Dietmar Rudolf Thal, Koen Poesen, Rik Vandenberghe, Steffi De Meyer
{"title":"Alzheimer's disease neuropathology and its estimation with fluid and imaging biomarkers","authors":"Dietmar Rudolf Thal, Koen Poesen, Rik Vandenberghe, Steffi De Meyer","doi":"10.1186/s13024-025-00819-y","DOIUrl":"https://doi.org/10.1186/s13024-025-00819-y","url":null,"abstract":"Alzheimer’s disease (AD) is neuropathologically characterized by the extracellular deposition of the amyloid-β peptide (Aβ) and the intraneuronal accumulation of abnormal phosphorylated tau (τ)-protein (p-τ). Most frequently, these hallmark lesions are accompanied by other co-pathologies in the brain that may contribute to cognitive impairment, such as vascular lesions, intraneuronal accumulation of phosphorylated transactive-response DNA-binding protein 43 (TDP-43), and/or α-synuclein (αSyn) aggregates. To estimate the extent of these AD and co-pathologies in patients, several biomarkers have been developed. Specific tracers target and visualize Aβ plaques, p-τ and αSyn pathology or inflammation by positron emission tomography. In addition to these imaging biomarkers, cerebrospinal fluid, and blood-based biomarker assays reflecting AD-specific or non-specific processes are either already in clinical use or in development. In this review, we will introduce the pathological lesions of the AD brain, the related biomarkers, and discuss to what extent the respective biomarkers estimate the pathology determined at post-mortem histopathological analysis. It became evident that initial stages of Aβ plaque and p-τ pathology are not detected with the currently available biomarkers. Interestingly, p-τ pathology precedes Aβ deposition, especially in the beginning of the disease when biomarkers are unable to detect it. Later, Aβ takes the lead and accelerates p-τ pathology, fitting well with the known evolution of biomarker measures over time. Some co-pathologies still lack clinically established biomarkers today, such as TDP-43 pathology or cortical microinfarcts. In summary, specific biomarkers for AD-related pathologies allow accurate clinical diagnosis of AD based on pathobiological parameters. Although current biomarkers are excellent measures for the respective pathologies, they fail to detect initial stages of the disease for which post-mortem analysis of the brain is still required. Accordingly, neuropathological studies remain essential to understand disease development especially in early stages. Moreover, there is an urgent need for biomarkers reflecting co-pathologies, such as limbic predominant, age-related TDP-43 encephalopathy-related pathology, which is known to modify the disease by interacting with p-τ. Novel biomarker approaches such as extracellular vesicle-based assays and cryptic RNA/peptides may help to better detect these co-pathologies in the future.\u0000","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"26 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143618413","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Dmytro Morderer, Melissa C. Wren, Feilin Liu, Naomi Kouri, Anastasiia Maistrenko, Bilal Khalil, Nora Pobitzer, Michelle R. Salemi, Brett S. Phinney, Guojun Bu, Na Zhao, Dennis W. Dickson, Melissa E. Murray, Wilfried Rossoll
{"title":"Probe-dependent Proximity Profiling (ProPPr) Uncovers Similarities and Differences in Phospho-Tau-Associated Proteomes Between Tauopathies","authors":"Dmytro Morderer, Melissa C. Wren, Feilin Liu, Naomi Kouri, Anastasiia Maistrenko, Bilal Khalil, Nora Pobitzer, Michelle R. Salemi, Brett S. Phinney, Guojun Bu, Na Zhao, Dennis W. Dickson, Melissa E. Murray, Wilfried Rossoll","doi":"10.1186/s13024-025-00817-0","DOIUrl":"https://doi.org/10.1186/s13024-025-00817-0","url":null,"abstract":"Tauopathies represent a diverse group of neurodegenerative disorders characterized by the abnormal aggregation of the microtubule-associated protein tau. Despite extensive research, the mechanisms underlying the diversity of neuronal and glial tau pathology in different tauopathies are poorly understood. While there is a growing understanding of tauopathy-specific differences in tau isoforms and fibrillar structures, the specific composition of heterogenous tau lesions remains unknown. Here we study the protein composition of tau aggregates in four major tauopathies: Alzheimer's disease (AD), corticobasal degeneration (CBD), Pick's disease (PiD), and progressive supranuclear palsy (PSP). We developed an approach for in situ proximity labeling and isolation of aggregate-associated proteins using glass slides with formalin-fixed paraffin-embedded (FFPE) human postmortem brain tissue, termed Probe-dependent Proximity Profiling (ProPPr). We used ProPPr for the analysis of proteomes associated with AT8-positive cellular lesions from frontal cortices. Isolated proximity proteomes were analyzed by data-independent acquisition mass spectrometry. Co-immunofluorescence staining and quantitative data analysis for selected proteins in human brain tissue was performed to further investigate associations with diverse tau pathologies. Proteomics data analysis identified numerous common and tauopathy-specific proteins associated with phospho-tau aggregates. Extensive validations of candidates through quantitative immunofluorescence imaging of distinct aggregates across disease cases demonstrate successful implementation of ProPPr for unbiased discovery of aggregate-associated proteins in in human brain tissue. Our results reveal the association of retromer complex component vacuolar protein sorting-associated protein 35 (VPS35) and lysosome-associated membrane glycoprotein 2 (LAMP2) with specific types of phospho-tau lesions in tauopathies. Furthermore, we discovered a disease-specific association of certain proteins with distinct pathological lesions, including glycogen synthase kinase alpha (GSK3α), ferritin light chain (FTL), and the neuropeptide precursor VGF. Notably, the identification of FTL-positive microglia in CBD astrocytic plaques indicate their potential role in the pathogenesis of these lesions. Our findings demonstrate the suitability of the ProPPr approach in FFPE brain tissue for unbiased discovery of local proteomes that provide valuable insights into the underlying proteomic landscape of tauopathies, shedding light on the molecular mechanisms underlying tau pathology. This first comprehensive characterization of tau-associated proteomes in a range of distinct tauopathies enhances our understanding of disease heterogeneity and mechanisms, informing strategies for the development of diagnostic biomarkers and targeted therapies.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"113 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-03-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143608076","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Udit Sheth, Linn Öijerstedt, Michael G. Heckman, Launia J. White, Hilary W. Heuer, Argentina Lario Lago, Leah K. Forsberg, Kelley M. Faber, Tatiana M. Foroud, Rosa Rademakers, Eliana Marisa Ramos, Brian S. Appleby, Andrea C. Bozoki, R. Ryan Darby, Bradford C. Dickerson, Kimiko Domoto-Reilly, Douglas R. Galasko, Nupur Ghoshal, Neill R. Graff-Radford, Ian M. Grant, Chadwick M. Hales, Ging-Yuek Robin Hsiung, Edward D. Huey, David Irwin, Justin Y. Kwan, Irene Litvan, Ian R. Mackenzie, Joseph C. Masdeu, Mario F. Mendez, Chiadi U. Onyike, Belen Pascual, Peter S. Pressman, Erik D. Roberson, Allison Snyder, M. Carmela Tartaglia, William W. Seeley, Dennis W. Dickson, Howard J. Rosen, Bradley F. Boeve, Adam L. Boxer, Leonard Petrucelli, Tania F. Gendron
{"title":"Comprehensive cross-sectional and longitudinal comparisons of plasma glial fibrillary acidic protein and neurofilament light across FTD spectrum disorders","authors":"Udit Sheth, Linn Öijerstedt, Michael G. Heckman, Launia J. White, Hilary W. Heuer, Argentina Lario Lago, Leah K. Forsberg, Kelley M. Faber, Tatiana M. Foroud, Rosa Rademakers, Eliana Marisa Ramos, Brian S. Appleby, Andrea C. Bozoki, R. Ryan Darby, Bradford C. Dickerson, Kimiko Domoto-Reilly, Douglas R. Galasko, Nupur Ghoshal, Neill R. Graff-Radford, Ian M. Grant, Chadwick M. Hales, Ging-Yuek Robin Hsiung, Edward D. Huey, David Irwin, Justin Y. Kwan, Irene Litvan, Ian R. Mackenzie, Joseph C. Masdeu, Mario F. Mendez, Chiadi U. Onyike, Belen Pascual, Peter S. Pressman, Erik D. Roberson, Allison Snyder, M. Carmela Tartaglia, William W. Seeley, Dennis W. Dickson, Howard J. Rosen, Bradley F. Boeve, Adam L. Boxer, Leonard Petrucelli, Tania F. Gendron","doi":"10.1186/s13024-025-00821-4","DOIUrl":"https://doi.org/10.1186/s13024-025-00821-4","url":null,"abstract":"Therapeutic development for frontotemporal dementia (FTD) is hindered by the lack of biomarkers that inform susceptibility/risk, prognosis, and the underlying causative pathology. Blood glial fibrillary acidic protein (GFAP) has garnered attention as a FTD biomarker. However, investigations of GFAP in FTD have been hampered by symptomatic and histopathologic heterogeneity and small cohort sizes contributing to inconsistent findings. Therefore, we evaluated plasma GFAP as a FTD biomarker and compared its performance to that of neurofilament light (NfL) protein, a leading FTD biomarker. We availed ARTFL LEFFTDS Longitudinal Frontotemporal Lobar Degeneration (ALLFTD) study resources to conduct a comprehensive cross-sectional and longitudinal examination of the susceptibility/risk, prognostic, and predictive performance of GFAP and NfL in the largest series of well-characterized presymptomatic FTD mutation carriers and participants with sporadic or familial FTD syndromes. Utilizing single molecule array technology, we measured GFAP and NfL in plasma from 161 controls, 127 presymptomatic mutation carriers, 702 participants with a FTD syndrome, and 67 participants with mild behavioral and/or cognitive changes. We used multivariable linear regression and Cox proportional hazard models adjusted for co-variates to examine the biomarker utility of baseline GFAP and NfL concentrations or their rates of change. Compared to controls, GFAP and NfL were elevated in each FTD syndrome but GFAP, unlike NfL, poorly discriminated controls from participants with mild symptoms. Similarly, both baseline GFAP and NfL were higher in presymptomatic mutation carriers who later phenoconverted, but NfL better distinguished non-converters from phenoconverters. We additionally observed that GFAP and NfL were associated with disease severity indicators and survival, but NfL far outperformed GFAP. Nevertheless, we validated findings that the GFAP/NfL ratio may discriminate frontotemporal lobar degeneration with tau versus TDP-43 pathology. Our head-to-head comparison of plasma GFAP and NfL as biomarkers for FTD indicate that NfL consistently outmatched GFAP as a prognostic and predictive biomarker for participants with a FTD syndrome, and as a susceptibility/risk biomarker for people at genetic risk of FTD. Our findings underscore the need to include leading biomarkers in investigations evaluating new biomarkers if the field is to fully ascertain their performance and clinical value.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"26 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-03-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143608080","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lutgarde Serneels, Annerieke Sierksma, Emanuela Pasciuto, Ivana Geric, Arya Nair, Anna Martinez-Muriana, An Snellinx, Bart De Strooper
{"title":"A versatile mouse model to advance human microglia transplantation research in neurodegenerative diseases","authors":"Lutgarde Serneels, Annerieke Sierksma, Emanuela Pasciuto, Ivana Geric, Arya Nair, Anna Martinez-Muriana, An Snellinx, Bart De Strooper","doi":"10.1186/s13024-025-00823-2","DOIUrl":"https://doi.org/10.1186/s13024-025-00823-2","url":null,"abstract":"Recent studies highlight the critical role of microglia in neurodegenerative disorders, and emphasize the need for humanized models to accurately study microglial responses. Human-mouse microglia xenotransplantation models are a valuable platform for functional studies and for testing therapeutic approaches, yet currently those models are only available for academic research. This hampers their implementation for the development and testing of medication that targets human microglia. We developed the hCSF1Bdes mouse line, which is suitable as a new transplantation model and available to be crossed to any disease model of interest. The hCSF1Bdes model created by CRISPR gene editing is RAG2 deficient and expresses human CSF1. Additionally, we crossed this model with two humanized App KI mice, the AppHu and the AppSAA. Flow cytometry, immunohistochemistry and bulk sequencing was used to study the response of microglia in the context of Alzheimer’s disease. Our results demonstrate the successful transplantation of iPSC-derived human microglia into the brains of hCSF1Bdes mice without triggering a NK-driven immune response. Furthermore, we confirmed the multipronged response of microglia in the context of Alzheimer’s disease. The hCSF1Bdes and the crosses with the Alzheimer’s disease knock-in model AppSAA and the humanized App knock-in control mice, AppHu are deposited with EMMA and fully accessible to the research community. The hCSF1Bdes mouse is available for both non-profit and for-profit organisations, facilitating the use of the xenotransplantation paradigm for human microglia to study complex human disease.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"37 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-03-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143589764","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Srebf2 mediates successful optic nerve axon regeneration via the mevalonate synthesis pathway","authors":"Mengming Hu, Matthew B. Veldman","doi":"10.1186/s13024-025-00807-2","DOIUrl":"https://doi.org/10.1186/s13024-025-00807-2","url":null,"abstract":"Axon regeneration within the mammalian central nervous system is extremely limited. In optic neuropathy conditions like glaucoma, the inability of retinal ganglion cell (RGC) axons to regenerate is a major impediment to functional recovery. In contrast, adult teleost fish such as zebrafish can fully regenerate RGC axons enabling visual recovery from optic nerve (ON) injury making it an ideal model to probe the mechanisms of successful axon regeneration. Laser Capture Microdissection followed by RNA-sequencing (LCM-seq) was used to identify genes and pathways differentially expressed in RGCs during ON regeneration. We validate these findings by in situ hybridization and qRT-PCR. Using loss- and gain-of-function experiments we demonstrate the necessity of srebf2 for efficient ON regeneration and recovery of visual function. Finally, we use LCM-seq coupled with experimental manipulations to identify downstream srebf2 target genes and test the role of hmgcra/b and mevalonate in this process. Statistical analysis was performed using Student’s t-test, two-way ANOVA, or repeated measures with appropriate post-hoc tests where applicable. LCM-seq comparison of uninjured versus 3-day post ON injury RGCs identified significant upregulation of the cholesterol synthesis pathway during axon regeneration. The master regulator of this pathway, the transcription factor srebf2, is upregulated throughout the regeneration phase. Chemical inhibition or morpholino-based gene knockdown of srebf2 decreased axon regeneration into the ON and optic tectum and delayed recovery of visual behavior over the course of normal optic nerve regeneration without causing a significant loss of RGCs. Constitutively active srebf2 can fully rescue axon regeneration and visual behavior losses caused by inhibition of endogenous srebf2 but does not accelerate regeneration compared to the control group. LCM-seq confirms the expected regulation of predicted srebf2 target genes after loss- or gain-of-function in vivo. Downstream of srebf2, hmgcra/b knockdown or simvastatin treatment delayed axon regeneration and this effect was rescued by supplemental mevalonate. Mevalonate treatment alone was sufficient to accelerate ON regeneration. These results demonstrate that srebf2 and the downstream mevalonate synthesis pathway plays an important role in regulating efficient axon regeneration in the zebrafish visual system. Involvement of this pathway should be closely examined in failed mammalian ON regeneration.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"23 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-03-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143546416","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mª Salomé Sirerol-Piquer, Ana Perez-Villalba, Pere Duart-Abadia, Germán Belenguer, Ulises Gómez-Pinedo, Laura Blasco-Chamarro, Pau Carrillo-Barberà, Azucena Pérez-Cañamás, Victoria Navarro-Garrido, Benjamin Dehay, Miquel Vila, Javier Vitorica, Francisco Pérez-Sánchez, Isabel Fariñas
{"title":"Age-dependent progression from clearance to vulnerability in the early response of periventricular microglia to α-synuclein toxic species","authors":"Mª Salomé Sirerol-Piquer, Ana Perez-Villalba, Pere Duart-Abadia, Germán Belenguer, Ulises Gómez-Pinedo, Laura Blasco-Chamarro, Pau Carrillo-Barberà, Azucena Pérez-Cañamás, Victoria Navarro-Garrido, Benjamin Dehay, Miquel Vila, Javier Vitorica, Francisco Pérez-Sánchez, Isabel Fariñas","doi":"10.1186/s13024-025-00816-1","DOIUrl":"https://doi.org/10.1186/s13024-025-00816-1","url":null,"abstract":"Cytoplasmic alpha-synuclein (αSyn) aggregates are a typical feature of Parkinson’s disease (PD). Extracellular insoluble αSyn can induce pathology in healthy neurons suggesting that PD neurodegeneration may spread through cell-to-cell transfer of αSyn proteopathic seeds. Early pro-homeostatic reaction of microglia to toxic forms of αSyn remains elusive, which is especially relevant considering the recently uncovered microglial molecular diversity. Here, we show that periventricular microglia of the subependymal neurogenic niche monitor the cerebrospinal fluid and can rapidly phagocytize and degrade different aggregated forms of αSyn delivered into the lateral ventricle. However, this clearing ability worsens with age, leading to an increase in microglia with aggregates in aged treated mice, an accumulation also observed in human PD samples. We also show that exposure of aged microglia to aggregated αSyn isolated from human PD samples results in the phosphorylation of the endogenous protein and the generation of αSyn seeds that can transmit the pathology to healthy neurons. Our data indicate that while microglial phagocytosis rapidly clears toxic αSyn, aged microglia can contribute to synucleinopathy spreading.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"12 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-03-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143546417","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rong-Xin Zhu, Rui-Xue Han, Yue-Han Chen, Lei Huang, Ting Liu, Jingwei Jiang, Cong Wang, Lei Cao, Yang Liu, Ming Lu
{"title":"Inactivation of NLRP3 inflammasome by dephosphorylation at Serine 658 alleviates glial inflammation in the mouse model of Parkinson’s disease","authors":"Rong-Xin Zhu, Rui-Xue Han, Yue-Han Chen, Lei Huang, Ting Liu, Jingwei Jiang, Cong Wang, Lei Cao, Yang Liu, Ming Lu","doi":"10.1186/s13024-025-00818-z","DOIUrl":"https://doi.org/10.1186/s13024-025-00818-z","url":null,"abstract":"Parkinson’s disease (PD) is a leading neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons, contributing to considerable disability worldwide. Current treatments offer only symptomatic relief, highlighting the need for novel therapeutic strategies targeting disease progression. Neuroinflammation plays a pivotal role in PD pathogenesis, with the NLRP3 inflammasome emerging as a key contributor. The virtual screening of a natural product library comprising 5,088 compounds was applied to identify five potential NLRP3 inhibitors through molecular docking scores. Then surface plasmon resonance assays were used to detect their binding affinities to the NLRP3 protein. Functional studies in macrophages and glial cells were used to demonstrate the effect of Psoralen on NLRP3 phosphorylation and inflammasome activation. Psoralen treatment improved PD-like symptoms and reduced dopaminergic neuronal death by targeting glial NLRP3 inflammasome activation in the MPTP/p mouse model. By performing 4D label-free quantitative phosphorylation proteomics and site mutation assays, we identified that Psoralen prevents NLRP3 phosphorylation at Serine 658 by binding to its NACHT and LRR domains. These findings position Psoralen as a promising NLRP3 inflammasome inhibitor, offering a potential therapeutic avenue for PD and other NLRP3 inflammasome-related diseases. Additionally, this research highlights the innovative approach of targeting specific phosphorylation sites on the NLRP3 protein to reduce neuroinflammation.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"84 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-03-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143546634","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"The neuroimmune nexus: unraveling the role of the mtDNA-cGAS-STING signal pathway in Alzheimer’s disease","authors":"Shuiyue Quan, Xiaofeng Fu, Huimin Cai, Ziye Ren, Yinghao Xu, Longfei Jia","doi":"10.1186/s13024-025-00815-2","DOIUrl":"https://doi.org/10.1186/s13024-025-00815-2","url":null,"abstract":"The relationship between Alzheimer's disease (AD) and neuroimmunity has gradually begun to be unveiled. Emerging evidence indicates that cyclic GMP-AMP synthase (cGAS) acts as a cytosolic DNA sensor, recognizing cytosolic damage-associated molecular patterns (DAMPs), and inducing the innate immune response by activating stimulator of interferon genes (STING). Dysregulation of this pathway culminates in AD-related neuroinflammation and neurodegeneration. A substantial body of evidence indicates that mitochondria are involved in the critical pathogenic mechanisms of AD, whose damage leads to the release of mitochondrial DNA (mtDNA) into the extramitochondrial space. This leaked mtDNA serves as a DAMP, activating various pattern recognition receptors and immune defense networks in the brain, including the cGAS-STING pathway, ultimately leading to an imbalance in immune homeostasis. Therefore, modulation of the mtDNA-cGAS-STING pathway to restore neuroimmune homeostasis may offer promising prospects for improving AD treatment outcomes. In this review, we focus on the mechanisms of mtDNA release during stress and the activation of the cGAS-STING pathway. Additionally, we delve into the research progress on this pathway in AD, and further discuss the primary directions and potential hurdles in developing targeted therapeutic drugs, to gain a deeper understanding of the pathogenesis of AD and provide new approaches for its therapy.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"39 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143538404","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ji Hun Kim, Sumin Yang, Hyojung Kim, Dang-Khoa Vo, Han-Joo Maeng, Areum Jo, Joo-Heon Shin, Joo-Ho Shin, Hyeon-Man Baek, Gum Hwa Lee, Sung-Hyun Kim, Key-Hwan Lim, Valina L. Dawson, Ted M. Dawson, Jae-Yeol Joo, Yunjong Lee
{"title":"Preclinical studies and transcriptome analysis in a model of Parkinson’s disease with dopaminergic ZNF746 expression","authors":"Ji Hun Kim, Sumin Yang, Hyojung Kim, Dang-Khoa Vo, Han-Joo Maeng, Areum Jo, Joo-Heon Shin, Joo-Ho Shin, Hyeon-Man Baek, Gum Hwa Lee, Sung-Hyun Kim, Key-Hwan Lim, Valina L. Dawson, Ted M. Dawson, Jae-Yeol Joo, Yunjong Lee","doi":"10.1186/s13024-025-00814-3","DOIUrl":"https://doi.org/10.1186/s13024-025-00814-3","url":null,"abstract":"The parkin-interacting substrate (PARIS, also known as ZNF746) is a transcriptional repressor, whose accumulation and phosphorylation play central pathological roles in Parkinson’s disease (PD). PARIS-induced transcriptional repression of PGC-1α or MDM4 contributes to mitochondrial dysfunction and p53-dependent neuron loss in PD. Despite the important role of PARIS in PD pathogenesis, unbiased transcriptomic profiles influenced by PARIS accumulation in dopaminergic neurons remain unexplored. We engineered Tet-Off conditional transgenic mice expressing PARIS in dopaminergic neurons, driven by DAT-PF-tTA driver mice. The conditional PARIS transgenic mice were characterized by PD-associated pathologies, including progressive dopamine cell loss, neuroinflammation, PGC-1α repression, and mitochondrial proteome alteration. Motor impairment was assessed using pole and rotarod tests. L-DOPA and c-Abl inhibitors were administered to PARIS transgenic mice to evaluate their therapeutic efficacy. The transcriptomic profiles and gene ontology clusters were analyzed by bulk and single-nucleus RNA-seq for the ventral midbrains from PARIS transgenic and age-matched controls. Conditional dopaminergic PARIS expression in mice led to the robust and selective dopaminergic neuron degeneration, neuroinflammation, and striatal dopamine deficits, resulting in L-DOPA-responsive motor impairments. Consistent with the results of previous reports, PARIS suppressed dopaminergic PGC-1α expression, disturbed mitochondrial marker protein expression, and reduced COXIV-labeled mitochondria in dopamine neurons. Pharmacological inhibition of c-Abl activity in PARIS transgenic mice largely prevents PD-associated pathological features. Unbiased transcriptomic analysis revealed PARIS-regulated differentially expressed genes (DEGs), both collectively and in a cell-type-specific manner, along with enriched biological pathways linked to PD pathogenesis. Single-cell resolution transcriptomic analysis confirmed repression of PGC-1α and several mitochondria-related target genes in dopaminergic cells. Additionally, we identified distinct glial cell subpopulations and DEGs associated with PD pathogenesis. Conditional PARIS transgenic mice recapitulate robust and dopaminergic neuron-selective pathological features of PD, allowing the preclinical evaluation of antisymptomatic and disease-modifying therapeutic strategies within a couple of months. Based on this new PD mouse model, we provide unbiased bulk and single-nucleus transcriptomic profiles that are regulated by PARIS and potentially contribute to PD pathogenesis. A PD mouse model with flexible pathology induction capacity and a whole transcriptome could serve as a useful resource for translational PD research.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"13 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-02-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143518654","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Michael L. Alosco, Jhony Mejía Pérez, Julia E. Culhane, Ranjani Shankar, Christopher J. Nowinski, Samantha Bureau, Nidhi Mundada, Karen Smith, Alinda Amuiri, Breton Asken, Jenna R. Groh, Annalise Miner, Erika Pettway, Sydney Mosaheb, Yorghos Tripodis, Charles Windon, Gustavo Mercier, Robert A. Stern, Lea T. Grinberg, David N. Soleimani-Meigooni, Bradley T. Christian, Tobey J. Betthauser, Thor D. Stein, Ann C. McKee, Chester A. Mathis, Eric E. Abrahamson, Milos D. Ikonomovic, Sterling C. Johnson, Jesse Mez, Renaud La Joie, Daniel Schonhaut, Gil D. Rabinovici
{"title":"18F-MK-6240 tau PET in patients at-risk for chronic traumatic encephalopathy","authors":"Michael L. Alosco, Jhony Mejía Pérez, Julia E. Culhane, Ranjani Shankar, Christopher J. Nowinski, Samantha Bureau, Nidhi Mundada, Karen Smith, Alinda Amuiri, Breton Asken, Jenna R. Groh, Annalise Miner, Erika Pettway, Sydney Mosaheb, Yorghos Tripodis, Charles Windon, Gustavo Mercier, Robert A. Stern, Lea T. Grinberg, David N. Soleimani-Meigooni, Bradley T. Christian, Tobey J. Betthauser, Thor D. Stein, Ann C. McKee, Chester A. Mathis, Eric E. Abrahamson, Milos D. Ikonomovic, Sterling C. Johnson, Jesse Mez, Renaud La Joie, Daniel Schonhaut, Gil D. Rabinovici","doi":"10.1186/s13024-025-00808-1","DOIUrl":"https://doi.org/10.1186/s13024-025-00808-1","url":null,"abstract":"Molecular biomarkers of chronic traumatic encephalopathy (CTE) are lacking. We evaluated 18F-MK-6240 tau PET as a biomarker for CTE. Two studies were done: (1) 3H-MK-6240 autoradiography and an in-vitro brain homogenate binding studies on postmortem CTE tissue, (2) an in-vivo 18F-MK-6240 tau PET study in former American football players. Autoradiography and in-vitro binding studies were done using 3H-MK-6240 on frozen temporal and frontal cortex tissue from six autopsy cases with stage III CTE compared to Alzheimer’s disease. Thirty male former National Football League (NFL) players with cognitive concerns (mean age = 58.9, SD = 7.8) completed tau (18F-MK-6240) and Aβ (18F-Florbetapir) PET. Controls included 39 Aβ-PET negative, cognitively normal males (mean age = 65.7, SD = 6.3). 18F-MK-6240 SUVr images were created using 70–90 min post-injection data with inferior cerebellar gray matter as the reference. We compared SUVr between players and controls using voxelwise and region-of-interest approaches. Correlations between 18F-MK-6240 SUVr and cognitive scores were tested. All six CTE stage III cases had Braak NFT stage III but no neuritic plaques. Two had Thal Phase 1 for Aβ; one showed a laminar pattern of 3H-MK-6240 autoradiography binding in the superior temporal cortex and less so in the dorsolateral frontal cortex, corresponding to tau-immunoreactive lesions detected using the AT8 antibody (pSer202/pThr205 tau) in adjacent tissue sections. The other CTE cases had low frequencies of cortical tau-immunoreactive deposits and no well-defined autoradiography binding. In-vitro 3H-MK-6240 binding studies to CTE brain homogenates in the case with autoradiography signal indicated high binding affinity (KD = 2.0 ± 0.9 nM, Bmax = 97 ± 24 nM, n = 3). All NFL players had negative Aβ-PET. There was variable, low-to-intermediate intensity 18F-MK-6240 uptake across participants: 16 had no cortical signal, 7 had medial temporal lobe (MTL) uptake, 2 had frontal uptake, and 4 had MTL and frontal uptake. NFL players had higher SUVr in the entorhinal cortex (d = 0.86, p = 0.001), and the parahippocampal gyrus (d = 0.39, p = 0.08). Voxelwise regressions showed increased uptake in NFL players in two bilateral anterior MTL clusters (p < 0.05 FWE). Higher parahippocampal and frontal–temporal SUVrs correlated with worse memory (r = -0.38, r = -0.40) and semantic fluency (r = -0.38, r = -0.48), respectively. We present evidence of 3H-MK-6240 in-vitro binding to post-mortem CTE tissue homogenates and in vivo 18F-MK-6240 PET binding in the MTL among a subset of participants. Additional studies in larger samples and PET-to-autopsy correlations are required to further elucidate the potential of 18F-MK-6240 to detect tau pathology in CTE.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"22 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143485822","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}