1
|
Sadleir KR, Gomez KP, Edwards AE, Patel AJ, Ley ML, Khatri AW, Guo J, Mahesh S, Watkins EA, Popovic J, Karunakaran DKP, Prokopenko D, Tanzi RE, Bustos B, Lubbe SJ, Demonbruen AR, McNally EM, Vassar R. Annexin A6 membrane repair protein protects against amyloid-induced dystrophic neurites and tau phosphorylation in Alzheimer's disease model mice. Acta Neuropathol 2025; 149:51. [PMID: 40411591 PMCID: PMC12103342 DOI: 10.1007/s00401-025-02888-1] [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: 02/02/2025] [Revised: 04/23/2025] [Accepted: 04/30/2025] [Indexed: 05/26/2025]
Abstract
In Alzheimer's disease, accumulation of amyloid-β (Aβ) peptide is thought to cause formation of neurofibrillary tangles composed of hyperphosphorylated tau protein, which correlates with neuronal loss and cognitive impairment, but the mechanism linking Aβ and tau pathologies is unknown. Dystrophic neurites, which surround Aβ plaques and accumulate phosphorylated tau and other proteins, may play a role in seeding and spreading of pathologic tau. Here, we investigate the novel hypothesis that improved membrane repair capacity decreases dystrophic neurite formation by protecting axons from Aβ-induced membrane damage. Using a ratiometric calcium sensor and a FRET-based calpain cleavage sensor, we demonstrate that dystrophic neurites in 5XFAD mice have elevated resting calcium levels and calpain activity because of putative membrane damage. Annexin A6, a plasma membrane repair in muscle and neurons, is present at plasma membrane of neurons and dystrophic neurites in murine and human brains. Overexpression of annexin A6 in brains of 5XFAD mice decreased size and quantity of dystrophic neurites and accumulation of phospho-tau181, an early biomarker of amyloid pathology. Phospho-tau231, another early amyloid biomarker, and phosphorylated of tau kinases, c-jun N-terminal kinase (JNK) and Calmodulin Kinase II (CaMKII) accumulate in dystrophic neurites in the brains of amyloid pathology mice and humans with AD, suggesting that dystrophic neurites are sites of active tau phosphorylation. Overexpression of dominant-negative annexin A6 in 5XFAD mice increased dystrophic neurites and phospho-tau181. Intracerebral injection of recombinant annexin A6 in 5XFAD and APP-NLGF knock-in mice resulted in localization of recombinant A6 to membranes of dystrophic neurites, suggesting therapeutic potential of recombinant annexin A6 for AD. In conclusion, dystrophic neurites have Aβ-induced membrane damage characterized by calcium elevation, calpain activation, and accumulation of tau kinases and phosphorylated tau. Overexpression of annexin A6 reduces dystrophic neurites and phospho-tau accumulation, suggesting that annexin A6-mediated membrane repair may represent a novel therapeutic approach for AD.
Collapse
Affiliation(s)
- Katherine R Sadleir
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| | - Karen P Gomez
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Abigail E Edwards
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Armana J Patel
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Makenna L Ley
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Ammaarah W Khatri
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Joanna Guo
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Shreya Mahesh
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Elyse A Watkins
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jelena Popovic
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | | | - Dmitry Prokopenko
- Department of Neurology, Genetics and Aging Research Unit and the McCance Center for Brain Health, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Rudolph E Tanzi
- Department of Neurology, Genetics and Aging Research Unit and the McCance Center for Brain Health, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Bernabe Bustos
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Simpson Querrey Center for Neurogenetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Steven J Lubbe
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Simpson Querrey Center for Neurogenetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Alexis R Demonbruen
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Robert Vassar
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
2
|
Prakash P, Manchanda P, Paouri E, Bisht K, Sharma K, Rajpoot J, Wendt V, Hossain A, Wijewardhane PR, Randolph CE, Chen Y, Stanko S, Gasmi N, Gjojdeshi A, Card S, Fine J, Jethava KP, Clark MG, Dong B, Ma S, Crockett A, Thayer EA, Nicolas M, Davis R, Hardikar D, Allende D, Prayson RA, Zhang C, Davalos D, Chopra G. Amyloid-β induces lipid droplet-mediated microglial dysfunction via the enzyme DGAT2 in Alzheimer's disease. Immunity 2025:S1074-7613(25)00192-X. [PMID: 40393454 DOI: 10.1016/j.immuni.2025.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/30/2024] [Accepted: 04/22/2025] [Indexed: 05/22/2025]
Abstract
Microglial phagocytosis genes have been linked to increased risk for Alzheimer's disease (AD), but the mechanisms translating genetic association to cellular dysfunction remain unknown. Here, we showed that microglia formed lipid droplets (LDs) upon amyloid-β (Aβ) exposure and that LD loads increased with proximity to amyloid plaques in brains from individuals with AD and the 5xFAD mouse model. LD-laden microglia exhibited defects in Aβ phagocytosis, and unbiased lipidomic analyses identified a parallel decrease in free fatty acids (FFAs) and increase in triacylglycerols (TGs) as the key metabolic transition underlying LD formation. Diacylglycerol O-acyltransferase 2 (DGAT2)-a key enzyme that converts FFAs to TGs-promoted microglial LD formation and was increased in mouse 5xFAD and human AD brains. Pharmacologically targeting DGAT2 improved microglial uptake of Aβ and reduced plaque load and neuronal damage in 5xFAD mice. These findings identify a lipid-mediated mechanism underlying microglial dysfunction that could become a therapeutic target for AD.
Collapse
Affiliation(s)
- Priya Prakash
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Palak Manchanda
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Evi Paouri
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Kanchan Bisht
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Kaushik Sharma
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Jitika Rajpoot
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Victoria Wendt
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Ahad Hossain
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | | | - Caitlin E Randolph
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Yihao Chen
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Sarah Stanko
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nadia Gasmi
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Anxhela Gjojdeshi
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Sophie Card
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Jonathan Fine
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Krupal P Jethava
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Matthew G Clark
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Bin Dong
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Seohee Ma
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Alexis Crockett
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Elizabeth A Thayer
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Marlo Nicolas
- Division of Pathology, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Ryann Davis
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Dhruv Hardikar
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Daniela Allende
- Division of Pathology, Cleveland Clinic, Cleveland, OH 44106, USA
| | | | - Chi Zhang
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Dimitrios Davalos
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Gaurav Chopra
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA; Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
3
|
Pan W, Teng Y, Han X, Liu S, Pang X, Wang L, Zhao M. Value of blood neural cell-derived small extracellular vesicles in the diagnosis and prediction of Alzheimer's disease: A systematic revie. J Prev Alzheimers Dis 2025:100193. [PMID: 40316481 DOI: 10.1016/j.tjpad.2025.100193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/17/2025] [Accepted: 04/19/2025] [Indexed: 05/04/2025]
Abstract
Blood neural cell-derived small extracellular vesicles (sEVs) can directly reflect changes in brain tissue and are easier to obtain than cerebrospinal fluid. This article systematically reviews the alterations of proteins and miRNAs from neural cell-derived sEVs in patients with Alzheimer's disease (AD), and summarizes the biomarkers with clinical diagnostic and predictive value. PubMed, Web of Science, Embase, and Cochrane Library were searched for studies in blood neural cell-derived sEVs in AD patients up to May 2024. According to the inclusion and exclusion criteria, the literature was screened, the information was extracted and the quality was evaluated. Proteins and miRNAs from neural cell-derived sEVs were classified and summarized, focusing on target molecules with high diagnostic and predictive values for AD. A final 34 articles reporting 5601 participants were included. In cross-sectional studies, Aβ- and Tau-related proteins (Aβ42, Aβ42/40, p-S396-Tau, p-Tau181), p-S312-IRS-1, and cathepsin D were increased, conversely, synaptic proteins (neurogranin, synaptotagmin, synaptophysin, synaptopodin, NMDAR2A) and REST were decreased in blood neuron-derived sEVs (NDsEVs) of patients with AD. While miR-29c-3p was increased in blood NDsEVs and glial cell-derived sEVs. Each of these proteins and miRNAs demonstrated high AD diagnostic value. Additionally, blood astrocyte-derived sEVs (ADsEVs) showed increased complement effector proteins and decreased complement regulatory proteins with a moderate diagnostic value. In longitudinal cohort studies, three composite models displayed high predictive efficacy for early AD prediction, and could predict the occurrence of AD within 1-10 years. Therefore, Aβ- and Tau-related proteins, synaptic proteins, and miRNA in blood neural cell-derived sEVs demonstrate high AD diagnostic and predictive values serving as important biomarkers. Especially, synaptic proteins showed significant changes in the early clinical stage, which has early predictive value.
Collapse
Affiliation(s)
- Weibing Pan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Teng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaowan Han
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shaojiao Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xingxue Pang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Mingjing Zhao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
4
|
Cabral‐Miranda F, Araujo APB, Medinas DB, Gomes FCA. Astrocytic Hevin/SPARCL-1 Regulates Cognitive Decline in Pathological and Normal Brain Aging. Aging Cell 2025; 24:e14493. [PMID: 39935382 PMCID: PMC12074016 DOI: 10.1111/acel.14493] [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: 07/22/2024] [Revised: 12/20/2024] [Accepted: 01/08/2025] [Indexed: 02/13/2025] Open
Abstract
Dementia, characterized by loss of cognitive abilities in the elderly, poses a significant global health challenge. This study explores the role of astrocytes, one of most representative glial cells in the brain, in mitigating cognitive decline. Specifically, we investigated the impact of Hevin (also known as SPARC-like1/SPARCL-1), a secreted glycoprotein, on cognitive decline in both normal and pathological brain aging. By using adeno-associated viruses, we overexpressed Hevin in hippocampal astrocytes of middle-aged APP/PSEN mice, an established Alzheimer's disease (AD) model. Results demonstrated that Hevin overexpression attenuates cognitive decline, as evidenced by cognitive tests, increased pre- and postsynaptic markers colocalization, and altered expression of synaptic mediators, as revealed by proteomic profiling. Importantly, Hevin overexpression did not influence the deposition of Aβ plaques in the hippocampus, a hallmark of AD pathology. Furthermore, the study extended its findings to middle-aged wild-type animals, revealing improved cognitive performance following astrocytic Hevin overexpression. In conclusion, our results propose astrocytic Hevin as a potential therapeutic target for age-associated cognitive decline.
Collapse
Affiliation(s)
- Felipe Cabral‐Miranda
- Institute of Biomedical SciencesUniversidade Federal do Rio de JaneiroRio de JaneiroBrazil
| | | | - Danilo Bilches Medinas
- Department of Biochemistry, Institute of ChemistryUniversity of São PauloSão PauloBrazil
| | | |
Collapse
|
5
|
Alkhatabi HA, Pushparaj PN. Untangling the complex mechanisms associated with Alzheimer's disease in elderly patients using high-throughput RNA sequencing data and next-generation knowledge discovery methods: Focus on potential gene signatures and drugs for dementia. Heliyon 2025; 11:e41266. [PMID: 39834440 PMCID: PMC11743088 DOI: 10.1016/j.heliyon.2024.e41266] [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: 02/21/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 01/22/2025] Open
Abstract
Objectives Alzheimer's disease (AD) is a complex neurodegenerative disorder that primarily affects elderly individuals. This study aimed to elucidate the intricate mechanisms underlying AD in elderly patients compared with healthy aged individuals using high-throughput RNA sequencing (RNA-seq) data and next-generation knowledge discovery methods (NGKD), with a focus on identifying potential therapeutic agents. Methods High-throughput RNA-seq data were obtained from the Gene Expression Omnibus (GEO) database (accession number: GSE104704). These data were derived from healthy and diseased human brains (eight young healthy brains [young], 10 aged healthy brains [Old], and 12 aged diseased brains [AD]). We used NGKD tools such as GEO RNA-seq Experiments Interactive Navigator (GREIN) to obtain differentially expressed genes (DEGs) by comparing the AD versus Old RNA-seq data and further filtered and normalized to obtain differentially regulated Kyoto Encyclopedia of Genes and Genomes (KEGG), Reactome and Panther pathways using ExpressAnalyst tool. Besides, WebGestalt was used to identify differentially regulated Gene Ontologies (GO) and the pre-ranked Gene Set Enrichment Analysis (GSEA) was performed using GSEA software. The X2K web tool was used to infer upstream regulator networks and X2K Appyter tool for obtaining transcription factors (TFs) and kinase network information. LFW1000 and L1000CDS2 tools were used to identify specific drugs that reverse AD-associated gene signatures in elderly patients. Results Our study revealed significant downregulation of pathways related to neuroactive receptor-ligand interaction, synaptic vesicle cycle, and neuronal system in elderly individuals with AD. GO analysis showed negative enrichment of functions related to cognition, potassium ion transport, receptor-ligand activity, SNARE binding, and primary lysosomes. The transcription factors SUZ12 and REST, along with increased MAPK signaling, were identified as key regulators of downregulated genes. Several drugs and natural products, including dihydroergocristine, mepacrine, gedunin, amlodipine, and disulfiram have been identified as potential therapeutic agents for reversing AD-associated gene signatures. Conclusions This comprehensive analysis of AD in elderly individuals using RNA-seq data and NGKD tools revealed multiple differentially regulated pathways, gene signatures, and potential drugs for dementia treatment. These findings highlight the complex molecular mechanisms underlying AD and provide insights into potential therapeutic strategies. Further research is needed to validate these findings and to develop personalized treatment approaches for AD in elderly patients.
Collapse
Affiliation(s)
- Hind A. Alkhatabi
- Department of Biological Science, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Peter Natesan Pushparaj
- Institute of Genomic Medicine Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| |
Collapse
|
6
|
Hopkins PCR, Troakes C, King A, Tear G. Transmembrane and coiled-coil 2 associates with Alzheimer's disease pathology in the human brain. Brain Pathol 2025; 35:e13290. [PMID: 39084860 PMCID: PMC11669416 DOI: 10.1111/bpa.13290] [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: 01/09/2024] [Accepted: 07/05/2024] [Indexed: 08/02/2024] Open
Abstract
Transmembrane and coiled-coil 2 (TMCC2) is a human orthologue of the Drosophila gene dementin, mutant alleles of which cause neurodegeneration with features of Alzheimer's disease (AD). TMCC2 and Dementin further have an evolutionarily conserved interaction with the amyloid protein precursor (APP), a protein central to AD pathogenesis. To investigate if human TMCC2 might also participate in mechanisms of neurodegeneration, we examined TMCC2 expression in late onset AD human brain and age-matched controls, familial AD cases bearing a mutation in APP Val717, and Down syndrome AD. Consistent with previous observations of complex formation between TMCC2 and APP in the rat brain, the dual immunocytochemistry of control human temporal cortex showed highly similar distributions of TMCC2 and APP. In late onset AD cases stratified by APOE genotype, TMCC2 immunoreactivity was associated with dense core senile plaques and adjacent neuronal dystrophies, but not with Aβ surrounding the core, diffuse Aβ plaques or tauopathy. In Down syndrome AD, we observed in addition TMCC2-immunoreactive and methoxy-X04-positive pathological features that were morphologically distinct from those seen in the late onset and familial AD cases, suggesting enhanced pathological alteration of TMCC2 in Down syndrome AD. At the protein level, western blots of human brain extracts revealed that human brain-derived TMCC2 exists as at least three isoforms, the relative abundance of which varied between the temporal gyrus and cerebellum and was influenced by APOE and/or dementia status. Our findings thus implicate human TMCC2 in AD via its interactions with APP, its association with dense core plaques, as well as its alteration in Down syndrome AD.
Collapse
Affiliation(s)
| | - Claire Troakes
- London Neurodegenerative Diseases Brain BankInstitute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUK
| | - Andrew King
- London Neurodegenerative Diseases Brain BankInstitute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUK
| | - Guy Tear
- Centre for Developmental NeurobiologyKing's College LondonLondonUK
| |
Collapse
|
7
|
Feole M, Pozo Devoto VM, Dragišić N, Arnaiz C, Bianchelli J, Texlová K, Kovačovicova K, Novotny JS, Havas D, Falzone TL, Stokin GB. Swedish Alzheimer's disease variant perturbs activity of retrograde molecular motors and causes widespread derangement of axonal transport pathways. J Biol Chem 2024; 300:107137. [PMID: 38447793 PMCID: PMC10997842 DOI: 10.1016/j.jbc.2024.107137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
Experimental studies in flies, mice, and humans suggest a significant role of impaired axonal transport in the pathogenesis of Alzheimer's disease (AD). The mechanisms underlying these impairments in axonal transport, however, remain poorly understood. Here we report that the Swedish familial AD mutation causes a standstill of the amyloid precursor protein (APP) in the axons at the expense of its reduced anterograde transport. The standstill reflects the perturbed directionality of the axonal transport of APP, which spends significantly more time traveling in the retrograde direction. This ineffective movement is accompanied by an enhanced association of dynactin-1 with APP, which suggests that reduced anterograde transport of APP is the result of enhanced activation of the retrograde molecular motor dynein by dynactin-1. The impact of the Swedish mutation on axonal transport is not limited to the APP vesicles since it also reverses the directionality of a subset of early endosomes, which become enlarged and aberrantly accumulate in distal locations. In addition, it also reduces the trafficking of lysosomes due to their less effective retrograde movement. Altogether, our experiments suggest a pivotal involvement of retrograde molecular motors and transport in the mechanisms underlying impaired axonal transport in AD and reveal significantly more widespread derangement of axonal transport pathways in the pathogenesis of AD.
Collapse
Affiliation(s)
- Monica Feole
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St Anne's University Hospital, Brno, Czech Republic; Faculty of Medicine, Department of Biology, Masaryk University, Brno, Czech Republic; School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
| | - Victorio M Pozo Devoto
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St Anne's University Hospital, Brno, Czech Republic
| | - Neda Dragišić
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St Anne's University Hospital, Brno, Czech Republic
| | - Cayetana Arnaiz
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA-CONICET-MPSP), Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Julieta Bianchelli
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA-CONICET-MPSP), Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Kateřina Texlová
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St Anne's University Hospital, Brno, Czech Republic; PsychoGenics, Paramus, New Jersey, USA
| | | | - Jan S Novotny
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St Anne's University Hospital, Brno, Czech Republic; Institute for Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czech Republic
| | | | - Tomas L Falzone
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA-CONICET-MPSP), Partner Institute of the Max Planck Society, Buenos Aires, Argentina; Instituto de Biología Celular y Neurociencia IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gorazd B Stokin
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St Anne's University Hospital, Brno, Czech Republic; Institute for Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czech Republic; Division of Neurology, University Medical Centre, Ljubljana, Slovenia; Department of Neurosciences, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
8
|
Jagadeesan N, Roules GC, Chandrashekar DV, Yang J, Kolluru S, Sumbria RK. Modulation of hippocampal protein expression by a brain penetrant biologic TNF-α inhibitor in the 3xTg Alzheimer's disease mice. J Transl Med 2024; 22:291. [PMID: 38500108 PMCID: PMC10946165 DOI: 10.1186/s12967-024-05008-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: 09/29/2023] [Accepted: 02/19/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Biologic TNF-α inhibitors (bTNFIs) can block cerebral TNF-α in Alzheimer's disease (AD) if these macromolecules can cross the blood-brain barrier (BBB). Thus, a model bTNFI, the extracellular domain of type II TNF-α receptor (TNFR), which can bind to and sequester TNF-α, was fused with a mouse transferrin receptor antibody (TfRMAb) to enable brain delivery via BBB TfR-mediated transcytosis. Previously, we found TfRMAb-TNFR to be protective in a mouse model of amyloidosis (APP/PS1) and tauopathy (PS19), and herein we investigated its effects in mice that combine both amyloidosis and tauopathy (3xTg-AD). METHODS Eight-month-old female 3xTg-AD mice were injected intraperitoneally with saline (n = 11) or TfRMAb-TNFR (3 mg/kg; n = 11) three days per week for 12 weeks. Age-matched wild-type (WT) mice (n = 9) were treated similarly with saline. Brains were processed for immunostaining and high-resolution multiplex NanoString GeoMx spatial proteomics. RESULTS We observed regional differences in proteins relevant to Aβ, tau, and neuroinflammation in the hippocampus of 3xTg-AD mice compared with WT mice. From 64 target proteins studied using spatial proteomics, a comparison of the Aβ-plaque bearing vs. plaque-free regions in the 3xTg-AD mice yielded 39 differentially expressed proteins (DEP) largely related to neuroinflammation (39% of DEP) and Aβ and tau pathology combined (31% of DEP). Hippocampal spatial proteomics revealed that the majority of the proteins modulated by TfRMAb-TNFR in the 3xTg-AD mice were relevant to microglial function (⁓ 33%). TfRMAb-TNFR significantly reduced mature Aβ plaques and increased Aβ-associated microglia around larger Aβ deposits in the 3xTg-AD mice. Further, TfRMAb-TNFR increased mature Aβ plaque-associated microglial TREM2 in 3xTg-AD mice. CONCLUSION Overall, despite the low visual Aβ load in the 11-month-old female 3xTg-AD mice, our results highlight region-specific AD-relevant DEP in the hippocampus of these mice. Chronic TfRMAb-TNFR dosing modulated several DEP involved in AD pathology and showed a largely microglia-centric mechanism of action in the 3xTg-AD mice.
Collapse
Affiliation(s)
- Nataraj Jagadeesan
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - G Chuli Roules
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - Devaraj V Chandrashekar
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - Joshua Yang
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - Sanjana Kolluru
- Rancho Cucamonga High School, 11801 Lark Dr, Rancho Cucamonga, CA, 91701, USA
| | - Rachita K Sumbria
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA.
- Department of Neurology, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
9
|
Maniv I, Sarji M, Bdarneh A, Feldman A, Ankawa R, Koren E, Magid-Gold I, Reis N, Soteriou D, Salomon-Zimri S, Lavy T, Kesselman E, Koifman N, Kurz T, Kleifeld O, Michaelson D, van Leeuwen FW, Verheijen BM, Fuchs Y, Glickman MH. Altered ubiquitin signaling induces Alzheimer's disease-like hallmarks in a three-dimensional human neural cell culture model. Nat Commun 2023; 14:5922. [PMID: 37739965 PMCID: PMC10516951 DOI: 10.1038/s41467-023-41545-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 09/08/2023] [Indexed: 09/24/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by toxic protein accumulation in the brain. Ubiquitination is essential for protein clearance in cells, making altered ubiquitin signaling crucial in AD development. A defective variant, ubiquitin B + 1 (UBB+1), created by a non-hereditary RNA frameshift mutation, is found in all AD patient brains post-mortem. We now detect UBB+1 in human brains during early AD stages. Our study employs a 3D neural culture platform derived from human neural progenitors, demonstrating that UBB+1 alone induces extracellular amyloid-β (Aβ) deposits and insoluble hyperphosphorylated tau aggregates. UBB+1 competes with ubiquitin for binding to the deubiquitinating enzyme UCHL1, leading to elevated levels of amyloid precursor protein (APP), secreted Aβ peptides, and Aβ build-up. Crucially, silencing UBB+1 expression impedes the emergence of AD hallmarks in this model system. Our findings highlight the significance of ubiquitin signalling as a variable contributing to AD pathology and present a nonclinical platform for testing potential therapeutics.
Collapse
Affiliation(s)
- Inbal Maniv
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Mahasen Sarji
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Anwar Bdarneh
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Alona Feldman
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Roi Ankawa
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Elle Koren
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Inbar Magid-Gold
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Noa Reis
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Despina Soteriou
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Shiran Salomon-Zimri
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences, The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Tali Lavy
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Ellina Kesselman
- The Wolfson Department of Chemical Engineering, The Technion Center for Electron Microscopy of Soft Matter, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Naama Koifman
- The Wolfson Department of Chemical Engineering, The Technion Center for Electron Microscopy of Soft Matter, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Thimo Kurz
- School of Molecular Biosciences, University of Glasgow, Glasgow, G12 8QQ, Scotland, UK
| | - Oded Kleifeld
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Daniel Michaelson
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences, The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Fred W van Leeuwen
- Department of Neuroscience, Maastricht University, 6229 ER, Maastricht, the Netherlands
| | - Bert M Verheijen
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
- Department of Neuroscience, Maastricht University, 6229 ER, Maastricht, the Netherlands
| | - Yaron Fuchs
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel.
- Augmanity, Rehovot, 7670308, Israel.
| | - Michael H Glickman
- Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel.
| |
Collapse
|
10
|
Jang J, Yeo S, Baek S, Jung HJ, Lee MS, Choi SH, Choe Y. Abnormal accumulation of extracellular vesicles in hippocampal dystrophic axons and regulation by the primary cilia in Alzheimer's disease. Acta Neuropathol Commun 2023; 11:142. [PMID: 37667395 PMCID: PMC10478284 DOI: 10.1186/s40478-023-01637-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/15/2023] [Indexed: 09/06/2023] Open
Abstract
Dystrophic neurites (DNs) are abnormal axons and dendrites that are swollen or deformed in various neuropathological conditions. In Alzheimer's disease (AD), DNs play a crucial role in impairing neuronal communication and function, and they may also contribute to the accumulation and spread of amyloid beta (Aβ) in the brain of AD patients. However, it is still a challenge to understand the DNs of specific neurons that are vulnerable to Aβ in the pathogenesis of AD. To shed light on the development of radiating DNs, we examined enriched dystrophic hippocampal axons in a mouse model of AD using a three-dimensional rendering of projecting neurons. We employed the anterograde spread of adeno-associated virus (AAV)1 and conducted proteomic analysis of synaptic compartments obtained from hippocampo-septal regions. Our findings revealed that DNs were formed due to synaptic loss at the axon terminals caused by the accumulation of extracellular vesicle (EV). Abnormal EV-mediated transport and exocytosis were identified in association with primary cilia, indicating their involvement in the accumulation of EVs at presynaptic terminals. To further address the regulation of DNs by primary cilia, we conducted knockdown of the Ift88 gene in hippocampal neurons, which impaired EV-mediated secretion of Aβ and promoted accumulation of axonal spheroids. Using single-cell RNA sequencing, we identified the septal projecting hippocampal somatostatin neurons (SOM) as selectively vulnerable to Aβ with primary cilia dysfunction and vesicle accumulation. Our study suggests that DNs in AD are initiated by the ectopic accumulation of EVs at the neuronal axon terminals, which is affected by neuronal primary cilia.
Collapse
Affiliation(s)
| | - Seungeun Yeo
- Korea Brain Research Institute, Daegu, 41068, Korea
| | | | | | - Mi Suk Lee
- Korea Brain Research Institute, Daegu, 41068, Korea
| | | | - Youngshik Choe
- Korea Brain Research Institute, Daegu, 41068, Korea.
- , Daegu, Korea.
| |
Collapse
|
11
|
Lenz M, Eichler A, Kruse P, Galanis C, Kleidonas D, Andrieux G, Boerries M, Jedlicka P, Müller U, Deller T, Vlachos A. The Amyloid Precursor Protein Regulates Synaptic Transmission at Medial Perforant Path Synapses. J Neurosci 2023; 43:5290-5304. [PMID: 37369586 PMCID: PMC10359033 DOI: 10.1523/jneurosci.1824-22.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 05/20/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The perforant path provides the primary cortical excitatory input to the hippocampus. Because of its important role in information processing and coding, entorhinal projections to the dentate gyrus have been studied in considerable detail. Nevertheless, synaptic transmission between individual connected pairs of entorhinal stellate cells and dentate granule cells remains to be characterized. Here, we have used mouse organotypic entorhino-hippocampal tissue cultures of either sex, in which the entorhinal cortex (EC) to dentate granule cell (GC; EC-GC) projection is present, and EC-GC pairs can be studied using whole-cell patch-clamp recordings. By using cultures of wild-type mice, the properties of EC-GC synapses formed by afferents from the lateral and medial entorhinal cortex were compared, and differences in short-term plasticity were identified. As the perforant path is severely affected in Alzheimer's disease, we used tissue cultures of amyloid precursor protein (APP)-deficient mice to examine the role of APP at this synapse. APP deficiency altered excitatory neurotransmission at medial perforant path synapses, which was accompanied by transcriptomic and ultrastructural changes. Moreover, presynaptic but not postsynaptic APP deletion through the local injection of Cre-expressing adeno-associated viruses in conditional APPflox/flox tissue cultures increased the neurotransmission efficacy at perforant path synapses. In summary, these data suggest a physiological role for presynaptic APP at medial perforant path synapses that may be adversely affected under altered APP processing conditions.SIGNIFICANCE STATEMENT The hippocampus receives input from the entorhinal cortex via the perforant path. These projections to hippocampal dentate granule cells are of utmost importance for learning and memory formation. Although there is detailed knowledge about perforant path projections, the functional synaptic properties at the level of individual connected pairs of neurons are not well understood. In this study, we investigated the role of APP in mediating functional properties and transmission rules in individually connected neurons using paired whole-cell patch-clamp recordings and genetic tools in organotypic tissue cultures. Our results show that presynaptic APP expression limits excitatory neurotransmission via the perforant path, which could be compromised in pathologic conditions such as Alzheimer's disease.
Collapse
Affiliation(s)
- Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, 30625 Hannover, Germany
| | - Amelie Eichler
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Pia Kruse
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Christos Galanis
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Dimitrios Kleidonas
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- German Cancer Consortium, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Peter Jedlicka
- Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
- Frankfurt Institute for Advanced Studies, 60438 Frankfurt am Main, Germany
| | - Ulrike Müller
- Institute of Pharmacy and Molecular Biotechnology, Functional Genomics, Ruprecht-Karls University Heidelberg, 69120 Heidelberg, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Center for Basics in Neuromodulation, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
12
|
Hou X, Zhang X, Zou H, Guan M, Fu C, Wang W, Zhang ZR, Geng Y, Chen Y. Differential and substrate-specific inhibition of γ-secretase by the C-terminal region of ApoE2, ApoE3, and ApoE4. Neuron 2023; 111:1898-1913.e5. [PMID: 37040764 DOI: 10.1016/j.neuron.2023.03.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 01/16/2023] [Accepted: 03/17/2023] [Indexed: 04/13/2023]
Abstract
Aberrant low γ-secretase activity is associated with most of the presenilin mutations that underlie familial Alzheimer's disease (fAD). However, the role of γ-secretase in the more prevalent sporadic AD (sAD) remains unaddressed. Here, we report that human apolipoprotein E (ApoE), the most important genetic risk factor of sAD, interacts with γ-secretase and inhibits it with substrate specificity in cell-autonomous manners through its conserved C-terminal region (CT). This ApoE CT-mediated inhibitory activity is differentially compromised in different ApoE isoforms, resulting in an ApoE2 > ApoE3 > ApoE4 potency rank order inversely correlating to their associated AD risk. Interestingly, in an AD mouse model, neuronal ApoE CT migrates to amyloid plaques in the subiculum from other regions and alleviates the plaque burden. Together, our data reveal a hidden role of ApoE as a γ-secretase inhibitor with substrate specificity and suggest that this precision γ-inhibition by ApoE may protect against the risk of sAD.
Collapse
Affiliation(s)
- Xianglong Hou
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Rd., B13, Pudongxinqu, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuexin Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Rd., B13, Pudongxinqu, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huan Zou
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Rd., B13, Pudongxinqu, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingfeng Guan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Rd., B13, Pudongxinqu, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chaoying Fu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Rd., B13, Pudongxinqu, Shanghai 201210, China
| | - Wenyuan Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Rd., B13, Pudongxinqu, Shanghai 201210, China
| | - Zai-Rong Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Rd., B13, Pudongxinqu, Shanghai 201210, China
| | - Yang Geng
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Rd., B13, Pudongxinqu, Shanghai 201210, China.
| | - Yelin Chen
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Rd., B13, Pudongxinqu, Shanghai 201210, China.
| |
Collapse
|
13
|
Zhaoxia W, Chenyu W, ZhuangZhuang Y, Liangliang F, Xue L, Tieyu T. Whole-exome sequencing detected a novel APP variant in a Han-Chinese family with Alzheimer's disease. Mol Biol Rep 2023; 50:5267-5271. [PMID: 37145212 DOI: 10.1007/s11033-023-08400-w] [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: 10/17/2022] [Accepted: 03/23/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is an incurable and debilitating neurodegenerative disease that results in the progressive degeneration and death of nerve cells. Mutations in the APP gene, which encodes an amyloid precursor protein, is the strongest genetic risk factor for sporadic AD. METHODS AND RESULTS We studied the APP gene (NM_000484.3: c.2045A > T; p.E682V) variants carried by members of a family suffering from AD using whole-exome sequencing and Sanger sequencing. CONCLUSION In this study, we identified a new variant of the APP gene (NM_000484.3: c.2045A > T; p.E682V) in members of a family with AD. This provides potential targets for subsequent studies and information that can be used in genetic counselling.
Collapse
Affiliation(s)
- Wang Zhaoxia
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Wang Chenyu
- Department of Cell Biology, The School of Life Sciences, Central South University, Changsha, 410013, China
| | - Yuan ZhuangZhuang
- Department of Cell Biology, The School of Life Sciences, Central South University, Changsha, 410013, China
| | - Fan Liangliang
- Department of Cell Biology, The School of Life Sciences, Central South University, Changsha, 410013, China
| | - Lin Xue
- Department of Neurology, Yangzhou Oriental Hospital, Yangzhou, 225001, Jiangsu, China.
| | - Tang Tieyu
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou, 225001, Jiangsu, China.
| |
Collapse
|
14
|
Ozsan McMillan I, Li JP, Wang L. Heparan sulfate proteoglycan in Alzheimer's disease: aberrant expression and functions in molecular pathways related to amyloid-β metabolism. Am J Physiol Cell Physiol 2023; 324:C893-C909. [PMID: 36878848 PMCID: PMC10069967 DOI: 10.1152/ajpcell.00247.2022] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 02/28/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Currently, there is no effective treatment for AD, as its etiology remains poorly understood. Mounting evidence suggests that the accumulation and aggregation of amyloid-β peptides (Aβ), which constitute amyloid plaques in the brain, is critical for initiating and accelerating AD pathogenesis. Considerable efforts have been dedicated to shedding light on the molecular basis and fundamental origins of the impaired Aβ metabolism in AD. Heparan sulfate (HS), a linear polysaccharide of the glycosaminoglycan family, co-deposits with Aβ in plaques in the AD brain, directly binds and accelerates Aβ aggregation, and mediates Aβ internalization and cytotoxicity. Mouse model studies demonstrate that HS regulates Aβ clearance and neuroinflammation in vivo. Previous reviews have extensively explored these discoveries. Here, this review focuses on the recent advancements in understanding abnormal HS expression in the AD brain, the structural aspects of HS-Aβ interaction, and the molecules involved in modulating Aβ metabolism through HS interaction. Furthermore, this review presents a perspective on the potential effects of abnormal HS expression on Aβ metabolism and AD pathogenesis. In addition, the review highlights the importance of conducting further research to differentiate the spatiotemporal components of HS structure and function in the brain and AD pathogenesis.
Collapse
Affiliation(s)
- Ilayda Ozsan McMillan
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States
- Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology & The Biomedical Center, University of Uppsala, Uppsala, Sweden
- SciLifeLab Uppsala, University of Uppsala, Uppsala, Sweden
| | - Lianchun Wang
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States
- Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States
| |
Collapse
|
15
|
Tian C, Stewart T, Hong Z, Guo Z, Aro P, Soltys D, Pan C, Peskind ER, Zabetian CP, Shaw LM, Galasko D, Quinn JF, Shi M, Zhang J. Blood extracellular vesicles carrying synaptic function- and brain-related proteins as potential biomarkers for Alzheimer's disease. Alzheimers Dement 2023; 19:909-923. [PMID: 35779041 PMCID: PMC9806186 DOI: 10.1002/alz.12723] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Objective and accessible markers for Alzheimer's disease (AD) and other dementias are critically needed. METHODS We identified NMDAR2A, a protein related to synaptic function, as a novel marker of central nervous system (CNS)-derived plasma extracellular vesicles (EVs) and developed a flow cytometry-based technology for detecting such plasma EVs readily. The assay was initially tested in our local cross-sectional study to distinguish AD patients from healthy controls (HCs) or from Parkinson's disease (PD) patients, followed by a validation study using an independent cohort collected from multiple medical centers (the Alzheimer's Disease Neuroimaging Initiative). Cerebrospinal fluid AD molecular signature was used to confirm diagnoses of all AD participants. RESULTS Likely CNS-derived EVs in plasma were significantly reduced in AD compared to HCs in both cohorts. Integrative models including CNS-derived EV markers and AD markers present on EVs reached area under the curve of 0.915 in discovery cohort and 0.810 in validation cohort. DISCUSSION This study demonstrated that robust and rapid analysis of individual neuron-derived synaptic function-related EVs in peripheral blood may serve as a helpful marker of synaptic dysfunction in AD and dementia.
Collapse
Affiliation(s)
- Chen Tian
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Tessandra Stewart
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Zhen Hong
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
- Department of Neurology, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Zhen Guo
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| | - Patrick Aro
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - David Soltys
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Catherine Pan
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Elaine R Peskind
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
- Northwest (VISN-20) Mental Illness, Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Cyrus P. Zabetian
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Leslie M. Shaw
- Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Douglas Galasko
- Department of Neurology, University of California, San Diego, California, USA
| | - Joseph F Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Department of Neurology and Parkinson’s Disease Research Education and Clinical Care Center (PADRECC), VA Portland Healthcare System, Portland, OR, USA
| | - Min Shi
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Jing Zhang
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- National Health and Disease Human Brain Tissue Resource Center, Zhejiang University, Zhejiang, Hangzhou, China
| |
Collapse
|
16
|
Zhou YN, Jiang L, Zhang Y, Zhou CN, Yang H, He Q, Wang YY, Xiao Q, Huang DJ, Luo YM, Tang Y, Chao FL. Anti-LINGO-1 antibody protects neurons and synapses in the medial prefrontal cortex of APP/PS1 transgenic mice. Neurosci Res 2023:S0168-0102(23)00039-1. [PMID: 36804877 DOI: 10.1016/j.neures.2023.02.005] [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: 11/28/2022] [Revised: 02/11/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
The medial prefrontal cortex (mPFC), one of the most vulnerable brain regions in Alzheimer's disease (AD), plays a critical role in cognition. Leucine-rich repeat and immunoglobulin-like domain-containing nogo receptor-interacting protein-1 (LINGO-1) negatively affects nerve growth in the central nervous system; however, its role in the pathological damage to the mPFC remains to be studied in AD. In this study, an anti-LINGO-1 antibody was administered to 10-month-old APP/PS1 mice, and behavioral tests, stereological methods, immunohistochemistry and immunofluorescence were used to answer this question. Our results revealed that LINGO-1 was highly expressed in the neurons of the mPFC of AD mice, and the anti-LINGO-1 antibody improved prefrontal cortex-related function and reduced the protein level of LINGO-1, atrophy of the volume, Aβ deposition and massive losses of synapses and neurons in the mPFC of AD mice. Antagonizing LINGO-1 could effectively alleviate the pathological damage in the mPFC of AD mice, which might be an important structural basis for improving prefrontal cortex-related function. Abnormal expression of LINGO-1 in the mPFC may be one of the key targets of AD, and the effect initiated by the anti-LINGO-1 antibody may provide an important basis in the search for drugs for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Yu-Ning Zhou
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Lin Jiang
- Experimental Teaching Management Center, Chongqing Medical University, Chongqing 400016, PR China
| | - Yi Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Chun-Ni Zhou
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Hao Yang
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Qi He
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Yi-Ying Wang
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Qian Xiao
- Department of Radioactive Medicine, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Du-Juan Huang
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Yan-Min Luo
- Department of Physiology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Yong Tang
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China.
| | - Feng-Lei Chao
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
17
|
Lin L, Petralia RS, Holtzclaw L, Wang YX, Abebe D, Hoffman DA. Alzheimer's disease/dementia-associated brain pathology in aging DPP6-KO mice. Neurobiol Dis 2022; 174:105887. [PMID: 36209950 PMCID: PMC9617781 DOI: 10.1016/j.nbd.2022.105887] [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: 06/10/2022] [Revised: 08/25/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022] Open
Abstract
We have previously reported that the single transmembrane protein Dipeptidyl Peptidase Like 6 (DPP6) impacts neuronal and synaptic development. DPP6-KO mice are impaired in hippocampal-dependent learning and memory and exhibit smaller brain size. Recently, we have described novel structures in hippocampal area CA1 in aging mice, apparently derived from degenerating presynaptic terminals, that are significantly more prevalent in DPP6-KO mice compared to WT mice of the same age and that these structures were observed earlier in development in DPP6-KO mice. These novel structures appear as clusters of large puncta that colocalize NeuN, synaptophysin, and chromogranin A, and also partially label for MAP2, amyloid β, APP, α-synuclein, and phosphorylated tau, with synapsin-1 and VGluT1 labeling on their periphery. In this current study, using immunofluorescence and electron microscopy, we confirm that both APP and amyloid β are prevalent in these structures; and we show with immunofluorescence the presence of similar structures in humans with Alzheimer's disease. Here we also found evidence that aging DPP6-KO mutants show additional changes related to Alzheimer's disease. We used in vivo MRI to show reduced size of the DPP6-KO brain and hippocampus. Aging DPP6-KO hippocampi contained fewer total neurons and greater neuron death and had diagnostic biomarkers of Alzheimer's disease present including accumulation of amyloid β and APP and increase in expression of hyper-phosphorylated tau. The amyloid β and phosphorylated tau pathologies were associated with neuroinflammation characterized by increases in microglia and astrocytes. And levels of proinflammatory or anti-inflammatory cytokines increased in aging DPP6-KO mice. We finally show that aging DPP6-KO mice display circadian dysfunction, a common symptom of Alzheimer's disease. Together these results indicate that aging DPP6-KO mice show symptoms of enhanced neurodegeneration reminiscent of dementia associated with a novel structure resulting from synapse loss and neuronal death. This study continues our laboratory's work in discerning the function of DPP6 and here provides compelling evidence of a direct role of DPP6 in Alzheimer's disease.
Collapse
Affiliation(s)
- Lin Lin
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ronald S Petralia
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lynne Holtzclaw
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ya-Xian Wang
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Abebe
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dax A Hoffman
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
18
|
Zhou H, Gao JY, Chen Y. The paradigm and future value of the metaverse for the intervention of cognitive decline. Front Public Health 2022; 10:1016680. [PMID: 36339131 PMCID: PMC9631202 DOI: 10.3389/fpubh.2022.1016680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/29/2022] [Indexed: 01/28/2023] Open
Abstract
Cognitive decline is a gradual neurodegenerative process that is affected by genetic and environmental factors. The doctor-patient relationship in the healthcare for cognitive decline is in a "shallow" medical world. With the development of data science, virtual reality, artificial intelligence, and digital twin, the introduction of the concept of the metaverse in medicine has brought alternative and complementary strategies in the intervention of cognitive decline. This article technically analyzes the application scenarios and paradigms of the metaverse in medicine in the field of mental health, such as hospital management, diagnosis, prediction, prevention, rehabilitation, progression delay, assisting life, companionship, and supervision. The metaverse in medicine has made primary progress in education, immersive consultation, dental disease, and Parkinson's disease, bringing revolutionary prospects for non-pharmacological complementary treatment of cognitive decline and other mental problems. In particular, with the demand for non-face-to-face communication generated by the global COVID-19 epidemic, the needs for uncontactable healthcare service for the elderly have increased. The paradigm of self-monitoring, self-healing, and healthcare experienced by the elderly through the metaverse in medicine, especially from meta-platform, meta-community, and meta-hospital, will be generated, which will reconstruct the service modes for the elderly people. The future map of the metaverse in medicine is huge, which depends on the co-construction of community partners.
Collapse
Affiliation(s)
- Hao Zhou
- Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Jian-Yi Gao
- Institute of Medical Genetics, Nanjing Medical University Affiliated Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Ying Chen
- Institute of Medical Genetics, Nanjing Medical University Affiliated Wuxi Maternity and Child Health Care Hospital, Wuxi, China,Jiangnan University Affiliated Wuxi Maternity and Child Health Care Hospital, Wuxi, China,*Correspondence: Ying Chen
| |
Collapse
|
19
|
Guo Q, Wu G, Huang F, Wei Z, Wang JZ, Zhang B, Liu R, Yang Y, Wang X, Li HL. Novel small molecular compound 2JY-OBZ4 alleviates AD pathology in cell models via regulating multiple targets. Aging (Albany NY) 2022; 14:8077-8094. [PMID: 36227154 PMCID: PMC9596221 DOI: 10.18632/aging.204336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/23/2022] [Indexed: 11/25/2022]
Abstract
Alzheimer’s disease (AD) is the most common form of neurodegenerative dementia, characterized by cognitive deficits and memory dysfunction, which is clinically incurable so far. Novel small molecular compound 2JY-OBZ4 is one of structural analogue of Huperzine A (Hup-A), an anti-AD drug in China. In our previous work, 2JY-OBZ4 exhibited potent effects on tau hyperphosphorylation, Aβ production and acetylcholinesterase (AChE) activity. However, 2JY-OBZ4's anti-AD effects and the underlying molecular mechanisms remain unclear. We here reported that 2JY-OBZ4 resisted tau hyperphosphorylation at Thr181 and Ser396 sites in HEK293-hTau cells transfected with GSK-3β, decreased tau phosphorylation via upregulating the activity of PP2A in HEK293-hTau cells and reduced Aβ production through regulating protein levels of APP cleavage enzymes in N2a-hAPP cells. Meanwhile, we found that 2JY-OBZ4 had no adverse effects on cell viability of mice primary neuron even at high concentration, and ameliorated synaptic loss induced by human oligomeric Aβ42. 2JY-OBZ4 had moderate AChE inhibitory activity with the half maximal inhibitory concentration (IC50) to be 39.48 μg/ml in vitro, which is more than two times higher than Hup-A. Together, 2JY-OBZ4 showed promising therapeutic effects in AD cell models through regulating multiple targets. The research provides a new candidate for the therapeutic development of AD.
Collapse
Affiliation(s)
- Qian Guo
- School of Basic Medicine, Key Laboratory of Education Ministry, Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong JS 226001, China
| | - Gang Wu
- School of Basic Medicine, Key Laboratory of Education Ministry, Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong JS 226001, China
| | - Fang Huang
- School of Basic Medicine, Key Laboratory of Education Ministry, Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhen Wei
- School of Basic Medicine, Key Laboratory of Education Ministry, Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jian-Zhi Wang
- School of Basic Medicine, Key Laboratory of Education Ministry, Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong JS 226001, China.,Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Bin Zhang
- School of Basic Medicine, Key Laboratory of Education Ministry, Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rong Liu
- School of Basic Medicine, Key Laboratory of Education Ministry, Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Yang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaochuan Wang
- School of Basic Medicine, Key Laboratory of Education Ministry, Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong JS 226001, China.,Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen 518000, China
| | - Hong-Lian Li
- School of Basic Medicine, Key Laboratory of Education Ministry, Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
20
|
Guo Q, Cai Q, Huang F, Wei Z, Wang JZ, Zhang B, Liu R, Yang Y, Wang X, Li HL. The Therapeutic Effects of Seven Lycophyte Compounds on Cell Models of Alzheimer’s Disease. J Alzheimers Dis 2022; 90:795-809. [DOI: 10.3233/jad-220704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: As an acetylcholinesterase inhibitor (AChEI), Huperzine-A (Hup-A) is marketed for treatment of mild to moderate Alzheimer’s disease (AD) for decades in China. However, Hup-A causes some side effects. To search for new analogs or derivatives of Hup-A, we produced five Lycophyte alkaloids and two analogues by chemical synthesis: Lyconadins A-E, H-R-NOB, and 2JY-OBZ4. Objective: To systematically evaluated the therapeutic effects of the seven compounds on AD cell models. Methods: We assessed the effects of the seven compounds on cell viability via CCK-8 kit and used HEK293-hTau cells and N2a-hAPP cells as AD cell models to evaluate their potential therapeutic effects. We examined their effects on cholinesterase activity by employing the mice primary neuron. Results: All compounds did not affect cell viability; in addition, Lyconadin A and 2JY-OBZ4 particularly increased cell viability. Lyconadin D and Lyconadin E restored tau phosphorylation at Thr231, and H-R-NOB and 2JY-OBZ4 restored tau phosphorylation at Thr231 and Ser396 in GSK-3β-transfected HEK293-hTau cells. 2JY-OBZ4 decreased the level of PP2Ac-pY307 and increased the level of PP2Ac-mL309, supporting that 2JY-OBZ4 may activate PP2A. Lyconadin B, Lyconadin D, Lyconadin E, H-R-NOB, and 2JY-OBZ4 increased sAβPPα level in N2a-hAPP cells. 2JY-OBZ4 decreased the levels of BACE1 and sAβPPβ, thereby reduced Aβ production. Seven compounds exhibited weaker AChE activity inhibition efficiency than Hup-A. Among them, 2JY-OBZ4 showed the strongest AChE inhibition activity with an inhibition rate of 17% at 10μM. Conclusion: Among the seven lycophyte compounds, 2JY-OBZ4 showed the most expected effects on promoting cell viability, downregulating tau hyperphosphorylation, and Aβ production and inhibiting AChE in AD.
Collapse
Affiliation(s)
- Qian Guo
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, China
| | - Qinfeng Cai
- School of Mental Health and Psychological Science, Anhui Medical University, Hefei, Anhui, China
| | - Fang Huang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
| | - Zhen Wei
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
| | - Jian-Zhi Wang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, China
| | - Bin Zhang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
| | - Rong Liu
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
| | - Yang Yang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaochuan Wang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, China
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China
| | - Hong-Lian Li
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Scienceand Technology, Wuhan, China
| |
Collapse
|
21
|
Oh Y, Lee W, Kim SH, Lee S, Kim BC, Lee KH, Kim SH, Song WK. SPIN90 Deficiency Ameliorates Amyloid β Accumulation by Regulating APP Trafficking in AD Model Mice. Int J Mol Sci 2022; 23:ijms231810563. [PMID: 36142484 PMCID: PMC9504172 DOI: 10.3390/ijms231810563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD), a common form of dementia, is caused in part by the aggregation and accumulation in the brain of amyloid β (Aβ), a product of the proteolytic cleavage of amyloid precursor protein (APP) in endosomes. Trafficking of APP, such as surface-intracellular recycling, is an early critical step required for Aβ generation. Less is known, however, about the molecular mechanism regulating APP trafficking. This study investigated the mechanism by which SPIN90, along with Rab11, modulates APP trafficking, Aβ motility and accumulation, and synaptic functionality. Brain Aβ deposition was lower in the progeny of 5xFAD-SPIN90KO mice than in 5xFAD-SPIN90WT mice. Analysis of APP distribution and trafficking showed that the surface fraction of APP was locally distinct in axons and dendrites, with these distributions differing significantly in 5xFAD-SPIN90WT and 5xFAD-SPIN90KO mice, and that neural activity-driven APP trafficking to the surface and intracellular recycling were more actively mobilized in 5xFAD-SPIN90KO neurons. In addition, SPIN90 was found to be cotrafficked with APP via axons, with ablation of SPIN90 reducing the intracellular accumulation of APP in axons. Finally, synaptic transmission was restored over time in 5xFAD-SPIN90KO but not in 5xFAD-SPIN90WT neurons, suggesting SPIN90 is implicated in Aβ production through the regulation of APP trafficking.
Collapse
Affiliation(s)
- Youngsoo Oh
- Cell Logistics Research Center, School of Life Science, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
| | - Wongyoung Lee
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - So Hee Kim
- Cell Logistics Research Center, School of Life Science, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
| | - Sooji Lee
- Department of Medicine, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Byeong C. Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju 61469, Korea
| | - Kun Ho Lee
- Gwangju Alzheimer’s Disease and Related Dementia Cohort Research Center, Chosun University, Gwangju 61452, Korea
| | - Sung Hyun Kim
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Department of Physiology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (S.H.K.); (W.K.S.)
| | - Woo Keun Song
- Cell Logistics Research Center, School of Life Science, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
- Correspondence: (S.H.K.); (W.K.S.)
| |
Collapse
|