Β-site amyloid precursor protein (APP) cleaving enzyme (BACE1) is a crucial protease in the production of amyloid-β (Aβ) in Alzheimer’s disease (AD) patients. However, the side effects observed in clinical trials of BACE1 inhibitors, including reduction in brain volume and cognitive worsening, suggest that the exact role of BACE1 in AD pathology is not fully understood. To further investigate this, we examined cerebrospinal fluid (CSF) levels of BACE1 and its cleaved product sAPPβ that reflects BACE1 activity in the China Aging and Neurodegenerative Disorder Initiative cohort. We found significant correlations between CSF BACE1 or sAPPβ levels and CSF Aβ40, Aβ42, and Aβ42/Aβ40 ratio, but not with amyloid deposition detected by 18F-Florbetapir PET. Additionally, CSF BACE1 and sAPPβ levels were positively associated with cortical thickness in multiple brain regions, and higher levels of sAPPβ were linked to increased cortical glucose metabolism in frontal and supramarginal areas. Interestingly, individuals with higher baseline levels of CSF BACE1 exhibited slower rates of brain volume reduction and cognitive worsening over time. This suggests that increased levels and activity of BACE1 may not be the determining factor for amyloid deposition, but instead, may be associated with increased neuronal activity and potentially providing protection against neurodegeneration in AD.
{"title":"Associations of CSF BACE1 with amyloid pathology, neurodegeneration, and cognition in Alzheimer’s disease","authors":"Feng Gao, Mengguo Zhang, Qiong Wang, Ming Ni, Chang Liu, Kexue Deng, Qiang Xie, Shicung Wang, Jiong Shi, Yong Shen, For CANDI Consortium","doi":"10.1007/s00401-024-02750-w","DOIUrl":"10.1007/s00401-024-02750-w","url":null,"abstract":"<div><p>Β-site amyloid precursor protein (APP) cleaving enzyme (BACE1) is a crucial protease in the production of amyloid-β (Aβ) in Alzheimer’s disease (AD) patients. However, the side effects observed in clinical trials of BACE1 inhibitors, including reduction in brain volume and cognitive worsening, suggest that the exact role of BACE1 in AD pathology is not fully understood. To further investigate this, we examined cerebrospinal fluid (CSF) levels of BACE1 and its cleaved product sAPPβ that reflects BACE1 activity in the China Aging and Neurodegenerative Disorder Initiative cohort. We found significant correlations between CSF BACE1 or sAPPβ levels and CSF Aβ40, Aβ42, and Aβ42/Aβ40 ratio, but not with amyloid deposition detected by 18F-Florbetapir PET. Additionally, CSF BACE1 and sAPPβ levels were positively associated with cortical thickness in multiple brain regions, and higher levels of sAPPβ were linked to increased cortical glucose metabolism in frontal and supramarginal areas. Interestingly, individuals with higher baseline levels of CSF BACE1 exhibited slower rates of brain volume reduction and cognitive worsening over time. This suggests that increased levels and activity of BACE1 may not be the determining factor for amyloid deposition, but instead, may be associated with increased neuronal activity and potentially providing protection against neurodegeneration in AD.</p></div>","PeriodicalId":7012,"journal":{"name":"Acta Neuropathologica","volume":"147 1","pages":""},"PeriodicalIF":9.3,"publicationDate":"2024-06-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141295384","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}
Pub Date : 2024-06-09DOI: 10.1007/s00401-024-02747-5
Sriram Balusu, Bart De Strooper
Although apoptosis, pyroptosis, and ferroptosis have been implicated in AD, none fully explains the extensive neuronal loss observed in AD brains. Recent evidence shows that necroptosis is abundant in AD, that necroptosis is closely linked to the appearance of Tau pathology, and that necroptosis markers accumulate in granulovacuolar neurodegeneration vesicles (GVD). We review here the neuron-specific activation of the granulovacuolar mediated neuronal-necroptosis pathway, the potential AD-relevant triggers upstream of this pathway, and the interaction of the necrosome with the endo-lysosomal pathway, possibly providing links to Tau pathology. In addition, we underscore the therapeutic potential of inhibiting necroptosis in neurodegenerative diseases such as AD, as this presents a novel avenue for drug development targeting neuronal loss to preserve cognitive abilities. Such an approach seems particularly relevant when combined with amyloid-lowering drugs.
虽然凋亡、热凋亡和铁凋亡都与 AD 有关,但它们都不能完全解释在 AD 大脑中观察到的大量神经元丢失。最近的证据表明,坏死在AD中大量存在,坏死与Tau病理学的出现密切相关,而且坏死标记物在颗粒细胞神经变性囊泡中积聚。我们在此回顾了粒细胞介导的神经元-坏死通路的神经元特异性激活、该通路上游的潜在AD相关诱因、坏死体与内溶酶体通路的相互作用,这些可能与Tau病理学有关。此外,我们还强调了抑制坏死蛋白沉积对神经退行性疾病(如阿氏痴呆症)的治疗潜力,因为这为针对神经元缺失以保护认知能力的药物开发提供了一条新途径。当这种方法与降低淀粉样蛋白的药物相结合时,似乎尤为重要。
{"title":"The necroptosis cell death pathway drives neurodegeneration in Alzheimer’s disease","authors":"Sriram Balusu, Bart De Strooper","doi":"10.1007/s00401-024-02747-5","DOIUrl":"10.1007/s00401-024-02747-5","url":null,"abstract":"<div><p>Although apoptosis, pyroptosis, and ferroptosis have been implicated in AD, none fully explains the extensive neuronal loss observed in AD brains. Recent evidence shows that necroptosis is abundant in AD, that necroptosis is closely linked to the appearance of Tau pathology, and that necroptosis markers accumulate in granulovacuolar neurodegeneration vesicles (GVD). We review here the neuron-specific activation of the granulovacuolar mediated neuronal-necroptosis pathway, the potential AD-relevant triggers upstream of this pathway, and the interaction of the necrosome with the endo-lysosomal pathway, possibly providing links to Tau pathology. In addition, we underscore the therapeutic potential of inhibiting necroptosis in neurodegenerative diseases such as AD, as this presents a novel avenue for drug development targeting neuronal loss to preserve cognitive abilities. Such an approach seems particularly relevant when combined with amyloid-lowering drugs.</p></div>","PeriodicalId":7012,"journal":{"name":"Acta Neuropathologica","volume":"147 1","pages":""},"PeriodicalIF":9.3,"publicationDate":"2024-06-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11162975/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141292997","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-07DOI: 10.1007/s00401-024-02746-6
Konstantin Okonechnikov, Daniel Schrimpf, Jan Koster, Philipp Sievers, Till Milde, Felix Sahm, David T. W. Jones, Andreas von Deimling, Stefan M. Pfister, Marcel Kool, Andrey Korshunov
The non-WNT/non-SHH (Grp3/Grp4) medulloblastomas (MBs) include eight second-generation subgroups (SGS; I–VIII) each with distinct molecular and clinical characteristics. Recently, we also identified two prognostically relevant transcriptome subtypes within each SGS MB, which are associated with unique gene expression signatures and signaling pathways. These prognostic subsets may be in connection to the intra-tumoral cell landscape that underlies SGS MB clinical-molecular diversity. Here, we performed a deconvolution analysis of the Grp3/Grp4 MB bulk RNA profiles using the previously identified single-cell RNA-seq reference dataset and focusing on variability in the cellular composition of SGS MB. RNA deconvolution analysis of the Grp3/Grp4 MB disclosed the subgroup-specific neoplastic cell subpopulations. Neuronally differentiated axodendritic GP3-C1 and glutamatergic GP4-C1 subpopulations were distributed within Grp3- and Grp4-associated SGS MB, respectively. Progenitor GP3-B2 subpopulation was prominent in aggressive SGS II MB, whereas photoreceptor/visual perception GP3/4-C2 cell content was typical for SGS III/IV MB. The current study also revealed significant variability in the proportions of cell subpopulations between clinically relevant SGS MB transcriptome subtypes, where unfavorable cohorts were enriched with cell cycle and progenitor-like cell subpopulations and, vice versa, favorable subtypes were composed of neuronally differentiated cell fractions predominantly. A higher than median proportion of proliferating and progenitor cell subpopulations conferred the shortest survival of the Grp3 and Grp 4 MB, and similar survival associations were identified for all SGS MB except SGS IV MB. In summary, the recently identified clinically relevant Grp3/Grp4 MB transcriptome subtypes are composed of different cell populations. Future studies should aim to validate the prognostic and therapeutic role of the identified Grp3/Grp4 MB inter-tumoral cellular heterogeneity. The application of the single-cell techniques on each SGS MB separately could help to clarify the clinical significance of subgroup-specific variability in tumor cell content and its relation with prognostic transcriptome signatures identified before.
{"title":"Clinically unfavorable transcriptome subtypes of non-WNT/non-SHH medulloblastomas are associated with a predominance in proliferating and progenitor-like cell subpopulations","authors":"Konstantin Okonechnikov, Daniel Schrimpf, Jan Koster, Philipp Sievers, Till Milde, Felix Sahm, David T. W. Jones, Andreas von Deimling, Stefan M. Pfister, Marcel Kool, Andrey Korshunov","doi":"10.1007/s00401-024-02746-6","DOIUrl":"10.1007/s00401-024-02746-6","url":null,"abstract":"<div><p>The non-WNT/non-SHH (Grp3/Grp4) medulloblastomas (MBs) include eight second-generation subgroups (SGS; I–VIII) each with distinct molecular and clinical characteristics. Recently, we also identified two prognostically relevant transcriptome subtypes within each SGS MB, which are associated with unique gene expression signatures and signaling pathways. These prognostic subsets may be in connection to the intra-tumoral cell landscape that underlies SGS MB clinical-molecular diversity. Here, we performed a deconvolution analysis of the Grp3/Grp4 MB bulk RNA profiles using the previously identified single-cell RNA-seq reference dataset and focusing on variability in the cellular composition of SGS MB. RNA deconvolution analysis of the Grp3/Grp4 MB disclosed the subgroup-specific neoplastic cell subpopulations. Neuronally differentiated axodendritic GP3-C1 and glutamatergic GP4-C1 subpopulations were distributed within Grp3- and Grp4-associated SGS MB, respectively. Progenitor GP3-B2 subpopulation was prominent in aggressive SGS II MB, whereas photoreceptor/visual perception GP3/4-C2 cell content was typical for SGS III/IV MB. The current study also revealed significant variability in the proportions of cell subpopulations between clinically relevant SGS MB transcriptome subtypes, where unfavorable cohorts were enriched with cell cycle and progenitor-like cell subpopulations and, vice versa<i>,</i> favorable subtypes were composed of neuronally differentiated cell fractions predominantly. A higher than median proportion of proliferating and progenitor cell subpopulations conferred the shortest survival of the Grp3 and Grp 4 MB, and similar survival associations were identified for all SGS MB except SGS IV MB. In summary, the recently identified clinically relevant Grp3/Grp4 MB transcriptome subtypes are composed of different cell populations. Future studies should aim to validate the prognostic and therapeutic role of the identified Grp3/Grp4 MB inter-tumoral cellular heterogeneity. The application of the single-cell techniques on each SGS MB separately could help to clarify the clinical significance of subgroup-specific variability in tumor cell content and its relation with prognostic transcriptome signatures identified before.</p></div>","PeriodicalId":7012,"journal":{"name":"Acta Neuropathologica","volume":"147 1","pages":""},"PeriodicalIF":9.3,"publicationDate":"2024-06-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141282612","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}
A recent large genome-wide association study has identified EGFR (encoding the epidermal growth factor EGFR) as a new genetic risk factor for late-onset AD. SHIP2, encoded by INPPL1, is taking part in the signalling and interactome of several growth factor receptors, such as the EGFR. While INPPL1 has been identified as one of the most significant genes whose RNA expression correlates with cognitive decline, the potential alteration of SHIP2 expression and localization during the progression of AD remains largely unknown. Here we report that gene expression of both EGFR and INPPL1 was upregulated in AD brains. SHIP2 immunoreactivity was predominantly detected in plaque-associated astrocytes and dystrophic neurites and its increase was correlated with amyloid load in the brain of human AD and of 5xFAD transgenic mouse model of AD. While mRNA of INPPL1 was increased in AD, SHIP2 protein undergoes a significant solubility change being depleted from the soluble fraction of AD brain homogenates and co-enriched with EGFR in the insoluble fraction. Using FRET-based flow cytometry biosensor assay for tau-tau interaction, overexpression of SHIP2 significantly increased the FRET signal while siRNA-mediated downexpression of SHIP2 significantly decreased FRET signal. Genetic association analyses suggest that some variants in INPPL1 locus are associated with the level of CSF pTau. Our data support the hypothesis that SHIP2 is an intermediate key player of EGFR and AD pathology linking amyloid and tau pathologies in human AD.
最近的一项大型全基因组关联研究发现,表皮生长因子受体(EGFR)(编码表皮生长因子 EGFR)是晚发性注意力缺失症的一个新的遗传风险因素。由 INPPL1 编码的 SHIP2 参与了多种生长因子受体(如表皮生长因子受体)的信号传递和相互作用。虽然 INPPL1 已被确定为 RNA 表达与认知能力下降相关的最重要基因之一,但 SHIP2 的表达和定位在 AD 进展过程中的潜在变化在很大程度上仍不为人所知。在这里,我们报告了表皮生长因子受体和 INPPL1 在 AD 大脑中的基因表达上调。在人类 AD 和 5xFAD 转基因小鼠 AD 模型中,SHIP2 的免疫反应主要在斑块相关的星形胶质细胞和萎缩性神经元中检测到,其增加与淀粉样蛋白负荷相关。虽然INPPL1的mRNA在AD中增加,但SHIP2蛋白的溶解度发生了显著变化,从AD脑匀浆的可溶部分中消失,并与表皮生长因子受体共同富集在不溶部分中。利用基于FRET的流式细胞仪生物传感器检测tau-tau相互作用,SHIP2的过表达显著增加了FRET信号,而siRNA介导的SHIP2的下表达则显著降低了FRET信号。遗传关联分析表明,INPPL1位点的一些变异与CSF pTau水平有关。我们的数据支持这一假设:SHIP2 是表皮生长因子受体和 AD 病理学的中间关键角色,它将人类 AD 中的淀粉样蛋白和 tau 病理学联系在一起。
{"title":"Alteration of gene expression and protein solubility of the PI 5-phosphatase SHIP2 are correlated with Alzheimer’s disease pathology progression","authors":"Kunie Ando, Fahri Küçükali, Emilie Doeraene, Siranjeevi Nagaraj, Eugenia Maria Antonelli, May Thazin Htut, Zehra Yilmaz, Andreea-Claudia Kosa, Lidia Lopez-Guitierrez, Carolina Quintanilla-Sánchez, Emmanuel Aydin, Ana Raquel Ramos, Salwa Mansour, Sabrina Turbant, Stéphane Schurmans, Kristel Sleegers, Christophe Erneux, Jean-Pierre Brion, Karelle Leroy","doi":"10.1007/s00401-024-02745-7","DOIUrl":"10.1007/s00401-024-02745-7","url":null,"abstract":"<div><p>A recent large genome-wide association study has identified <i>EGFR</i> (encoding the epidermal growth factor EGFR) as a new genetic risk factor for late-onset AD. SHIP2, encoded by <i>INPPL1</i>, is taking part in the signalling and interactome of several growth factor receptors, such as the EGFR. While <i>INPPL1</i> has been identified as one of the most significant genes whose RNA expression correlates with cognitive decline, the potential alteration of SHIP2 expression and localization during the progression of AD remains largely unknown. Here we report that gene expression of both <i>EGFR</i> and <i>INPPL1</i> was upregulated in AD brains. SHIP2 immunoreactivity was predominantly detected in plaque-associated astrocytes and dystrophic neurites and its increase was correlated with amyloid load in the brain of human AD and of 5xFAD transgenic mouse model of AD. While mRNA of <i>INPPL1</i> was increased in AD, SHIP2 protein undergoes a significant solubility change being depleted from the soluble fraction of AD brain homogenates and co-enriched with EGFR in the insoluble fraction. Using FRET-based flow cytometry biosensor assay for tau-tau interaction, overexpression of SHIP2 significantly increased the FRET signal while siRNA-mediated downexpression of SHIP2 significantly decreased FRET signal. Genetic association analyses suggest that some variants in <i>INPPL1</i> locus are associated with the level of CSF pTau. Our data support the hypothesis that SHIP2 is an intermediate key player of EGFR and AD pathology linking amyloid and tau pathologies in human AD.</p></div>","PeriodicalId":7012,"journal":{"name":"Acta Neuropathologica","volume":"147 1","pages":""},"PeriodicalIF":9.3,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11150309/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141236295","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-05-27DOI: 10.1007/s00401-024-02743-9
Ann-Charlotte E. Granholm, Elisabet Englund, Anah Gilmore, Elizabeth Head, William H. Yong, Sylvia E. Perez, Samuel J. Guzman, Eric D. Hamlett, Elliott J. Mufson
The SARS-CoV-2 virus that led to COVID-19 is associated with significant and long-lasting neurologic symptoms in many patients, with an increased mortality risk for people with Alzheimer’s disease (AD) and/or Down syndrome (DS). However, few studies have evaluated the neuropathological and inflammatory sequelae in postmortem brain tissue obtained from AD and people with DS with severe SARS-CoV-2 infections. We examined tau, beta-amyloid (Aβ), inflammatory markers and SARS-CoV-2 nucleoprotein in DS, AD, and healthy non-demented controls with COVID-19 and compared with non-infected brain tissue from each disease group (total n = 24). A nested ANOVA was used to determine regional effects of the COVID-19 infection on arborization of astrocytes (Sholl analysis) and percent-stained area of Iba-1 and TMEM 119. SARS-CoV-2 antibodies labeled neurons and glial cells in the frontal cortex of all subjects with COVID-19, and in the hippocampus of two of the three DS COVID-19 cases. SARS-CoV-2-related alterations were observed in peri-vascular astrocytes and microglial cells in the gray matter of the frontal cortex, hippocampus, and para-hippocampal gyrus. Bright field microscopy revealed scattered intracellular and diffuse extracellular Aβ deposits in the hippocampus of controls with confirmed SARS-CoV-2 infections. Overall, the present preliminary findings suggest that SARS-CoV-2 infections induce abnormal inflammatory responses in Down syndrome.
{"title":"Neuropathological findings in Down syndrome, Alzheimer’s disease and control patients with and without SARS-COV-2: preliminary findings","authors":"Ann-Charlotte E. Granholm, Elisabet Englund, Anah Gilmore, Elizabeth Head, William H. Yong, Sylvia E. Perez, Samuel J. Guzman, Eric D. Hamlett, Elliott J. Mufson","doi":"10.1007/s00401-024-02743-9","DOIUrl":"10.1007/s00401-024-02743-9","url":null,"abstract":"<div><p>The SARS-CoV-2 virus that led to COVID-19 is associated with significant and long-lasting neurologic symptoms in many patients, with an increased mortality risk for people with Alzheimer’s disease (AD) and/or Down syndrome (DS). However, few studies have evaluated the neuropathological and inflammatory sequelae in <i>postmortem</i> brain tissue obtained from AD and people with DS with severe SARS-CoV-2 infections. We examined tau, beta-amyloid (Aβ), inflammatory markers and SARS-CoV-2 nucleoprotein in DS, AD, and healthy non-demented controls with COVID-19 and compared with non-infected brain tissue from each disease group (total <i>n</i> = 24). A nested ANOVA was used to determine regional effects of the COVID-19 infection on arborization of astrocytes (Sholl analysis) and percent-stained area of Iba-1 and TMEM 119. SARS-CoV-2 antibodies labeled neurons and glial cells in the frontal cortex of all subjects with COVID-19, and in the hippocampus of two of the three DS COVID-19 cases. SARS-CoV-2-related alterations were observed in peri-vascular astrocytes and microglial cells in the gray matter of the frontal cortex, hippocampus, and para-hippocampal gyrus. Bright field microscopy revealed scattered intracellular and diffuse extracellular Aβ deposits in the hippocampus of controls with confirmed SARS-CoV-2 infections. Overall, the present preliminary findings suggest that SARS-CoV-2 infections induce abnormal inflammatory responses in Down syndrome.</p></div>","PeriodicalId":7012,"journal":{"name":"Acta Neuropathologica","volume":"147 1","pages":""},"PeriodicalIF":9.3,"publicationDate":"2024-05-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11130011/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141155090","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
APOEε4 is the major genetic risk factor for sporadic Alzheimer’s disease (AD). Although APOEε4 is known to promote Aβ pathology, recent data also support an effect of APOE polymorphism on phosphorylated Tau (pTau) pathology. To elucidate these potential effects, the pTau interactome was analyzed across APOE genotypes in the frontal cortex of 10 advanced AD cases (n = 5 APOEε3/ε3 and n = 5 APOEε4/ε4), using a combination of anti-pTau pS396/pS404 (PHF1) immunoprecipitation (IP) and mass spectrometry (MS). This proteomic approach was complemented by an analysis of anti-pTau PHF1 and anti-Aβ 4G8 immunohistochemistry, performed in the frontal cortex of 21 advanced AD cases (n = 11 APOEε3/ε3 and n = 10 APOEε4/ε4). Our dataset includes 1130 and 1330 proteins enriched in IPPHF1 samples from APOEε3/ε3 and APOEε4/ε4 groups (fold change ≥ 1.50, IPPHF1vs IPIgG ctrl). We identified 80 and 68 proteins as probable pTau interactors in APOEε3/ε3 and APOEε4/ε4 groups, respectively (SAINT score ≥ 0.80; false discovery rate (FDR) ≤ 5%). A total of 47/80 proteins were identified as more likely to interact with pTau in APOEε3/ε3 vs APOEε4/ε4 cases. Functional enrichment analyses showed that they were significantly associated with the nucleoplasm compartment and involved in RNA processing. In contrast, 35/68 proteins were identified as more likely to interact with pTau in APOEε4/ε4 vs APOEε3/ε3 cases. They were significantly associated with the synaptic compartment and involved in cellular transport. A characterization of Tau pathology in the frontal cortex showed a higher density of plaque-associated neuritic crowns, made of dystrophic axons and synapses, in APOEε4 carriers. Cerebral amyloid angiopathy was more frequent and severe in APOEε4/ε4 cases. Our study supports an influence of APOE genotype on pTau-subcellular location in AD. These results suggest a facilitation of pTau progression to Aβ-affected brain regions in APOEε4 carriers, paving the way to the identification of new therapeutic targets.
{"title":"The influence of APOEε4 on the pTau interactome in sporadic Alzheimer’s disease","authors":"Manon Thierry, Jackeline Ponce, Mitchell Martà-Ariza, Manor Askenazi, Arline Faustin, Dominique Leitner, Geoffrey Pires, Evgeny Kanshin, Eleanor Drummond, Beatrix Ueberheide, Thomas Wisniewski","doi":"10.1007/s00401-024-02744-8","DOIUrl":"10.1007/s00401-024-02744-8","url":null,"abstract":"<div><p><i>APOE</i><sup><i>ε4</i></sup> is the major genetic risk factor for sporadic Alzheimer’s disease (AD). Although <i>APOE</i><sup><i>ε4</i></sup> is known to promote Aβ pathology, recent data also support an effect of <i>APOE</i> polymorphism on phosphorylated Tau (pTau) pathology. To elucidate these potential effects, the pTau interactome was analyzed across <i>APOE</i> genotypes in the frontal cortex of 10 advanced AD cases (<i>n</i> = 5 <i>APOE</i><sup>ε3/ε3</sup> and <i>n</i> = 5 <i>APOE</i><sup><i>ε4/ε4</i></sup>), using a combination of anti-pTau pS396/pS404 (PHF1) immunoprecipitation (IP) and mass spectrometry (MS). This proteomic approach was complemented by an analysis of anti-pTau PHF1 and anti-Aβ 4G8 immunohistochemistry, performed in the frontal cortex of 21 advanced AD cases (<i>n</i> = 11 <i>APOE</i><sup>ε3/ε3</sup> and <i>n</i> = 10 <i>APOE</i><sup><i>ε4/ε4</i></sup>). Our dataset includes 1130 and 1330 proteins enriched in IP<sub>PHF1</sub> samples from <i>APOE</i><sup>ε3/ε3</sup> and <i>APOE</i><sup><i>ε4/ε4</i></sup> groups (fold change ≥ 1.50, IP<sub>PHF1</sub> <i>vs</i> IP<sub>IgG ctrl</sub>). We identified 80 and 68 proteins as probable pTau interactors in <i>APOE</i><sup>ε3/ε3</sup> and <i>APOE</i><sup><i>ε4/ε4</i></sup> groups, respectively (SAINT score ≥ 0.80; false discovery rate (FDR) ≤ 5%). A total of 47/80 proteins were identified as more likely to interact with pTau in <i>APOE</i><sup><i>ε3/ε3</i></sup><i> vs APOE</i><sup><i>ε4/ε4</i></sup> cases. Functional enrichment analyses showed that they were significantly associated with the nucleoplasm compartment and involved in RNA processing. In contrast, 35/68 proteins were identified as more likely to interact with pTau in <i>APOE</i><sup><i>ε4/ε4</i></sup><i> vs APOE</i><sup><i>ε3/ε3</i></sup> cases. They were significantly associated with the synaptic compartment and involved in cellular transport. A characterization of Tau pathology in the frontal cortex showed a higher density of plaque-associated neuritic crowns, made of dystrophic axons and synapses, in <i>APOE</i><sup><i>ε4</i></sup> carriers. Cerebral amyloid angiopathy was more frequent and severe in <i>APOE</i><sup><i>ε4/ε4</i></sup> cases. Our study supports an influence of <i>APOE</i> genotype on pTau-subcellular location in AD. These results suggest a facilitation of pTau progression to Aβ-affected brain regions in <i>APOE</i><sup><i>ε4</i></sup> carriers, paving the way to the identification of new therapeutic targets.</p></div>","PeriodicalId":7012,"journal":{"name":"Acta Neuropathologica","volume":"147 1","pages":""},"PeriodicalIF":9.3,"publicationDate":"2024-05-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11108952/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141074591","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-05-21DOI: 10.1007/s00401-024-02742-w
Alyse de Boer, Aletta M. R. van den Bosch, Nienke J. Mekkes, Nina L. Fransen, Ekaterina Dagkesamanskaia, Eric Hoekstra, Jörg Hamann, Joost Smolders, Inge Huitinga, Inge R. Holtman
Multiple sclerosis (MS) is a heterogeneous neurological disorder with regards to clinical presentation and pathophysiology. Here, we investigated the heterogeneity of MS by performing an exploratory factor analysis on quantitative and qualitative neuropathology data collected for 226 MS donors in the Netherlands Brain Bank autopsy cohort. Three promising dimensions were identified and subsequently validated with clinical, neuropathological, and genetic data. Dimension 1 ranged from a predominance of remyelinated and inactive lesions to extensive pathological changes, higher proportions of active and mixed lesions, and foamy microglia morphology. This pattern was positively correlated with more severe disease, the presence of B and T cells, and neuroaxonal damage. Scoring high on dimension 2 was associated with active lesions, reactive sites, and the presence of nodules. These donors had less severe disease, a specific pattern of cortical lesions, and MS risk variants in the human leukocyte antigen region, the latter indicating a connection between disease onset and this neuropathological dimension. Donors scoring high on dimension 3 showed increased lesional pathology with relatively more mixed and inactive lesions and ramified microglia morphology. This pattern was associated with longer disease duration, subpial cortical lesions, less involvement of the adaptive immune system, and less axonal damage. Taken together, the three dimensions may represent (1) demyelination and immune cell activity associated with pathological and clinical progression, (2) microglia (re)activity and possibly lesion initiation, and (3) loss of lesion activity and scar formation. Our findings highlight that a thorough understanding of the interplay between multiple pathological characteristics is crucial to understand the heterogeneity of MS pathology, as well as its association with genetic predictors and disease outcomes. The scores of donors on the dimensions can serve as an important starting point for further disentanglement of MS heterogeneity and translation into observations and interventions in living cohorts with MS.
多发性硬化症(MS)在临床表现和病理生理学方面是一种异质性神经系统疾病。在此,我们通过对荷兰脑库尸检队列中 226 名多发性硬化症供体的定量和定性神经病理学数据进行探索性因子分析,研究了多发性硬化症的异质性。结果发现了三个有前景的维度,并随后通过临床、神经病理学和遗传学数据进行了验证。维度 1 的范围从再髓鞘化和非活动性病变为主到广泛的病理变化、较高比例的活动性和混合性病变以及泡沫状小胶质细胞形态。这种模式与更严重的疾病、B 细胞和 T 细胞的存在以及神经轴损伤呈正相关。维度 2 的高分与活动性病变、反应性部位和结节的存在有关。这些捐献者的病情较轻,皮质病变模式特殊,人类白细胞抗原区域存在多发性硬化症风险变异,后者表明发病与这一神经病理学维度有关。在维度3上得分较高的供体表现出更多的病变病理,混合性和非活动性病变相对较多,小胶质细胞形态呈横纹状。这种模式与较长的病程、皮质下病变、较少的适应性免疫系统参与和较少的轴突损伤有关。综合来看,这三个维度可能代表(1)与病理和临床进展相关的脱髓鞘和免疫细胞活动,(2)小胶质细胞(再)活动和可能的病变启动,以及(3)病变活动消失和瘢痕形成。我们的研究结果突出表明,透彻了解多种病理特征之间的相互作用对于了解多发性硬化症病理的异质性及其与遗传预测因素和疾病预后的关联至关重要。捐献者在这些维度上的得分可以作为一个重要的起点,进一步揭示多发性硬化症的异质性,并将其转化为对多发性硬化症患者群体的观察和干预。
{"title":"Disentangling the heterogeneity of multiple sclerosis through identification of independent neuropathological dimensions","authors":"Alyse de Boer, Aletta M. R. van den Bosch, Nienke J. Mekkes, Nina L. Fransen, Ekaterina Dagkesamanskaia, Eric Hoekstra, Jörg Hamann, Joost Smolders, Inge Huitinga, Inge R. Holtman","doi":"10.1007/s00401-024-02742-w","DOIUrl":"10.1007/s00401-024-02742-w","url":null,"abstract":"<div><p>Multiple sclerosis (MS) is a heterogeneous neurological disorder with regards to clinical presentation and pathophysiology. Here, we investigated the heterogeneity of MS by performing an exploratory factor analysis on quantitative and qualitative neuropathology data collected for 226 MS donors in the Netherlands Brain Bank autopsy cohort. Three promising dimensions were identified and subsequently validated with clinical, neuropathological, and genetic data. Dimension 1 ranged from a predominance of remyelinated and inactive lesions to extensive pathological changes, higher proportions of active and mixed lesions, and foamy microglia morphology. This pattern was positively correlated with more severe disease, the presence of B and T cells, and neuroaxonal damage. Scoring high on dimension 2 was associated with active lesions, reactive sites, and the presence of nodules. These donors had less severe disease, a specific pattern of cortical lesions, and MS risk variants in the human leukocyte antigen region, the latter indicating a connection between disease onset and this neuropathological dimension. Donors scoring high on dimension 3 showed increased lesional pathology with relatively more mixed and inactive lesions and ramified microglia morphology. This pattern was associated with longer disease duration, subpial cortical lesions, less involvement of the adaptive immune system, and less axonal damage. Taken together, the three dimensions may represent (1) demyelination and immune cell activity associated with pathological and clinical progression, (2) microglia (re)activity and possibly lesion initiation, and (3) loss of lesion activity and scar formation. Our findings highlight that a thorough understanding of the interplay between multiple pathological characteristics is crucial to understand the heterogeneity of MS pathology, as well as its association with genetic predictors and disease outcomes. The scores of donors on the dimensions can serve as an important starting point for further disentanglement of MS heterogeneity and translation into observations and interventions in living cohorts with MS.</p></div>","PeriodicalId":7012,"journal":{"name":"Acta Neuropathologica","volume":"147 1","pages":""},"PeriodicalIF":9.3,"publicationDate":"2024-05-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11108935/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141070255","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-05-18DOI: 10.1007/s00401-024-02729-7
Michael J. Ellis, Christiana Lekka, Katie L. Holden, Hanna Tulmin, Faheem Seedat, Darragh P. O’Brien, Shalinee Dhayal, Marie-Louise Zeissler, Jakob G. Knudsen, Benedikt M. Kessler, Noel G. Morgan, John A. Todd, Sarah J. Richardson, M. Irina Stefana
Antibodies are essential research tools whose performance directly impacts research conclusions and reproducibility. Owing to its central role in Alzheimer’s disease and other dementias, hundreds of distinct antibody clones have been developed against the microtubule-associated protein Tau and its multiple proteoforms. Despite this breadth of offer, limited understanding of their performance and poor antibody selectivity have hindered research progress. Here, we validate a large panel of Tau antibodies by Western blot (79 reagents) and immunohistochemistry (35 reagents). We address the reagents’ ability to detect the target proteoform, selectivity, the impact of protein phosphorylation on antibody binding and performance in human brain samples. While most antibodies detected Tau at high levels, many failed to detect it at lower, endogenous levels. By WB, non-selective binding to other proteins affected over half of the antibodies tested, with several cross-reacting with the related MAP2 protein, whereas the “oligomeric Tau” T22 antibody reacted with monomeric Tau by WB, thus calling into question its specificity to Tau oligomers. Despite the presumption that “total” Tau antibodies are agnostic to post-translational modifications, we found that phosphorylation partially inhibits binding for many such antibodies, including the popular Tau-5 clone. We further combine high-sensitivity reagents, mass-spectrometry proteomics and cDNA sequencing to demonstrate that presumptive Tau “knockout” human cells continue to express residual protein arising through exon skipping, providing evidence of previously unappreciated gene plasticity. Finally, probing of human brain samples with a large panel of antibodies revealed the presence of C-term-truncated versions of all main Tau brain isoforms in both control and tauopathy donors. Ultimately, we identify a validated panel of Tau antibodies that can be employed in Western blotting and/or immunohistochemistry to reliably detect even low levels of Tau expression with high selectivity. This work represents an extensive resource that will enable the re-interpretation of published data, improve reproducibility in Tau research, and overall accelerate scientific progress.
抗体是重要的研究工具,其性能直接影响研究结论和可重复性。由于 Tau 在阿尔茨海默病和其他痴呆症中的核心作用,针对微管相关蛋白 Tau 及其多种蛋白形式开发了数百种不同的抗体克隆。尽管提供的抗体种类繁多,但对其性能的了解有限以及抗体选择性差阻碍了研究的进展。在此,我们通过 Western 印迹(79 种试剂)和免疫组化(35 种试剂)验证了大量 Tau 抗体。我们讨论了这些试剂检测目标蛋白形式的能力、选择性、蛋白磷酸化对抗体结合的影响以及在人脑样本中的表现。虽然大多数抗体都能检测到高水平的 Tau,但许多抗体却不能检测到较低水平的内源性 Tau。通过WB检测,与其他蛋白质的非选择性结合影响了半数以上的受试抗体,其中几种抗体与相关的MAP2蛋白发生交叉反应,而 "寡聚Tau "T22抗体则与单体Tau发生WB反应,从而使其对Tau寡聚体的特异性受到质疑。尽管 "全 "Tau抗体与翻译后修饰无关,但我们发现磷酸化会部分抑制许多此类抗体的结合,包括流行的Tau-5克隆。我们进一步将高灵敏度试剂、质谱蛋白质组学和 cDNA 测序结合起来,证明推定的 Tau "敲除 "人体细胞继续表达通过外显子跳越产生的残余蛋白,为以前未被认识到的基因可塑性提供了证据。最后,用大量抗体对人脑样本进行检测发现,在对照组和牛磺酸脑病供体中都存在所有主要Tau脑异构体的C端截短版本。最终,我们确定了一组经过验证的 Tau 抗体,可用于 Western 印迹分析和/或免疫组化,以高选择性可靠地检测低水平的 Tau 表达。这项工作代表了一种广泛的资源,将有助于重新解释已发表的数据,提高 Tau 研究的可重复性,并全面加速科学进步。
{"title":"Identification of high-performing antibodies for the reliable detection of Tau proteoforms by Western blotting and immunohistochemistry","authors":"Michael J. Ellis, Christiana Lekka, Katie L. Holden, Hanna Tulmin, Faheem Seedat, Darragh P. O’Brien, Shalinee Dhayal, Marie-Louise Zeissler, Jakob G. Knudsen, Benedikt M. Kessler, Noel G. Morgan, John A. Todd, Sarah J. Richardson, M. Irina Stefana","doi":"10.1007/s00401-024-02729-7","DOIUrl":"10.1007/s00401-024-02729-7","url":null,"abstract":"<div><p>Antibodies are essential research tools whose performance directly impacts research conclusions and reproducibility. Owing to its central role in Alzheimer’s disease and other dementias, hundreds of distinct antibody clones have been developed against the microtubule-associated protein Tau and its multiple proteoforms. Despite this breadth of offer, limited understanding of their performance and poor antibody selectivity have hindered research progress. Here, we validate a large panel of Tau antibodies by Western blot (79 reagents) and immunohistochemistry (35 reagents). We address the reagents’ ability to detect the target proteoform, selectivity, the impact of protein phosphorylation on antibody binding and performance in human brain samples. While most antibodies detected Tau at high levels, many failed to detect it at lower, endogenous levels. By WB, non-selective binding to other proteins affected over half of the antibodies tested, with several cross-reacting with the related MAP2 protein, whereas the “oligomeric Tau” T22 antibody reacted with monomeric Tau by WB, thus calling into question its specificity to Tau oligomers. Despite the presumption that “total” Tau antibodies are agnostic to post-translational modifications, we found that phosphorylation partially inhibits binding for many such antibodies, including the popular Tau-5 clone. We further combine high-sensitivity reagents, mass-spectrometry proteomics and cDNA sequencing to demonstrate that presumptive Tau “knockout” human cells continue to express residual protein arising through exon skipping, providing evidence of previously unappreciated gene plasticity. Finally, probing of human brain samples with a large panel of antibodies revealed the presence of C-term-truncated versions of all main Tau brain isoforms in both control and tauopathy donors. Ultimately, we identify a validated panel of Tau antibodies that can be employed in Western blotting and/or immunohistochemistry to reliably detect even low levels of Tau expression with high selectivity. This work represents an extensive resource that will enable the re-interpretation of published data, improve reproducibility in Tau research, and overall accelerate scientific progress.</p></div>","PeriodicalId":7012,"journal":{"name":"Acta Neuropathologica","volume":"147 1","pages":""},"PeriodicalIF":9.3,"publicationDate":"2024-05-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://link.springer.com/content/pdf/10.1007/s00401-024-02729-7.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140953964","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}