Molecular Neurodegeneration最新文献

筛选
英文 中文
Glycerophospholipids in ALS: insights into disease mechanisms and clinical implication 肌萎缩侧索硬化症中的甘油磷脂:疾病机制和临床意义的见解
IF 15.1 1区 医学
Molecular Neurodegeneration Pub Date : 2025-07-26 DOI: 10.1186/s13024-025-00876-3
Thibaut Burg, Ludo Van Den Bosch
{"title":"Glycerophospholipids in ALS: insights into disease mechanisms and clinical implication","authors":"Thibaut Burg, Ludo Van Den Bosch","doi":"10.1186/s13024-025-00876-3","DOIUrl":"https://doi.org/10.1186/s13024-025-00876-3","url":null,"abstract":"Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease affecting the adult motor system, with no effective treatments available. Despite extensive research efforts, the exact pathological cascade leading to progressive motor neuron degeneration remains elusive. Recent evidence highlights significant modifications in lipid metabolism during ALS progression, even before the onset of motor symptoms. Glycerophospholipids, the primary components of cellular membranes, are frequently altered in ALS patients and models. These lipids not only play a structural role in membranes, but also contribute to cellular metabolism, signaling pathways, and cell type-specific processes such as neuronal transmission and muscle contraction. In this review, we discuss glycerophospholipid physiological functions in the motor system and review recent studies demonstrating their alterations and the possible underlying pathological mechanisms in ALS. Furthermore, we discuss challenges emerging from studying lipid alterations in neurodegeneration and evaluate the therapeutic potential of glycerophospholipids.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"14 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-07-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144710637","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}
引用次数: 0
The role of endolysosomal progranulin and TMEM106B in neurodegenerative diseases 内溶酶体颗粒前蛋白和TMEM106B在神经退行性疾病中的作用
IF 15.1 1区 医学
Molecular Neurodegeneration Pub Date : 2025-07-26 DOI: 10.1186/s13024-025-00873-6
Hideyuki Takahashi, Stephen M. Strittmatter
{"title":"The role of endolysosomal progranulin and TMEM106B in neurodegenerative diseases","authors":"Hideyuki Takahashi, Stephen M. Strittmatter","doi":"10.1186/s13024-025-00873-6","DOIUrl":"https://doi.org/10.1186/s13024-025-00873-6","url":null,"abstract":"Although different neurodegenerative diseases are defined by distinct pathological proteins, they share many common features including protein aggregation. Despite this commonality, most current therapeutic approaches in the field, such as anti-aggregate antibodies, are focused on individual diseases or single neuropathologies with only limited success. The endolysosomal proteins progranulin and TMEM106B were both initially associated with frontotemporal lobar degeneration but have subsequently also been linked to other neurodegenerative diseases. Thus, these proteins are predicted to participate in common pathogenic pathways shared across various neurodegenerative diseases. Importantly, recent discoveries of TMEM106B amyloid fibrils in varied neurodegenerative diseases and glycosphingolipid regulation by progranulin and TMEM106B further support their central roles in cross-disease neurodegenerative mechanisms. This review summarizes recent advances in progranulin and TMEM106B function within the endolysosomal system and neurodegenerative diseases. It describes preclinical models and therapeutic approaches for progranulin- and TMEM106B-associated diseases. We also discuss future direction leading to novel alternative therapies targeting shared mechanisms in neurodegenerative diseases.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"21 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-07-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144710705","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}
引用次数: 0
Endothelium-specific endoglin triggers astrocyte reactivity via extracellular vesicles in a mouse model of Alzheimer’s disease 在阿尔茨海默病小鼠模型中,内皮特异性内啡肽通过细胞外囊泡触发星形胶质细胞反应性
IF 15.1 1区 医学
Molecular Neurodegeneration Pub Date : 2025-07-23 DOI: 10.1186/s13024-025-00875-4
Pingao Zhang, Chenghuan Song, Jiyun Shi, Zijie Wei, Jing Wang, Wanying Huang, Rui Zhang, Jintao Wang, Xiaoli Yang, Gang Wang, Xiaoling Gao, Yongfang Zhang, Hongzhuan Chen, Hao Wang
{"title":"Endothelium-specific endoglin triggers astrocyte reactivity via extracellular vesicles in a mouse model of Alzheimer’s disease","authors":"Pingao Zhang, Chenghuan Song, Jiyun Shi, Zijie Wei, Jing Wang, Wanying Huang, Rui Zhang, Jintao Wang, Xiaoli Yang, Gang Wang, Xiaoling Gao, Yongfang Zhang, Hongzhuan Chen, Hao Wang","doi":"10.1186/s13024-025-00875-4","DOIUrl":"https://doi.org/10.1186/s13024-025-00875-4","url":null,"abstract":"Alzheimer’s disease (AD) is a multifaceted neurodegenerative disorder with a complex etiology that extends beyond the well-documented amyloid-β and tau pathologies. Growing evidence implicates cerebrovascular dysfunction, particularly brain microvascular endothelial cells (BMECs) dysfunction, as an early contributor to AD pathogenesis. However, how BMECs influence on neighboring astrocytes needs to be further explored. We employed a multi-omics approach integrating bulk RNA sequencing of human BMECs with proteomic analysis of cerebrospinal fluid (CSF) from AD patients and cerebrovascular endothelial extracellular vesicles (CEEVs). The role of identified candidate proteins was investigated in vitro and in vivo utilizing CEEVs transplantation and BMEC-astrocyte co-cultures. Endothelial cell-specific knockdown or treatment with a monoclonal antibody was used to assess the functional consequences on cognitive impairment and AD pathology via two-photon imaging and behavioral experiments on APP/PS1 mice. The elevated endothelium-specific protein Endoglin (ENG) was identified in the brain and serum of AD individuals and APP/PS1 mice, and the supernatant of injured BMECs. ENG was released and delivered to adjacent astrocytes via CEEVs, and subsequently upregulated TGFBRI/Smad3 pathway in astrocytes, leading to astrocyte reactivity and the release of pro-inflammatory cytokines. Endothelial cell-specific ENG knockdown or treating with ENG monoclonal antibody Carotuximab significantly suppressed reactive astrocytes, reduced neuroinflammation, and improved cognitive performance of APP/PS1 mice. This study reveals a novel mechanism by which BMECs-derived ENG, delivered via CEEVs, drives astrocyte reactivity. These findings redefine the role of cerebrovascular dysfunction in AD pathogenesis and identify ENG as both a potential biomarker and a promising therapeutic target for AD.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"112 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144684670","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}
引用次数: 0
Proteomic landscape of Alzheimer’s disease: emerging technologies, advances and insights (2021 – 2025) 阿尔茨海默病的蛋白质组学前景:新兴技术,进展和见解(2021 - 2025)
IF 15.1 1区 医学
Molecular Neurodegeneration Pub Date : 2025-07-14 DOI: 10.1186/s13024-025-00874-5
Jay M. Yarbro, Him K. Shrestha, Zhen Wang, Xue Zhang, Masihuz Zaman, Mengqi Chu, Xusheng Wang, Gang Yu, Junmin Peng
{"title":"Proteomic landscape of Alzheimer’s disease: emerging technologies, advances and insights (2021 – 2025)","authors":"Jay M. Yarbro, Him K. Shrestha, Zhen Wang, Xue Zhang, Masihuz Zaman, Mengqi Chu, Xusheng Wang, Gang Yu, Junmin Peng","doi":"10.1186/s13024-025-00874-5","DOIUrl":"https://doi.org/10.1186/s13024-025-00874-5","url":null,"abstract":"The advancements of proteomics technologies are shaping Alzheimer's disease (AD) research, revealing new molecular insights and improving biomarker discovery. Here, we summarize major AD proteomics studies since our 2021 review, focusing on disease mechanisms and biomarker identification. Enhanced sensitivity and throughput in proteome profiling have been driven by mass spectrometry (MS)-based approaches and affinity-based platforms (e.g., Olink and SomaScan). Emerging techniques, including single-cell, spatial, and single-molecule proteomics, provide unprecedented resolution in studying cellular heterogeneity and pathological microenvironments (e.g., amyloidome). Multi-cohort analyses of AD brain tissues have revealed consensus protein alterations (n = 866), identifying novel disease-associated proteins validated in functional studies (e.g., MDK/PTN, NTN1, SMOC1, GPNMB, NPTX2, NRN1, VGF, and U1 snRNP). Proteomic studies of AD biofluids have identified distinct disease subtypes, offering candidate proteins for early detection. Comparisons between human tissues and AD mouse models highlight shared pathways in amyloid pathology while underscoring limitations in recapitulating human disease. Combining proteomics with genomics enables protein quantitative trait locus (pQTL) analysis in AD, linking genetic risk factors to protein expression changes. Discrepancies between proteome and transcriptome suggest altered protein turnover in AD. Overall, AD proteomics continues to provide mechanistic insights into disease progression and potential biomarkers for precision medicine.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"13 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-07-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144622228","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}
引用次数: 0
Biomarkers and therapeutic strategies targeting microglia in neurodegenerative diseases: current status and future directions 神经退行性疾病中针对小胶质细胞的生物标志物和治疗策略:现状和未来方向
IF 15.1 1区 医学
Molecular Neurodegeneration Pub Date : 2025-07-10 DOI: 10.1186/s13024-025-00867-4
Min-Young Noh, Hyuk Sung  Kwon, Min-Soo Kwon, Minyeop Nahm, Hee Kyung Jin, Jae-sung Bae, Seung Hyun Kim
{"title":"Biomarkers and therapeutic strategies targeting microglia in neurodegenerative diseases: current status and future directions","authors":"Min-Young Noh, Hyuk Sung  Kwon, Min-Soo Kwon, Minyeop Nahm, Hee Kyung Jin, Jae-sung Bae, Seung Hyun Kim","doi":"10.1186/s13024-025-00867-4","DOIUrl":"https://doi.org/10.1186/s13024-025-00867-4","url":null,"abstract":"Recent advances in our understanding of non-cell-autonomous mechanisms in neurodegenerative diseases (NDDs) have highlighted microglial dysfunction as a core driver of disease progression. Conditions such as Alzheimer’s disease (AD), amyotrophic lateral sclerosis (ALS), Parkinson’s disease (PD), and frontotemporal dementia (FTD) share features of impaired microglial phagocytosis, chronic neuroinflammation, and metabolic dysregulation. These insights have prompted new therapeutic strategies targeting microglial function and emphasized the need for reliable biomarkers to monitor disease progression and treatment response. Well-established therapeutic targets, such as triggering receptor expressed on myeloid cells 2 (TREM2), progranulin (PGRN), and sortilin (SORT1), along with emerging candidates including LILRB4, P2Y6R, TAM receptors, and neuroinflammation-related markers, are discussed alongside novel blood, cerebrospinal fluid (CSF), and imaging biomarkers. Despite notable progress, many of these biomarkers remain restricted to preclinical studies and face translational challenges due to species-specific differences, lack of standardization, and clinical heterogeneity. Emerging technologies—including single-cell omics, spatial transcriptomics, and artificial intelligence (AI)-driven integration of multimodal data—offer new opportunities to align biomarker profiles with evolving disease states and improve patient stratification. Building on the model of companion diagnostics (CDx) in oncology, integrating multimodal biomarker strategies holds promise for guiding personalized interventions, improving clinical outcomes, and deepening our mechanistic understanding of microglial contributions across the neurodegenerative spectrum.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"26 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144594115","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}
引用次数: 0
Glial phagocytosis for synapse and toxic proteins in neurodegenerative diseases. 神经退行性疾病中突触和毒性蛋白的胶质吞噬作用。
IF 15.1 1区 医学
Molecular Neurodegeneration Pub Date : 2025-07-09 DOI: 10.1186/s13024-025-00870-9
Yeseong Choi,Won-Suk Chung
{"title":"Glial phagocytosis for synapse and toxic proteins in neurodegenerative diseases.","authors":"Yeseong Choi,Won-Suk Chung","doi":"10.1186/s13024-025-00870-9","DOIUrl":"https://doi.org/10.1186/s13024-025-00870-9","url":null,"abstract":"Glia, as resident immune and supportive cells of the central nervous system, play a critical role in maintaining brain homeostasis. One of their key homeostatic functions is phagocytic capacity in pruning synapses and removing cellular debris/protein aggregates, a process vital for synaptic plasticity and brain maintenance. However, these phagocytic functions are often dysregulated with aging and in neurodegenerative diseases (NDs), such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and frontotemporal dementia. This review aims to examine the phagocytic roles of glia under both physiological and pathological conditions, with a special focus on their interactions with misfolded protein aggregates, including amyloid beta, tau, alpha synuclein, prion, huntingtin, and TAR DNA-binding protein 43. We also explore the fate of ingested molecules after being phagocytosed by glia-whether they are degraded, accumulate intracellularly, or are transferred between cells-and their implications for disease progression. Finally, we review current therapeutic strategies and the potential approaches for modulating glial phagocytosis to mitigate several NDs. We believe that understanding the exact mechanisms of glial phagocytosis and clearance will serve as key elements in developing future treatments for NDs.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"26 1","pages":"81"},"PeriodicalIF":15.1,"publicationDate":"2025-07-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144586441","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}
引用次数: 0
Methylome analysis of FTLD patients with TDP-43 pathology identifies epigenetic signatures specific to pathological subtypes 具有TDP-43病理的FTLD患者的甲基组分析确定了病理亚型特异性的表观遗传特征
IF 15.1 1区 医学
Molecular Neurodegeneration Pub Date : 2025-07-06 DOI: 10.1186/s13024-025-00869-2
Cristina T. Vicente, Tejasvi Niranjan, Elise Coopman, Júlia Faura, Sara Alidadiani, Claudia Schrauwen, Billie J. Matchett, Bavo Heeman, Marleen Van den Broeck, Wouter De Coster, Thuy Nguyen, Julie S. Lau, Saurabh Baheti, Tim de Pooter, Peter De Rijk, Mojca Strazisar, Matt Baker, Mariely DeJesus-Hernandez, NiCole A. Finch, Cyril Pottier, Marka van Blitterswijk, Yan Asmann, Melissa E. Murray, Leonard Petrucelli, Andrew King, Claire Troakes, Safa Al-Sarraj, Robert A. Rissman, Annie Hiniker, Margaret Flanagan, Bret M. Evers, Charles L. White, Carlos Cruchaga, Rudolph Castellani, Jeroen G.J. van Rooij, Merel O. Mol, Harro Seelaar, John C. van Swieten, Björn Oskarsson, Robert Ross Reichard, Aivi T. Nguyen, Keith A. Josephs, Ronald C. Petersen, Nilüfer Ertekin-Taner, Bradley F. Boeve, Neill R. Graff-Radford, Sarah Weckhuysen, Dennis W. Dickson, Rosa Rademakers
{"title":"Methylome analysis of FTLD patients with TDP-43 pathology identifies epigenetic signatures specific to pathological subtypes","authors":"Cristina T. Vicente, Tejasvi Niranjan, Elise Coopman, Júlia Faura, Sara Alidadiani, Claudia Schrauwen, Billie J. Matchett, Bavo Heeman, Marleen Van den Broeck, Wouter De Coster, Thuy Nguyen, Julie S. Lau, Saurabh Baheti, Tim de Pooter, Peter De Rijk, Mojca Strazisar, Matt Baker, Mariely DeJesus-Hernandez, NiCole A. Finch, Cyril Pottier, Marka van Blitterswijk, Yan Asmann, Melissa E. Murray, Leonard Petrucelli, Andrew King, Claire Troakes, Safa Al-Sarraj, Robert A. Rissman, Annie Hiniker, Margaret Flanagan, Bret M. Evers, Charles L. White, Carlos Cruchaga, Rudolph Castellani, Jeroen G.J. van Rooij, Merel O. Mol, Harro Seelaar, John C. van Swieten, Björn Oskarsson, Robert Ross Reichard, Aivi T. Nguyen, Keith A. Josephs, Ronald C. Petersen, Nilüfer Ertekin-Taner, Bradley F. Boeve, Neill R. Graff-Radford, Sarah Weckhuysen, Dennis W. Dickson, Rosa Rademakers","doi":"10.1186/s13024-025-00869-2","DOIUrl":"https://doi.org/10.1186/s13024-025-00869-2","url":null,"abstract":"In the last decade, the importance of DNA methylation in the functioning of the central nervous system has been highlighted through associations between methylation changes and differential expression of key genes involved in aging and neurodegenerative diseases. In frontotemporal lobar degeneration (FTLD), aberrant methylation has been reported in causal disease genes including GRN and C9orf72; however, the genome-wide contribution of epigenetic changes to the development of FTLD remains largely unexplored. We performed reduced representation bisulfite sequencing of matched pairs of post-mortem tissue from frontal cortex (FCX) and cerebellum (CER) from pathologically confirmed FTLD patients with TDP-43 pathology (FTLD-TDP) further divided into five subtypes and including both sporadic and genetic forms (N = 25 pairs per group), and neuropathologically normal controls (N = 42 pairs). Case-control differential methylation analyses were performed, both at the individual CpG level, and in regions of grouped CpGs (differentially methylated regions; DMRs), either including all genomic locations or only gene promoters. Gene Ontology (GO) analyses were then performed using all differentially methylated genes in each group of sporadic patients. Finally, additional datasets were queried to prioritize candidate genes for follow-up. Using the largest FTLD-TDP DNA methylation dataset generated to date, we identified thousands of differentially methylated CpGs (FCX = 6,520; CER = 7,134) and several hundred DMRs in FTLD-TDP brains (FCX = 134; CER = 219). Of these, less than 10% are shared between pathological subgroups. Combining additional datasets, we identified, validated and replicated hypomethylation of CAMTA1 in TDP-A potentially also impacting additional genes in the locus. GO analysis further implicated DNA methylation in myelination and developmental processes, as well as important disease-relevant mechanisms with subtype specificity such as protein phosphorylation and DNA damage repair in TDP-A, cholesterol biosynthesis in TDP-B, and protein localization in TDP-C. We identify methylation changes in all FTLD-TDP patient groups and show that most changes are unique to a specific pathological FTLD-TDP subtype, suggesting that these subtypes not only have distinct transcriptomic and genetic signatures, but are also epigenetically distinct. Our study constitutes an invaluable resource to the community and highlights the need for further studies to profile additional epigenetic layers within each FTLD-TDP pathological subtype.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"46 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-07-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144565679","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}
引用次数: 0
Skin biopsy: an emerging tool for the diagnosis of protein misfolding diseases of the central nervous system 皮肤活组织检查:一种用于诊断中枢神经系统蛋白质错误折叠疾病的新兴工具
IF 15.1 1区 医学
Molecular Neurodegeneration Pub Date : 2025-07-03 DOI: 10.1186/s13024-025-00871-8
Wei Zhu, Hao-Lun Sun, Yan-Jiang Wang, Xia Lei, Xian-Le Bu
{"title":"Skin biopsy: an emerging tool for the diagnosis of protein misfolding diseases of the central nervous system","authors":"Wei Zhu, Hao-Lun Sun, Yan-Jiang Wang, Xia Lei, Xian-Le Bu","doi":"10.1186/s13024-025-00871-8","DOIUrl":"https://doi.org/10.1186/s13024-025-00871-8","url":null,"abstract":"<p>Abnormal aggregation of misfolded proteins is a common characteristic of many neurodegenerative diseases [1]. Brain protein aggregates formed by phosphorylated tau, amyloid-beta (Aβ), α-synuclein, or prion proteins are toxic and can cause gradual neuronal loss and death in the central nervous system (CNS). These aggregates are linked to many neurodegenerative diseases such as Alzheimer’s disease (AD), and Parkinson’s disease (PD). Rapid advancements in brain PET imaging and body fluid biomarkers have enabled the precise diagnosis of these disorders. However, brain PET scans are expensive, cerebrospinal fluid (CSF) collection is an invasive procedure, and precise diagnostic tools remain limited. Therefore, it is imperative to develop simpler and more accessible diagnostic approaches. Skin tissue shares a common developmental origin with the CNS, as both are derived from the ectoderm during embryogenesis, and skin is rich in nerve fibers. Multiple misfolded proteins, including α-synuclein, tau, and Aβ, are deposited not only in the brain but also in skin tissue [2,3,4]. The biological classification of PD emphasizes the necessity of detecting pathological α-synuclein for diagnosis, with skin biomarkers being particularly noted as an accessible and promising detection method [5, 6]. Skin biopsy samples are more accessible than brain PET and CSF samples. Therefore, skin biopsy holds potential as a biomarker-based method for the diagnosis of protein aggregation diseases.</p><p>Synucleinopathies are defined as a group of neurodegenerative disorders characterized by the abnormal accumulation of α-synuclein in the brain. These include PD, dementia with Lewy bodies (DLB), multiple system atrophy (MSA), and pure autonomic failure (PAF). Recently, Gibbons et al. conducted a blinded, multicenter, cross-sectional study to evaluate the rate of positivity for cutaneous α-synuclein deposition among patients with synucleinopathies (PD, DLB, MSA, and PAF) [7]. The researchers enrolled 151 controls and 277 patients with synucleinopathies diagnosed on the basis of clinical consensus criteria and the findings were confirmed by an expert review panel. Among the participants, 343 participants (223 clinically diagnosed with a specific synucleinopathy and 120 who met the criteria for serving as controls without a synucleinopathy) were included in the primary analysis. Skin biopsy samples were taken from the distal leg, the distal thigh, and the posterior cervical region. Among those patients clinically diagnosed with a specific synucleinopathy, 95.5% (213 of 223) had a skin biopsy sample that was positive for phosphorylated α-synuclein. Specifically, phosphorylated α-synuclein positivity in skin biopsy samples was observed for 92.7% of PD patients, 98.2% of MSA patients, 96% of DLB patients, and 100% of PAF patients. In contrast, only 3.3% (4 of 120) of those in the control group had skin biopsy samples that were positive for phosphorylated α-synuclein. In the seconda","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"18 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144547419","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}
引用次数: 0
Current strategies in the management of dementia with lewy bodies and future directions based on disease pathophysiology 路易体痴呆的当前治疗策略及基于疾病病理生理学的未来方向
IF 15.1 1区 医学
Molecular Neurodegeneration Pub Date : 2025-07-01 DOI: 10.1186/s13024-025-00856-7
Daniel Erskine, John-Paul Taylor
{"title":"Current strategies in the management of dementia with lewy bodies and future directions based on disease pathophysiology","authors":"Daniel Erskine, John-Paul Taylor","doi":"10.1186/s13024-025-00856-7","DOIUrl":"https://doi.org/10.1186/s13024-025-00856-7","url":null,"abstract":"Dementia with Lewy bodies (DLB) is thought to be the second most common form of dementia after Alzheimer’s disease, and is characterised by a combination of cognitive, neuropsychiatric and motor symptoms. The present review seeks to discuss current strategies for the management of DLB, and future directions for novel disease-modifying therapies. Current best practice for the clinical management of DLB is based upon therapies that target specific symptom domains due to the lack of disease-modifying therapies. Cholinesterase inhibitors are the frontline treatment for treating cognitive decline in DLB, whereas the treatment of motor symptoms remains challenging due to poor response to dopaminergic therapies and the potential for exacerbation of neuropsychiatric features. There is emerging evidence suggesting a range of further pharmacological and non-pharmacological therapies may be effective in treating specific symptom domains of DLB, but further evidence is warranted to demonstrate their efficacy. A key challenge in the treatment of DLB is incomplete understanding of disease pathophysiology, which has limited attempts to develop disease-modifying therapies. In the present article, we discuss the multi-faceted nature of DLB neuropathology, from Lewy body pathology to mitochondrial dysfunction, and discuss therapies in development that target particular aspects of DLB neuropathology. In particular, we highlight antibody-based therapies to attenuate protein aggregation, compounds that enhance the generation of cellular energy and autophagy-enhancing agents as particular areas of promise. Furthermore, we discuss how optimal strategies for disease modification will be centred on agents that treat DLB neuropathology more holistically, and will be underpinned by a more complete understanding of the pathogenic events that underlie the full spectrum of pathological changes observed in the DLB brain.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"9 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144520837","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}
引用次数: 0
Alpha-synuclein seeding amplification assays in Lewy body dementia: a brief review 路易体痴呆中α -突触核蛋白种子扩增试验:综述
IF 15.1 1区 医学
Molecular Neurodegeneration Pub Date : 2025-07-01 DOI: 10.1186/s13024-025-00868-3
Maria Bregendahl, Zeynep Bengisu Kaya, Wolfgang Singer, Pamela J. McLean
{"title":"Alpha-synuclein seeding amplification assays in Lewy body dementia: a brief review","authors":"Maria Bregendahl, Zeynep Bengisu Kaya, Wolfgang Singer, Pamela J. McLean","doi":"10.1186/s13024-025-00868-3","DOIUrl":"https://doi.org/10.1186/s13024-025-00868-3","url":null,"abstract":"Lewy body dementia (LBD), which includes dementia with Lewy bodies (DLB) and Parkinson’s disease dementia (PDD), is characterized by cognitive decline, sleep disturbances, motor dysfunction, and other debilitating clinical symptoms. Neuropathologically, LBD is characterized by the progressive accumulation of alpha-synuclein (aSYN) in vulnerable cellular populations in the brain. Diagnosing LBD is challenging due to the overlap of clinical symptoms with Alzheimer’s disease (AD) and other neurodegenerative disorders with current diagnostic tools, including clinical examinations by specialized neurologists and brain imaging, limited by accessibility. Taken together, LBD is often misdiagnosed, especially at early disease stages. Seed amplification assays to detect pathogenic aSYN (aSYN SAAs) are emerging as promising tools to detect aSYN pathology in biological specimens. These assays amplify trace amounts of misfolded aSYN, enabling their potential detection in brain, CSF, saliva, skin, and blood. This review compares the sensitivity and specificity of aSYN SAAs across different biological samples and explores the potential of the assay as a diagnostic in LBD. We also highlight challenges that will need to be addressed going forward if the aSYN SAA is to be widely adopted as a diagnostic test. Despite current limitations, aSYN SAAs hold promise for early and precise diagnosis, paving the way for targeted treatments that could significantly improve patient care and outcomes.","PeriodicalId":18800,"journal":{"name":"Molecular Neurodegeneration","volume":"48 1","pages":""},"PeriodicalIF":15.1,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144520953","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}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信