaging of Tau Proteins in Isolated and Immobilized Brain Synaptosomes. Methods Mol Biol 2024; 2754:445-456. [PMID: 38512681 DOI: 10.1007/978-1-0716-3629-9_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Tau protein has important physiological functions at both presynaptic and postsynaptic terminals. Pathological tau species are also associated with synaptic dysfunctions in several neurodegenerative disorders, especially Alzheimer's disease. To understand tau distribution inside synaptic compartments, super-resolution imaging is required. Here, we describe a facile protocol to immobilize and image brain synaptosomes without aggregation artefacts, by substituting the standard fixative paraformaldehyde with ethylene glycol bis(succinimidyl succinate) (EGS). Super-resolution imaging of tau proteins is achieved through three-color direct stochastic optical reconstruction microscopy (dSTORM). Tau protein is found to colocalize with synaptic vesicles as well as postsynaptic densities.
Collapse
Affiliation(s)
- Qixu Cai
- State Key Laboratory of Vaccines for Infectious Diseases, School of Public Health, Xiamen University, Xiamen, People's Republic of China
| | - Hwan-Ching Tai
- State Key Laboratory of Vaccines for Infectious Diseases, School of Public Health, Xiamen University, Xiamen, People's Republic of China.
| |
Collapse
|
35
|
Wang D, Zheng J, Sun X, Xie L, Yang Y. Study on the Pharmacological Mechanism of Icariin for the Treatment of Alzheimer's Disease Based on Network Pharmacology and Molecular Docking Techniques. Metabolites 2023; 14:1. [PMID: 38276291 PMCID: PMC10820555 DOI: 10.3390/metabo14010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
The purpose of this study is to explore the pharmacological mechanism of icariin (ICA) in the treatment of Alzheimer's disease (AD) based on network pharmacology and network molecular docking technology. In order to investigate the regulatory effect of ICA on the expression level of AD pathological phosphorylation regulatory proteins, this study further explored the possible molecular mechanism of ICA regulating AD autophagy through network pharmacology. Macromolecular docking network was verified by Autodock Vina 1.1.2 software. The main active ingredients of ICA, the physicochemical properties, and pharmacokinetic information of ICA were predicted using online databases and relevant information. The results showed that the targets of MAPK3, AKT1, HSP90AA1, ESR1, and HSP90AA1 were more critical in the treatment of AD. Autophagy, apoptosis, senescence factors, phosphatidylinositide 3-kinase/protein kinase B (P13K/AKT) signaling pathway, MAKP, mTOR, and other pathways were significantly associated with AD. Docking of ICA with HIF-1, BNIP3, PINK1, and Parkin pathway molecules showed that the key targets of the signaling pathway were more stably bound to ICA, which may provide a better pathway for ICA to regulate autophagy by providing a better pathway. ICA can improve AD, and its mechanism may be related to the P13K/AKT, MAKP, and mTOR signaling pathways, thereby regulating autophagy-related proteins.
Collapse
Affiliation(s)
- Dongwei Wang
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
| | - Jilong Zheng
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
| | - Xingsheng Sun
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
| | - Liuwei Xie
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
- The Second Affiliated Hospital of Shenyang Medical College, Shenyang 110031, China
| | - Yang Yang
- The Second Affiliated Hospital of Shenyang Medical College, Shenyang 110031, China
| |
Collapse
|
36
|
Chang YL, Yang CC, Huang YY, Chen YA, Yang CW, Liao CY, Li H, Wu CS, Lin CH, Teng SC. The HSP40 family chaperone isoform DNAJB6b prevents neuronal cells from tau aggregation. BMC Biol 2023; 21:293. [PMID: 38110916 PMCID: PMC10729500 DOI: 10.1186/s12915-023-01798-6] [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: 02/23/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disorder with clinical presentations of progressive cognitive and memory deterioration. The pathologic hallmarks of AD include tau neurofibrillary tangles and amyloid plaque depositions in the hippocampus and associated neocortex. The neuronal aggregated tau observed in AD cells suggests that the protein folding problem is a major cause of AD. J-domain-containing proteins (JDPs) are the largest family of cochaperones, which play a vital role in specifying and directing HSP70 chaperone functions. JDPs bind substrates and deliver them to HSP70. The association of JDP and HSP70 opens the substrate-binding domain of HSP70 to help the loading of the clients. However, in the initial HSP70 cycle, which JDP delivers tau to the HSP70 system in neuronal cells remains unclear. RESULTS We screened the requirement of a diverse panel of JDPs for preventing tau aggregation in the human neuroblastoma cell line SH-SY5Y by a filter retardation method. Interestingly, knockdown of DNAJB6, one of the JDPs, displayed tau aggregation and overexpression of DNAJB6b, one of the isoforms generated from the DNAJB6 gene by alternative splicing, reduced tau aggregation. Further, the tau bimolecular fluorescence complementation assay confirmed the DNAJB6b-dependent tau clearance. The co-immunoprecipitation and the proximity ligation assay demonstrated the protein-protein interaction between tau and the chaperone-cochaperone complex. The J-domain of DNAJB6b was critical for preventing tau aggregation. Moreover, reduced DNAJB6 expression and increased tau aggregation were detected in an age-dependent manner in immunohistochemical analysis of the hippocampus tissues of a mouse model of tau pathology. CONCLUSIONS In summary, downregulation of DNAJB6b increases the insoluble form of tau, while overexpression of DNAJB6b reduces tau aggregation. Moreover, DNAJB6b associates with tau. Therefore, this study reveals that DNAJB6b is a direct sensor for its client tau in the HSP70 folding system in neuronal cells, thus helping to prevent AD.
Collapse
Affiliation(s)
- Ya-Lan Chang
- Department of Microbiology, College of Medicine, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei, 10051, Taiwan
| | - Chan-Chih Yang
- Department of Microbiology, College of Medicine, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei, 10051, Taiwan
| | - Yun-Yu Huang
- Department of Microbiology, College of Medicine, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei, 10051, Taiwan
| | - Yi-An Chen
- Department of Microbiology, College of Medicine, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei, 10051, Taiwan
| | - Chia-Wei Yang
- Department of Microbiology, College of Medicine, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei, 10051, Taiwan
| | - Chia-Yu Liao
- Department of Microbiology, College of Medicine, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei, 10051, Taiwan
| | - Hsun Li
- Department of Neurology, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 10051, Taiwan
| | - Ching-Shyi Wu
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 10051, Taiwan.
| | - Shu-Chun Teng
- Department of Microbiology, College of Medicine, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei, 10051, Taiwan.
- Center of Precision Medicine, National Taiwan University, Taipei, 10051, Taiwan.
| |
Collapse
|
37
|
Younas N, Saleem T, Younas A, Zerr I. Nuclear face of Tau: an inside player in neurodegeneration. Acta Neuropathol Commun 2023; 11:196. [PMID: 38087392 PMCID: PMC10714511 DOI: 10.1186/s40478-023-01702-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Tau (Tubulin associated unit) protein is a major hallmark of Alzheimer's disease (AD) and tauopathies. Tau is predominantly an axonal protein with a crucial role in the stabilization and dynamics of the microtubules. Since the discovery of Tau protein in 1975, research efforts were concentrated on the pathophysiological role of Tau protein in the context of the microtubules. Although, for more than three decades, different localizations of Tau protein have been discovered e.g., in the nuclear compartments. Discovery of the role of Tau protein in various cellular compartments especially in the nucleus opens up a new fold of complexity in tauopathies. Data from cellular models, animal models, and the human brain indicate that nuclear Tau is crucial for genome stability and to cope with cellular distress. Moreover, it's nature of nuclear translocation, its interactions with the nuclear DNA/RNA and proteins suggest it could play multiple roles in the nucleus. To comprehend Tau pathophysiology and efficient Tau-based therapies, there is an urgent need to understand whole repertoire of Tau species (nuclear and cytoplasmic) and their functional relevance. To complete the map of Tau repertoire, understanding of various species of Tau in the nucleus and cytoplasm, identification if specific transcripts of Tau, isoforms and post-translational modifications could foretell Tau's localizations and functions, and how they are modified in neurodegenerative diseases like AD, is urgently required. In this review, we explore the nuclear face of Tau protein, its nuclear localizations and functions and its linkage with Alzheimer's disease.
Collapse
Affiliation(s)
- Neelam Younas
- University Medical Center Göttingen, National Reference Center for Surveillance of TSE, Department of Neurology, Robert-Koch strasse 40, 37075, Göttingen, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, 37075, Germany.
| | - Tayyaba Saleem
- University Medical Center Göttingen, National Reference Center for Surveillance of TSE, Department of Neurology, Robert-Koch strasse 40, 37075, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, 37075, Germany
| | - Abrar Younas
- University Medical Center Göttingen, National Reference Center for Surveillance of TSE, Department of Neurology, Robert-Koch strasse 40, 37075, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, 37075, Germany
| | - Inga Zerr
- University Medical Center Göttingen, National Reference Center for Surveillance of TSE, Department of Neurology, Robert-Koch strasse 40, 37075, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, 37075, Germany
| |
Collapse
|
38
|
Hamilton K, Morrow K, Markantoni E, Harvey J. Leptin prevents aberrant targeting of tau to hippocampal synapses via PI 3 kinase driven inhibition of GSK3β. J Neurochem 2023; 167:520-537. [PMID: 37822142 DOI: 10.1111/jnc.15980] [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: 05/09/2023] [Revised: 09/11/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023]
Abstract
Amyloid-β (Aβ) and hyper-phosphorylated tau are key hallmarks of Alzheimer's disease (AD), with an accumulation of both proteins linked to hippocampal synaptic dysfunction. Recent evidence indicates that Aβ drives mis-localisation of tau from axons to synapses, resulting in AMPA receptor (AMPAR) internalisation and impaired excitatory synaptic function. These tau-driven synaptic impairments are thought to underlie the cognitive deficits in AD. Consequently, limiting the synapto-toxic effects of tau may prevent AD-related cognitive deficits. Increasing evidence links leptin dysfunction with higher AD risk, and numerous studies have identified neuroprotective properties of leptin in AD models of Aβ-induced toxicity. However, it is unclear if leptin protects against tau-related synaptic dysfunction. Here we show that Aβ1-42 significantly increases dendritic and synaptic levels of tau and p-tau in hippocampal neurons, and these effects were blocked by leptin. In accordance with GSK-3β being involved in tau phosphorylation, the protective effects of leptin involve PI 3-kinase (PI3K) activation and inhibition of GSK-3β. Aβ1-42 -driven synaptic targeting of tau was associated with the removal of GluA1-containing AMPARs from synapses, which was also inhibited by leptin-driven inhibition of GSK-3β. Direct application of oligomeric tau to hippocampal neurons caused internalisation of GluA1-containing AMPARs and this effect was blocked by prior application of leptin. Similarly, leptin prevented the ability of tau to block induction of activity-dependent long-term potentiation (LTP) at hippocampal SC-CA1 synapses. These findings increase our understanding of the neuroprotective actions of leptin in the early pre-clinical stages of AD and further validate the leptin system as a therapeutic target in AD.
Collapse
Affiliation(s)
- Kirsty Hamilton
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Kate Morrow
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Ermione Markantoni
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Jenni Harvey
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| |
Collapse
|
39
|
Wang L, Zhu R, Zhou X, Zhang Z, Peng D. Altered local and remote functional connectivity in mild Alzheimer's disease patients with sleep disturbances. Front Aging Neurosci 2023; 15:1269582. [PMID: 37920381 PMCID: PMC10619161 DOI: 10.3389/fnagi.2023.1269582] [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: 07/30/2023] [Accepted: 10/03/2023] [Indexed: 11/04/2023] Open
Abstract
Objectives This study aimed to investigate local and remote functional connectivity in mild Alzheimer's disease patients with sleep disturbances (ADSD) and those without sleep disturbances (ADNSD). Methods Thirty eight mild AD patients with sleep disturbances and 21 mild AD patients without sleep disturbances participated in this study. All subjects underwent neuropsychological assessments and 3.0 Tesla magnetic resonance scanning. Static and dynamic regional homogeneity (ReHo) were used to represent the local functional connectivity. Seed-based whole-brain functional connectivity was used to represent the remote functional connectivity. The seed was chosen based on the results of ReHo. Results Compared to ADNSD, ADSD showed decreased static ReHo in the left posterior central gyrus and the right cuneus and increased dynamic ReHo in the left posterior central gyrus. As for the remote functional connectivity, comparing ADSD to ADNSD, it was found that there was a decreased functional connection between the left posterior central gyrus and the left cuneus as well as the left calcarine. Conclusion The current study demonstrated that, compared with ADNSD, ADSD is impaired in both local and remote functional connectivity, manifested as reduced functional connectivity involving the primary sensory network and the primary visual network. The abnormality of the above functional connectivity is one of the reasons why sleep disorders promote cognitive impairment in AD. Moreover, sleep disorders change the temporal sequence of AD pathological damage to brain functional networks, but more evidence is needed to support this conclusion.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurology, Beijing Geriatric Hospital, Beijing, China
| | - Rui Zhu
- Department of Neurology, Beijing Geriatric Hospital, Beijing, China
| | - Xiao Zhou
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhiyong Zhang
- Department of Neurology, Beijing Geriatric Hospital, Beijing, China
| | - Dantao Peng
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
40
|
Zuo WF, Pang Q, Yao LP, Zhang Y, Peng C, Huang W, Han B. Gut microbiota: A magical multifunctional target regulated by medicine food homology species. J Adv Res 2023; 52:151-170. [PMID: 37269937 PMCID: PMC10555941 DOI: 10.1016/j.jare.2023.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/27/2023] [Accepted: 05/28/2023] [Indexed: 06/05/2023] Open
Abstract
BACKGROUND The relationship between gut microbiota and human health has gradually been recognized. Increasing studies show that the disorder of gut microbiota is related to the occurrence and development of many diseases. Metabolites produced by the gut microbiota are responsible for their extensive regulatory roles. In addition, naturally derived medicine food homology species with low toxicity and high efficiency have been clearly defined owing to their outstanding physiological and pharmacological properties in disease prevention and treatment. AIM OF REVIEW Based on supporting evidence, the current review summarizes the representative work of medicine food homology species targeting the gut microbiota to regulate host pathophysiology and discusses the challenges and prospects in this field. It aims to facilitate the understanding of the relationship among medicine food homology species, gut microbiota, and human health and further stimulate the advancement of more relevant research. KEY SCIENTIFIC CONCEPTS OF REVIEW As this review reveals, from the initial practical application to more mechanism studies, the relationship among medicine food homology species, gut microbiota, and human health has evolved into an irrefutable interaction. On the one hand, through affecting the population structure, metabolism, and function of gut microbiota, medicine food homology species maintain the homeostasis of the intestinal microenvironment and human health by affecting the population structure, metabolism, and function of gut microbiota. On the other hand, the gut microbiota is also involved in the bioconversion of the active ingredients from medicine food homology species and thus influences their physiological and pharmacological properties.
Collapse
Affiliation(s)
- Wei-Fang Zuo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qiwen Pang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Lai-Ping Yao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yang Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
41
|
Buccellato FR, D’Anca M, Tartaglia GM, Del Fabbro M, Scarpini E, Galimberti D. Treatment of Alzheimer's Disease: Beyond Symptomatic Therapies. Int J Mol Sci 2023; 24:13900. [PMID: 37762203 PMCID: PMC10531090 DOI: 10.3390/ijms241813900] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
In an ever-increasing aged world, Alzheimer's disease (AD) represents the first cause of dementia and one of the first chronic diseases in elderly people. With 55 million people affected, the WHO considers AD to be a disease with public priority. Unfortunately, there are no final cures for this pathology. Treatment strategies are aimed to mitigate symptoms, i.e., acetylcholinesterase inhibitors (AChEI) and the N-Methyl-D-aspartate (NMDA) antagonist Memantine. At present, the best approaches for managing the disease seem to combine pharmacological and non-pharmacological therapies to stimulate cognitive reserve. Over the last twenty years, a number of drugs have been discovered acting on the well-established biological hallmarks of AD, deposition of β-amyloid aggregates and accumulation of hyperphosphorylated tau protein in cells. Although previous efforts disappointed expectations, a new era in treating AD has been working its way recently. The Food and Drug Administration (FDA) gave conditional approval of the first disease-modifying therapy (DMT) for the treatment of AD, aducanumab, a monoclonal antibody (mAb) designed against Aβ plaques and oligomers in 2021, and in January 2023, the FDA granted accelerated approval for a second monoclonal antibody, Lecanemab. This review describes ongoing clinical trials with DMTs and non-pharmacological therapies. We will also present a future scenario based on new biomarkers that can detect AD in preclinical or prodromal stages, identify people at risk of developing AD, and allow an early and curative treatment.
Collapse
Affiliation(s)
- Francesca R. Buccellato
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
- Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Marianna D’Anca
- Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Gianluca Martino Tartaglia
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
- Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Massimo Del Fabbro
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
- Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Elio Scarpini
- Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
- Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
42
|
Lu J, Liang F, Bai P, Liu C, Xu M, Sun Z, Tian W, Dong Y, Zhang Y, Quan Q, Khatri A, Shen Y, Marcantonio E, Crosby G, Culley D, Wang C, Yang G, Xie Z. Blood tau-PT217 contributes to the anesthesia/surgery-induced delirium-like behavior in aged mice. Alzheimers Dement 2023; 19:4110-4126. [PMID: 37249148 PMCID: PMC10524579 DOI: 10.1002/alz.13118] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 05/31/2023]
Abstract
INTRODUCTION Blood phosphorylated tau at threonine 217 (tau-PT217) is a newly established biomarker for Alzheimer's disease and postoperative delirium in patients. However, the mechanisms and consequences of acute changes in blood tau-PT217 remain largely unknown. METHODS We investigated the effects of anesthesia/surgery on blood tau-PT217 in aged mice, and evaluated the associated changes in B cell populations, neuronal excitability in anterior cingulate cortex, and delirium-like behavior using positron emission tomography imaging, nanoneedle technology, flow cytometry, electrophysiology, and behavioral tests. RESULTS Anesthesia/surgery induced acute increases in blood tau-PT217 via enhanced generation in the lungs and release from B cells. Tau-PT217 might cross the blood-brain barrier, increasing neuronal excitability and inducing delirium-like behavior. B cell transfer and WS635, a mitochondrial function enhancer, mitigated the anesthesia/surgery-induced changes. DISCUSSION Acute increases in blood tau-PT217 may contribute to brain dysfunction and postoperative delirium. Targeting B cells or mitochondrial function may have therapeutic potential for preventing or treating these conditions.
Collapse
Affiliation(s)
- Jing Lu
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, 610072, China
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, United States
| | - Feng Liang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, United States
| | - Ping Bai
- Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, United States
| | - Chenghao Liu
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, United States
- Chinese Academy of Sciences, Institute of Automation, Beijing, 100080, China
| | - Miao Xu
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, United States
- Department of Anesthesiology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| | - Zhengwang Sun
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, United States
| | - Wenjie Tian
- Department of Cardiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, 610072, China
- Cardiovascular Research Center and Cardiology Division of the Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, United States
| | - Yuanlin Dong
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, United States
| | - Yiying Zhang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, United States
| | - Qimin Quan
- NanoMosaic, Inc., Woburn, MA, 01801, United States
| | - Ashok Khatri
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129, United States
| | - Yuan Shen
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, United States
- Anesthesia and Brain Research Institute, Tongji University School of Medicine, Shanghai, 200092, China
- Mental Health Center affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Edward Marcantonio
- Divisions of General Medicine and Primary Care and Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02115, United States
| | - Gregory Crosby
- Department of Anesthesiology, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, 02115, United States
| | - Deborah Culley
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania Health System, Philadelphia, PA, 19104, United States
| | - Changning Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, United States
| | - Guang Yang
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, 10032, United States
| | - Zhongcong Xie
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, United States
| |
Collapse
|
43
|
Forrest SL, Lee S, Nassir N, Martinez-Valbuena I, Sackmann V, Li J, Ahmed A, Tartaglia MC, Ittner LM, Lang AE, Uddin M, Kovacs GG. Cell-specific MAPT gene expression is preserved in neuronal and glial tau cytopathologies in progressive supranuclear palsy. Acta Neuropathol 2023; 146:395-414. [PMID: 37354322 PMCID: PMC10412651 DOI: 10.1007/s00401-023-02604-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/11/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023]
Abstract
Microtubule-associated protein tau (MAPT) aggregates in neurons, astrocytes and oligodendrocytes in a number of neurodegenerative diseases, including progressive supranuclear palsy (PSP). Tau is a target of therapy and the strategy includes either the elimination of pathological tau aggregates or reducing MAPT expression, and thus the amount of tau protein made to prevent its aggregation. Disease-associated tau affects brain regions in a sequential manner that includes cell-to-cell spreading. Involvement of glial cells that show tau aggregates is interpreted as glial cells taking up misfolded tau assuming that glial cells do not express enough MAPT. Although studies have evaluated MAPT expression in human brain tissue homogenates, it is not clear whether MAPT expression is compromised in cells accumulating pathological tau. To address these perplexing aspects of disease pathogenesis, this study used RNAscope combined with immunofluorescence (AT8), and single-nuclear(sn) RNAseq to systematically map and quantify MAPT expression dynamics across different cell types and brain regions in controls (n = 3) and evaluated whether tau cytopathology affects MAPT expression in PSP (n = 3). MAPT transcripts were detected in neurons, astrocytes and oligodendrocytes, and varied between brain regions and within each cell type, and were preserved in all cell types with tau aggregates in PSP. These results propose a complex scenario in all cell types, where, in addition to the ingested misfolded tau, the preserved cellular MAPT expression provides a pool for local protein production that can (1) be phosphorylated and aggregated, or (2) feed the seeding of ingested misfolded tau by providing physiological tau, both accentuating the pathological process. Since tau cytopathology does not compromise MAPT gene expression in PSP, a complete loss of tau protein expression as an early pathogenic component is less likely. These observations provide rationale for a dual approach to therapy by decreasing cellular MAPT expression and targeting removal of misfolded tau.
Collapse
Affiliation(s)
- Shelley L Forrest
- Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Krembil Discovery Tower, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Seojin Lee
- Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Krembil Discovery Tower, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
| | - Nasna Nassir
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Ivan Martinez-Valbuena
- Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Krembil Discovery Tower, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
| | - Valerie Sackmann
- Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Krembil Discovery Tower, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
| | - Jun Li
- Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Krembil Discovery Tower, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
| | - Awab Ahmed
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Krembil Discovery Tower, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
- University Health Network Memory Clinic, Krembil Brain Institute, Toronto, ON, Canada
| | - Lars M Ittner
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Anthony E Lang
- Edmond J. Safra Program in Parkinson's Disease, Rossy PSP Centre and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, Canada
| | - Mohammed Uddin
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
- Cellular Intelligence (Ci) Lab, GenomeArc Inc., Toronto, ON, Canada
| | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Krembil Discovery Tower, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada.
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia.
- Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, ON, Canada.
- Edmond J. Safra Program in Parkinson's Disease, Rossy PSP Centre and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology and Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
44
|
Mate De Gerando A, Welikovitch LA, Khasnavis A, Commins C, Glynn C, Chun JE, Perbet R, Hyman BT. Tau seeding and spreading in vivo is supported by both AD-derived fibrillar and oligomeric tau. Acta Neuropathol 2023; 146:191-210. [PMID: 37341831 PMCID: PMC10329061 DOI: 10.1007/s00401-023-02600-1] [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: 03/22/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/22/2023]
Abstract
Insoluble fibrillar tau, the primary constituent of neurofibrillary tangles, has traditionally been thought to be the biologically active, toxic form of tau mediating neurodegeneration in Alzheimer's disease. More recent studies have implicated soluble oligomeric tau species, referred to as high molecular weight (HMW), due to their properties on size-exclusion chromatography, in tau propagation across neural systems. These two forms of tau have never been directly compared. We prepared sarkosyl-insoluble and HMW tau from the frontal cortex of Alzheimer patients and compared their properties using a variety of biophysical and bioactivity assays. Sarkosyl-insoluble fibrillar tau comprises abundant paired-helical filaments (PHF) as quantified by electron microscopy (EM) and is more resistant to proteinase K, compared to HMW tau, which is mostly in an oligomeric form. Sarkosyl-insoluble and HMW tau are nearly equivalent in potency in HEK cell bioactivity assay for seeding aggregates, and their injection reveals similar local uptake into hippocampal neurons in PS19 Tau transgenic mice. However, the HMW preparation appears to be far more potent in inducing a glial response including Clec7a-positive rod microglia in the absence of neurodegeneration or synapse loss and promotes more rapid propagation of misfolded tau to distal, anatomically connected regions, such as entorhinal and perirhinal cortices. These data suggest that soluble HMW tau has similar properties to fibrillar sarkosyl-insoluble tau with regard to tau seeding potential, but may be equal or even more bioactive with respect to propagation across neural systems and activation of glial responses, both relevant to tau-related Alzheimer phenotypes.
Collapse
Affiliation(s)
- Anastasie Mate De Gerando
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Cambridge, MA, USA
| | - Lindsay A Welikovitch
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Cambridge, MA, USA
| | - Anita Khasnavis
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Cambridge, MA, USA
| | - Caitlin Commins
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Cambridge, MA, USA
| | - Calina Glynn
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Cambridge, MA, USA
| | - Joshua E Chun
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Cambridge, MA, USA
| | - Romain Perbet
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Cambridge, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Cambridge, MA, USA.
| |
Collapse
|
45
|
van Hummel A, Taleski G, Sontag J, Feiten AF, Ke YD, Ittner LM, Sontag E. Methyl donor supplementation reduces phospho-Tau, Fyn and demethylated protein phosphatase 2A levels and mitigates learning and motor deficits in a mouse model of tauopathy. Neuropathol Appl Neurobiol 2023; 49:e12931. [PMID: 37565253 PMCID: PMC10947299 DOI: 10.1111/nan.12931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 07/27/2023] [Accepted: 08/05/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Reduced folate status and elevated levels of circulating homocysteine are modifiable risk factors for cognitive decline and dementia. Disturbances in one-carbon metabolism are associated with the pathological accumulation of phosphorylated tau, a hallmark feature of prevalent dementia, including Alzheimer's disease and subgroups of frontotemporal dementia. METHODS Here, using transgenic TAU58/2 mouse models of human tauopathy, we tested whether dietary supplementation with L-methylfolate (the active folate form), choline and betaine can reduce tau phosphorylation and associated behavioural phenotypes. RESULTS TAU58/2 mice fed with the methyl donor-enriched diet showed reduced phosphorylation of tau at the pathological S202 (CP13) and S396/S404 (PHF-1) epitopes and alleviation of associated motor and learning deficits. Compared with mice on the control diet, the decrease in cortical phosphorylated tau levels in mice fed with the methyl donor-enriched diet was associated with enhanced methylation of protein phosphatase 2A, the major brain tau Ser/Thr phosphatase. It also correlated with a reduction in protein levels of Fyn, a tau tyrosine kinase that plays a central role in mediating pathological tau-induced neurodegeneration. Conversely, Fyn expression levels were increased in mice with deficiencies in folate metabolism. CONCLUSIONS Our findings provide the first experimental evidence that boosting one-carbon metabolism with L-methylfolate, choline and betaine can mitigate key pathological, learning and motor deficits in a tauopathy mouse model. They give support to using a combination of methyl donors as a preventive or disease-modifying strategy for tauopathies.
Collapse
Affiliation(s)
- Annika van Hummel
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Goce Taleski
- School of Biomedical Sciences and PharmacyUniversity of NewcastleCallaghanNew South WalesAustralia
- Hunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Jean‐Marie Sontag
- School of Biomedical Sciences and PharmacyUniversity of NewcastleCallaghanNew South WalesAustralia
- Hunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Astrid Feentje Feiten
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Yazi D. Ke
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Lars M. Ittner
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Estelle Sontag
- School of Biomedical Sciences and PharmacyUniversity of NewcastleCallaghanNew South WalesAustralia
- Hunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| |
Collapse
|
46
|
Yang G, Tong Y, Wang X, Zhao C, Ba Z, Ahelijiang R, Liu X, Gao W, Zhao Y, Gu Y, Yang J, Xu Y. Guizhi Fuling capsule relieves memory deficits by inhibition of microglial neuroinflammation through blocking JAK2/STAT3 pathway in presenilin1/2 conditional double knockout mice. Front Immunol 2023; 14:1185570. [PMID: 37465679 PMCID: PMC10350565 DOI: 10.3389/fimmu.2023.1185570] [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: 03/13/2023] [Accepted: 06/09/2023] [Indexed: 07/20/2023] Open
Abstract
Chronic neuroinflammation has been regarded as an important part of the pathological initiation of Alzheimer's disease (AD), which is associated with the regulation of microglial activation. Preventing microglial activation to inhibit neuroinflammation may become a potential target for the treatment of neurodegenerative diseases. Guizhi Fuling capsule (GZFL) has a strong repression on inflammatory responses. Here, the presenilin1/2 conditional double knockout (PS cDKO) mice, a well-established mouse model of AD, were divided into: WT mice (WT), WT mice+GZFL (WT+GZFL), PS cDKO mice (cDKO), and PS cDKO mice+GZFL (cDKO+GZFL). Mice in the WT+GZFL and cDKO+GZFL group were fed standard chow containing 2000 ppm GZFL for 90 days. After 60 days of GZFL treatment, mice were given to behavioral tests for 30 days in order to explore the effects of GZFL on cognitive and motor function. Then, mice were sacrificed for examining the effects of GZFL on inflammation. Furthermore, primary microglia were obtained from neonatal Sprague-Dawley rats and pretreated with or without GZFL (50 μg/ml) for 1 h in the absence or presence of lipopolysaccharide (LPS) (100 ng/ml) stimulation to speculate whether the underlying mechanism of GZFL's anti-inflammatory potential was closely associated with Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway. Our findings indicated that GZFL has the ability to alleviate memory deficits in PS cDKO mice, which attributes to the improvement of neuroinflammation by inhibiting microglial activation and the levels of pro-inflammatory mediators. In addition, GZFL could inverse the tau hyperphosphorylation and the lessened expression of synaptic proteins in hippocampus of PS cDKO mice. Furthermore, GZFL prevented LPS-induced neuroinflammatory responses in primary microglia by decreasing the levels of pro-inflammatory mediators. It is noteworthy that therapeutic effects of GZFL on memory impairment are depended on the inhibition of neuroinflammatory responses by the blockage of JAK2/STAT3 signaling pathway. Taken together, GZFL may be an effective compound Chinese medicine for the improvement and postponement of neurodegenerative progression in AD.
Collapse
Affiliation(s)
- Guang Yang
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuting Tong
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xingyu Wang
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenyi Zhao
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zongtao Ba
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Reaila Ahelijiang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinjuan Liu
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Waimao Gao
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Zhao
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yining Gu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Yangzhi Rehabilitation Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianmei Yang
- Department of Traditional Chinese Medicine, Shanghai Xuhui District Central Hospital, Shanghai, China
| | - Ying Xu
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
47
|
Hurtle BT, Xie L, Donnelly CJ. Disrupting pathologic phase transitions in neurodegeneration. J Clin Invest 2023; 133:e168549. [PMID: 37395272 DOI: 10.1172/jci168549] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023] Open
Abstract
Solid-like protein deposits found in aged and diseased human brains have revealed a relationship between insoluble protein accumulations and the resulting deficits in neurologic function. Clinically diverse neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, frontotemporal lobar degeneration, and amyotrophic lateral sclerosis, exhibit unique and disease-specific biochemical protein signatures and abnormal protein depositions that often correlate with disease pathogenesis. Recent evidence indicates that many pathologic proteins assemble into liquid-like protein phases through the highly coordinated process of liquid-liquid phase separation. Over the last decade, biomolecular phase transitions have emerged as a fundamental mechanism of cellular organization. Liquid-like condensates organize functionally related biomolecules within the cell, and many neuropathology-associated proteins reside within these dynamic structures. Thus, examining biomolecular phase transitions enhances our understanding of the molecular mechanisms mediating toxicity across diverse neurodegenerative diseases. This Review explores the known mechanisms contributing to aberrant protein phase transitions in neurodegenerative diseases, focusing on tau and TDP-43 proteinopathies and outlining potential therapeutic strategies to regulate these pathologic events.
Collapse
Affiliation(s)
- Bryan T Hurtle
- Center for Neuroscience at the University of Pittsburgh Graduate Program
- Medical Scientist Training Program, University of Pittsburgh; and
- LiveLikeLou Center for ALS Research at the University of Pittsburgh Brain Institute; Pittsburgh, Pennsylvania, USA
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Longxin Xie
- LiveLikeLou Center for ALS Research at the University of Pittsburgh Brain Institute; Pittsburgh, Pennsylvania, USA
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- School of Medicine, Tsinghua University, Beijing, China
| | - Christopher J Donnelly
- Center for Neuroscience at the University of Pittsburgh Graduate Program
- Medical Scientist Training Program, University of Pittsburgh; and
- LiveLikeLou Center for ALS Research at the University of Pittsburgh Brain Institute; Pittsburgh, Pennsylvania, USA
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
48
|
Yao Y, Chang Y, Li S, Zhu J, Wu Y, Jiang X, Li L, Liu R, Ma R, Li G. Complement C3a Receptor Antagonist Alleviates Tau Pathology and Ameliorates Cognitive Deficits in P301S Mice. Brain Res Bull 2023:110685. [PMID: 37330021 DOI: 10.1016/j.brainresbull.2023.110685] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023]
Abstract
Human tauopathies, including Alzheimer's disease (AD), are a major class of neurodegenerative diseases characterized by intracellular deposition of pathological hyperphosphorylated forms of Tau protein. Complement system is composed of many proteins, which form a complex regulatory network to modulate the immune activity in the brain. Emerging studies have demonstrated a critical role of complement C3a receptor (C3aR) in the development of tauopathy and AD. The underlying mechanisms by which C3aR activation mediates tau hyperphosphorylation in tauopathies, however, remains largely unknown. Here, we observed that the expression of C3aR is upregulated in the brains of P301S mice - a mouse model of tauopathy and AD. Pharmacologic blockade of C3aR ameliorates synaptic integrity and reduced tau hyperphosphorylation in P301S mice. Besides, the administration of C3aR antagonist (C3aRA: SB 290157) improved spatial memory as tested in the Morris water maze. Moreover, C3a receptor antagonist inhibited tau hyperphosphorylation by regulating p35/CDK5 signaling. In summary, results suggest that the C3aR plays an essential role in the accumulation of hyperphosphorylated Tau and behavioral deficits in P301S mice. C3aR could be a feasible therapeutic target for the treatment of tauopathy disorders, including AD. AVAILABILITY OF DATA AND MATERIALS: The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Yi Yao
- Department of Neurology, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yanmin Chang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jiahui Zhu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yanqing Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xingjun Jiang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lulu Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ruitian Liu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Science, Haidian District, Beijing 100190, China
| | - Rong Ma
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Gang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
49
|
Whitfield JF, Rennie K, Chakravarthy B. Alzheimer's Disease and Its Possible Evolutionary Origin: Hypothesis. Cells 2023; 12:1618. [PMID: 37371088 PMCID: PMC10297544 DOI: 10.3390/cells12121618] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/29/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
The enormous, 2-3-million-year evolutionary expansion of hominin neocortices to the current enormity enabled humans to take over the planet. However, there appears to have been a glitch, and it occurred without a compensatory expansion of the entorhinal cortical (EC) gateway to the hippocampal memory-encoding system needed to manage the processing of the increasing volume of neocortical data converging on it. The resulting age-dependent connectopathic glitch was unnoticed by the early short-lived populations. It has now surfaced as Alzheimer's disease (AD) in today's long-lived populations. With advancing age, processing of the converging neocortical data by the neurons of the relatively small lateral entorhinal cortex (LEC) inflicts persistent strain and high energy costs on these cells. This may result in their hyper-release of harmless Aβ1-42 monomers into the interstitial fluid, where they seed the formation of toxic amyloid-β oligomers (AβOs) that initiate AD. At the core of connectopathic AD are the postsynaptic cellular prion protein (PrPC). Electrostatic binding of the negatively charged AβOs to the positively charged N-terminus of PrPC induces hyperphosphorylation of tau that destroys synapses. The spread of these accumulating AβOs from ground zero is supported by Aβ's own production mediated by target cells' Ca2+-sensing receptors (CaSRs). These data suggest that an early administration of a strongly positively charged, AβOs-interacting peptide or protein, plus an inhibitor of CaSR, might be an effective AD-arresting therapeutic combination.
Collapse
Affiliation(s)
- James F. Whitfield
- Human Health Therapeutics, National Research Council, Ottawa, ON K1A 0R6, Canada
| | | | | |
Collapse
|
50
|
Martin SC, Joyce KK, Harper KM, Nikolova VD, Cohen TJ, Moy SS, Diering GH. Sleep disruption precedes forebrain synaptic Tau burden and contributes to cognitive decline in a sex-dependent manner in the P301S Tau transgenic mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544101. [PMID: 37333395 PMCID: PMC10274785 DOI: 10.1101/2023.06.07.544101] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Background Sleep is an essential process that supports brain health and cognitive function in part through the modification of neuronal synapses. Sleep disruption, and impaired synaptic processes, are common features in neurodegenerative diseases, including Alzheimer's disease (AD). However, the casual role of sleep disruption in disease progression is not clear. Neurofibrillary tangles, made from hyperphosphorylated and aggregated Tau protein, form one of the major hallmark pathologies seen in AD and contribute to cognitive decline, synapse loss and neuronal death.Tau has been shown to aggregate in synapses which may impair restorative synapse processes occurring during sleep. However, it remains unclear how sleep disruption and synaptic Tau pathology interact to drive cognitive decline. It is also unclear whether the sexes show differential vulnerability to the effects of sleep loss in the context of neurodegeneration. Methods We used a piezoelectric home-cage monitoring system to measure sleep behavior in 3-11month-old transgenic hTau P301S Tauopathy model mice (PS19) and littermate controls of both sexes. Subcellular fractionation and Western blot was used to examine Tau pathology in mouse forebrain synapse fractions. To examine the role of sleep disruption in disease progression, mice were exposed to acute or chronic sleep disruption. The Morris water maze test was used to measure spatial learning and memory performance. Results PS19 mice exhibited a selective loss of sleep during the dark phase, referred to as hyperarousal, as an early symptom with an onset of 3months in females and 6months in males. At 6months, forebrain synaptic Tau burden did not correlate with sleep measures and was not affected by acute or chronic sleep disruption. Chronic sleep disruption accelerated the onset of decline of hippocampal spatial memory in PS19 males, but not females. Conclusions Dark phase hyperarousal is an early symptom in PS19 mice that precedes robust Tau aggregation. We find no evidence that sleep disruption is a direct driver of Tau pathology in the forebrain synapse. However, sleep disruption synergized with Tau pathology to accelerate the onset of cognitive decline in males. Despite the finding that hyperarousal appears earlier in females, female cognition was resilient to the effects of sleep disruption.
Collapse
|