8,702 results
Search Results
2. Nothing is wrong with descriptive papers.
- Author
-
Paulus W
- Subjects
- Animals, Humans, Nervous System Diseases pathology, Nervous System Diseases physiopathology, Neurology methods, Pathology methods, Observational Studies as Topic, Publishing, Research Design
- Published
- 2014
- Full Text
- View/download PDF
3. The controversial paper.
- Author
-
Paulus W
- Subjects
- Editorial Policies, Neurology standards, Pathology standards, Peer Review, Research, Publishing standards, Neurology trends, Pathology trends, Periodicals as Topic
- Published
- 2013
- Full Text
- View/download PDF
4. Nothing is wrong with descriptive papers
- Author
-
Werner Paulus
- Subjects
Publishing ,Cognitive science ,Identifying problems ,Disease ,Neuropathology ,Pathology and Forensic Medicine ,Reality check ,Observational Studies as Topic ,Cellular and Molecular Neuroscience ,Neurology ,Research Design ,Nothing ,Human material ,Pathology ,Animals ,Humans ,Relevance (law) ,Neurology (clinical) ,Nervous System Diseases ,Psychology - Abstract
while descriptive studies often tend to be more relevant. this means that mechanistic studies need to be backed up by a reality check to make them relevant, and neuropathology is in a unique position to undertake mechanistic studies AND verification using human specimens. Studies on mechanisms without checking against the reality of human tissues may be an excellent science, but may be in danger of becoming a kind of neuroscience in the clinical nirvana, and do not represent neuropathology. this self-conception is reflected in the subtitle of Acta Neuropathologica, namely “Pathology and Mechanisms of Neurological Disease”. Accordingly, we love to publish excellent, interesting AND relevant papers analyzing mechanisms of disease using in vitro or in vivo models and complemented by appropriate analyses of human tissues. the same high priority is granted to excellent descriptive papers on tissue-based morphological or molecular features of neurological disease bearing an important clinical impact. Mechanistic studies without verification in human tissues may be important and can also be published in this journal, but their priority is somewhat lower if there are no apparent clinical implications. the same is true for descriptive studies using animal models without providing experimental insight into pathogenesis. Neuropathology is not merely neuroscience of neurological disease. Neuropathology is identifying problems through the examination of human pathological material, then designing experiments to test mechanistic hypotheses and finally confirming the relevance of these hypotheses by expert re-examination of human material. Acta Neuropathologica is about to promote this view.
- Published
- 2014
- Full Text
- View/download PDF
5. Addendum to the paper
- Published
- 1966
- Full Text
- View/download PDF
6. Mission not yet completed: on the ups and downs of being the editor of ANP.
- Author
-
Paulus, Werner
- Subjects
PUBLICATIONS ,EDITORIAL boards - Abstract
The author presents his experience of working as an editor for the journal "Acta Neuropathologica" (ANP), various publications, submissions, and quality and relevance of published papers and also gives thanks to the editorial boards and discusses his emotional and cognitive experience.
- Published
- 2019
- Full Text
- View/download PDF
7. Diverse human astrocyte and microglial transcriptional responses to Alzheimer’s pathology
- Author
-
Amy M. Smith, Paul M. Matthews, David R. Owen, Maria Weinert, Robert C. J. Muirhead, Aisling McGarry, Karen Davey, Steve M. Gentleman, Combiz Khozoie, Eirini Liaptsi, Stergios Tsartsalis, Nurun Fancy, and See Swee Tang
- Subjects
Male ,Apolipoprotein E ,Pathology ,medicine.medical_specialty ,Cell type ,Amyloid beta ,Inflammation ,Biology ,Pathology and Forensic Medicine ,ddc:616.89 ,Cellular and Molecular Neuroscience ,Alzheimer Disease ,Gene expression ,medicine ,Humans ,Aged ,Aged, 80 and over ,Metal ion homeostasis ,Original Paper ,GPNMB ,snRNA sequencing ,Microglia ,Brain ,medicine.anatomical_structure ,Proteostasis ,Astrocytes ,biology.protein ,Female ,Neurology (clinical) ,medicine.symptom ,Amyloid-beta ,Tau ,Transcriptome ,Alzheimer’s disease ,Astrocyte - Abstract
To better define roles that astrocytes and microglia play in Alzheimer’s disease (AD), we used single-nuclei RNA-sequencing to comprehensively characterise transcriptomes in astrocyte and microglia nuclei selectively enriched during isolation post-mortem from neuropathologically defined AD and control brains with a range of amyloid-beta and phospho-tau (pTau) pathology. Significant differences in glial gene expression (including AD risk genes expressed in both the astrocytes [CLU, MEF2C, IQCK] and microglia [APOE, MS4A6A, PILRA]) were correlated with tissue amyloid or pTau expression. The differentially expressed genes were distinct between with the two cell types and pathologies, although common (but cell-type specific) gene sets were enriched with both pathologies in each cell type. Astrocytes showed enrichment for proteostatic, inflammatory and metal ion homeostasis pathways. Pathways for phagocytosis, inflammation and proteostasis were enriched in microglia and perivascular macrophages with greater tissue amyloid, but IL1-related pathway enrichment was found specifically in association with pTau. We also found distinguishable sub-clusters in the astrocytes and microglia characterised by transcriptional signatures related to either homeostatic functions or disease pathology. Gene co-expression analyses revealed potential functional associations of soluble biomarkers of AD in astrocytes (CLU) and microglia (GPNMB). Our work highlights responses of both astrocytes and microglia for pathological protein clearance and inflammation, as well as glial transcriptional diversity in AD.
- Published
- 2021
- Full Text
- View/download PDF
8. Recurrent fusions in PLAGL1 define a distinct subset of pediatric-type supratentorial neuroepithelial tumors
- Author
-
Konstantin Okonechnikov, Christina Blume, Mariëtte E.G. Kranendonk, Stephanie Bunkowski, Dominik Sturm, Matija Snuderl, David R Ghasemi, Damian Stichel, Philipp Sievers, David T.W. Jones, Richard Grundy, Christian Mawrin, Ofelia Cruz, Andreas von Deimling, David W. Ellison, Tuyu Zheng, Daniel Schrimpf, Mark R. Gilbert, Lenka Krskova, Pascale Varlet, Hildegard Dohmen, Till Acker, Henning B. Boldt, Sophie C Henneken, Kenneth Aldape, Stefan Rutkowski, Mariona Suñol, Andrey Korshunov, Stefan M. Pfister, Julia Benzel, David Capper, Wolf Mueller, Ulrich Schüller, Sebastian Brandner, Patrick N. Harter, Zied Abdullaev, Celso Pouget, Rudi Beschorner, Kendra K Maaß, Viktoria Ruf, Pieter Wesseling, Mélanie Pagès, Nada Jabado, Terri S. Armstrong, Patricia Kohlhof-Meinecke, Martin Sill, Marcel Kool, Koichi Ichimura, Felix Sahm, Guido Reifenberger, Kristian W. Pajtler, Wolfgang Wick, David E. Reuss, Leonille Schweizer, Christine Stadelmann, Cinzia Lavarino, Ales Vicha, Michal Zapotocky, Noreen Akhtar, Pathology, CCA - Cancer biology and immunology, and CCA - Imaging and biomarkers
- Subjects
Ependymoma ,Male ,Pathology ,medicine.medical_specialty ,SOX10 ,Cell Cycle Proteins ,Biology ,Supratentorial ,Pathology and Forensic Medicine ,Neuroepithelial tumor ,Fusion gene ,Pleomorphic adenoma ,OLIG2 ,Cellular and Molecular Neuroscience ,FOXO1 ,medicine ,Humans ,Oncogene Fusion ,ddc:610 ,EP300 ,Child ,PLAGL1 ,Original Paper ,Tumor Suppressor Proteins ,EWSR1 ,Supratentorial Neoplasms ,medicine.disease ,DNA methylation ,Gene fusion ,Female ,Neurology (clinical) ,Genomic imprinting ,Transcription Factors - Abstract
Ependymomas encompass a heterogeneous group of central nervous system (CNS) neoplasms that occur along the entire neuroaxis. In recent years, extensive (epi-)genomic profiling efforts have identified several molecular groups of ependymoma that are characterized by distinct molecular alterations and/or patterns. Based on unsupervised visualization of a large cohort of genome-wide DNA methylation data, we identified a highly distinct group of pediatric-type tumors (n = 40) forming a cluster separate from all established CNS tumor types, of which a high proportion were histopathologically diagnosed as ependymoma. RNA sequencing revealed recurrent fusions involving the pleomorphic adenoma gene-like 1 (PLAGL1) gene in 19 of 20 of the samples analyzed, with the most common fusion being EWSR1:PLAGL1 (n = 13). Five tumors showed a PLAGL1:FOXO1 fusion and one a PLAGL1:EP300 fusion. High transcript levels of PLAGL1 were noted in these tumors, with concurrent overexpression of the imprinted genes H19 and IGF2, which are regulated by PLAGL1. Histopathological review of cases with sufficient material (n = 16) demonstrated a broad morphological spectrum of tumors with predominant ependymoma-like features. Immunohistochemically, tumors were GFAP positive and OLIG2- and SOX10 negative. In 3/16 of the cases, a dot-like positivity for EMA was detected. All tumors in our series were located in the supratentorial compartment. Median age of the patients at the time of diagnosis was 6.2 years. Median progression-free survival was 35 months (for 11 patients with data available). In summary, our findings suggest the existence of a novel group of supratentorial neuroepithelial tumors that are characterized by recurrent PLAGL1 fusions and enriched for pediatric patients.
- Published
- 2021
- Full Text
- View/download PDF
9. Immunisation with UB-312 in the Thy1SNCA mouse prevents motor performance deficits and oligomeric α-synuclein accumulation in the brain and gut
- Author
-
James A. R. Nicoll, Harry Smith, Jean-Cosme Dodart, Ajay Verma, Jacqui T. Nimmo, Zhi Liu, Chang Yi Wang, Jessica L. Teeling, Roxana O. Carare, and Feng Lin
- Subjects
Thy1SNCA ,Gastrointestinal pathology ,Synucleinopathies ,medicine.medical_treatment ,Motor behaviour ,Hippocampus ,Mice, Transgenic ,Striatum ,Protein Aggregation, Pathological ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Mice ,Atrophy ,Parkinsonian Disorders ,medicine ,Alpha synuclein ,Animals ,Humans ,Alpha-synuclein ,Original Paper ,business.industry ,Dementia with Lewy bodies ,Vaccination ,Brain ,Immunotherapy ,medicine.disease ,Intestines ,Disease Models, Animal ,medicine.anatomical_structure ,chemistry ,Cerebral cortex ,Immunology ,Vaccines, Subunit ,alpha-Synuclein ,Neurology (clinical) ,business - Abstract
Alpha synuclein has a key role in the pathogenesis of Parkinson’s disease (PD), Dementia with Lewy Bodies (LBD) and Multiple System Atrophy (MSA). Immunotherapies aiming at neutralising toxic αSyn species are being investigated in the clinic as potential disease modifying therapies for PD and other synucleinopathies. In this study, the effects of active immunisation against αSyn with the UB-312 vaccine were investigated in the Thy1SNCA/15 mouse model of PD. Young transgenic and wild-type mice received an immunisation regimen over a period of 6 weeks, then observed for an additional 9 weeks. Behavioural assessment was conducted before immunisation and at 15 weeks after the first dose. UB-312 immunisation prevented the development of motor impairment in the wire test and challenging beam test, which was associated with reduced levels of αSyn oligomers in the cerebral cortex, hippocampus and striatum of Thy1SNCA/15 mice. UB-312 immunotherapy resulted in a significant reduction of theαSyn load in the colon, accompanied by a reduction in enteric glial cell reactivity in the colonic ganglia. Our results demonstrate that immunisation with UB-312 prevents functional deficits and both central and peripheral pathology in Thy1SNCA/15 mice. Supplementary Information The online version contains supplementary material available at 10.1007/s00401-021-02381-5.
- Published
- 2021
10. Frontal white matter lesions in Alzheimer’s disease are associated with both small vessel disease and AD-associated cortical pathology
- Author
-
Mohi Miah, Charles DeCarli, Mary Johnson, Lauren Walker, David J. Koss, Georgina M. Hadfield, Sophie Graham, Johannes Attems, Sean J. Colloby, Alan J. Thomas, and Kirsty E. McAleese
- Subjects
Male ,Aging ,Pathology ,Axonal loss ,Pilot Projects ,Neurodegenerative ,Alzheimer's Disease ,80 and over ,2.1 Biological and endogenous factors ,White matter hyperintensity ,Aetiology ,Aged, 80 and over ,biology ,Parietal lobe ,Intracranial Arteriosclerosis ,White Matter ,Small vessel disease ,Frontal Lobe ,medicine.anatomical_structure ,Neurological ,Female ,Amyloid-beta ,Alzheimer’s disease ,medicine.medical_specialty ,Amyloid beta ,Clinical Sciences ,Arteriolosclerosis ,Ischemia ,Pathology and Forensic Medicine ,White matter ,Cellular and Molecular Neuroscience ,Neuroimaging ,Clinical Research ,White matter lesion ,Alzheimer Disease ,Acquired Cognitive Impairment ,medicine ,Humans ,Dementia ,Hyperphosphorylated tau ,Aged ,Original Paper ,Neurology & Neurosurgery ,business.industry ,Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,medicine.disease ,Brain Disorders ,Cerebral Small Vessel Diseases ,biology.protein ,Neurology (clinical) ,business - Abstract
Cerebral white matter lesions (WML) encompass axonal loss and demyelination and are assumed to be associated with small vessel disease (SVD)-related ischaemia. However, our previous study in the parietal lobe white matter revealed that WML in Alzheimer’s disease (AD) are linked with degenerative axonal loss secondary to the deposition of cortical AD pathology. Furthermore, neuroimaging data suggest that pathomechanisms for the development of WML differ between anterior and posterior lobes with AD-associated degenerative mechanism driving posterior white matter disruption, and both AD-associated degenerative and vascular mechanisms contributed to anterior matter disruption. In this pilot study, we used human post-mortem brain tissue to investigate the composition and aetiology of frontal WML from AD and non-demented controls to determine if frontal WML are SVD-associated and to reveal any regional differences in the pathogenesis of WML. Frontal WML tissue sections from 40 human post-mortem brains (AD, n = 19; controls, n = 21) were quantitatively assessed for demyelination, axonal loss, cortical hyperphosphorylated tau (HPτ) and amyloid-beta (Aβ) burden, and arteriolosclerosis as a measure of SVD. Biochemical assessment included Wallerian degeneration-associated protease calpain and the myelin-associated glycoprotein to proteolipid protein ratio as a measure of ante-mortem ischaemia. Arteriolosclerosis severity was found to be associated with and a significant predictor of frontal WML severity in both AD and non-demented controls. Interesting, frontal axonal loss was also associated with HPτ and calpain levels were associated with increasing Aβ burden in the AD group, suggestive of an additional degenerative influence. To conclude, this pilot data suggest that frontal WML in AD may result from both increased arteriolosclerosis and AD-associated degenerative changes. These preliminary findings in combination with previously published data tentatively indicate regional differences in the aetiology of WML in AD, which should be considered in the clinical diagnosis of dementia subtypes: posterior WML maybe associated with degenerative mechanisms secondary to AD pathology, while anterior WML could be associated with both SVD-associated and degenerative mechanisms.
- Published
- 2021
- Full Text
- View/download PDF
11. Remarks on the papers by C.-D. agardh et al./H. Kalimo et al
- Author
-
A. W. Brown and J. B. Brierley
- Subjects
Cellular and Molecular Neuroscience ,Philosophy ,Neurology (clinical) ,Humanities ,Pathology and Forensic Medicine - Published
- 1981
- Full Text
- View/download PDF
12. The subcellular arrangement of alpha-synuclein proteoforms in the Parkinson’s disease brain as revealed by multicolor STED microscopy
- Author
-
Markus Britschgi, Thomas Kremer, Klaus Kaluza, Jeroen Kole, Evelien Timmermans-Huisman, Christina A. Maat, Wagner Zago, Mirko Ritter, Olaf Mundigl, Tim Moors, Robin Barbour, Daniel Niedieker, Samir F. El-Mashtoly, Jeroen J. G. Geurts, Liz Spycher, Melanie N. Hug, Sebastian Dziadek, Klaus Gerwert, Wilma D.J. van de Berg, Sylwia Huber, Dennis Petersen, Daniel Mona, Anatomy and neurosciences, Amsterdam Neuroscience - Neurodegeneration, and Amsterdam Neuroscience - Neuroinfection & -inflammation
- Subjects
Male ,Cytoplasm ,Parkinson's disease ,Pathology and Forensic Medicine ,Alpha-synuclein ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,medicine ,Humans ,Super-resolution microscopy ,Cytoskeleton ,Aged ,Biological Specimen Banks ,Brain Chemistry ,Inclusion Bodies ,Neurons ,Original Paper ,Microscopy, Confocal ,Post-mortem human brain ,STED microscopy ,Parkinson Disease ,Human brain ,Middle Aged ,medicine.disease ,Phenotype ,Cell biology ,medicine.anatomical_structure ,chemistry ,Lewy Bodies ,Neurology (clinical) ,Protein Processing, Post-Translational ,Subcellular Fractions ,Post-translational modifications - Abstract
Various post-translationally modified (PTM) proteoforms of alpha-synuclein (aSyn)—including C-terminally truncated (CTT) and Serine 129 phosphorylated (Ser129-p) aSyn—accumulate in Lewy bodies (LBs) in different regions of the Parkinson’s disease (PD) brain. Insight into the distribution of these proteoforms within LBs and subcellular compartments may aid in understanding the orchestration of Lewy pathology in PD. We applied epitope-specific antibodies against CTT and Ser129-p aSyn proteoforms and different aSyn domains in immunohistochemical multiple labelings on post-mortem brain tissue from PD patients and non-neurological, aged controls, which were scanned using high-resolution 3D multicolor confocal and stimulated emission depletion (STED) microscopy. Our multiple labeling setup highlighted a consistent onion skin-type 3D architecture in mature nigral LBs in which an intricate and structured-appearing framework of Ser129-p aSyn and cytoskeletal elements encapsulates a core enriched in CTT aSyn species. By label-free CARS microscopy we found that enrichments of proteins and lipids were mainly localized to the central portion of nigral aSyn-immunopositive (aSyn+) inclusions. Outside LBs, we observed that 122CTT aSyn+ punctae localized at mitochondrial membranes in the cytoplasm of neurons in PD and control brains, suggesting a physiological role for 122CTT aSyn outside of LBs. In contrast, very limited to no Ser129-p aSyn immunoreactivity was observed in brains of non-neurological controls, while the alignment of Ser129-p aSyn in a neuronal cytoplasmic network was characteristic for brains with (incidental) LB disease. Interestingly, Ser129-p aSyn+ network profiles were not only observed in neurons containing LBs but also in neurons without LBs particularly in donors at early disease stage, pointing towards a possible subcellular pathological phenotype preceding LB formation. Together, our high-resolution and 3D multicolor microscopy observations in the post-mortem human brain provide insights into potential mechanisms underlying a regulated LB morphogenesis. Supplementary Information The online version contains supplementary material available at 10.1007/s00401-021-02329-9.
- Published
- 2021
- Full Text
- View/download PDF
13. Apolipoprotein E regulates lipid metabolism and α-synuclein pathology in human iPSC-derived cerebral organoids
- Author
-
Guojun Bu, Wenyan Lu, Xianlin Han, Jing Zhao, Xue Wang, Mary D. Davis, Meixia Pan, Nilufer Ertekin-Taner, Steven G. Younkin, Chia Chen Liu, Fuyao Li, Zbigniew K. Wszolek, Yuka A. Martens, Yan W. Asmann, Ziying Xu, Yuan Fu, Kai Chen, Neill R. Graff-Radford, Francis Shue, David A. Brafman, Takahisa Kanekiyo, Yingxue Ren, and Dennis W. Dickson
- Subjects
Apolipoprotein E ,Pathology ,medicine.medical_specialty ,Synucleinopathies ,Induced Pluripotent Stem Cells ,Biology ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Mice ,Apolipoproteins E ,Lipidomics ,mental disorders ,Organoid ,medicine ,Animals ,Humans ,Protein Isoforms ,Original Paper ,Cholesterol ,Dementia with Lewy bodies ,Correction ,Lipid metabolism ,medicine.disease ,Lipid Metabolism ,Organoids ,chemistry ,alpha-Synuclein ,lipids (amino acids, peptides, and proteins) ,Neurology (clinical) ,Cerebral organoid - Abstract
APOE4 is a strong genetic risk factor for Alzheimer’s disease and Dementia with Lewy bodies; however, how its expression impacts pathogenic pathways in a human-relevant system is not clear. Here using human iPSC-derived cerebral organoid models, we find that APOE deletion increases α-synuclein (αSyn) accumulation accompanied with synaptic loss, reduction of GBA levels, lipid droplet accumulation and dysregulation of intracellular organelles. These phenotypes are partially rescued by exogenous apoE2 and apoE3, but not apoE4. Lipidomics analysis detects the increased fatty acid utilization and cholesterol ester accumulation in apoE-deficient cerebral organoids. Furthermore, APOE4 cerebral organoids have increased αSyn accumulation compared to those with APOE3. Carrying APOE4 also increases apoE association with Lewy bodies in postmortem brains from patients with Lewy body disease. Our findings reveal the predominant role of apoE in lipid metabolism and αSyn pathology in iPSC-derived cerebral organoids, providing mechanistic insights into how APOE4 drives the risk for synucleinopathies.
- Published
- 2021
14. Seizure-mediated iron accumulation and dysregulated iron metabolism after status epilepticus and in temporal lobe epilepsy
- Author
-
James D. Mills, Eleonora Aronica, Wim Van Hecke, Till S. Zimmer, Stefanie Dedeurwaerdere, Johannes C. Baayen, Sander Idema, Nicole N. van der Wel, Anatoly Korotkov, Jonathan van Eyll, Peter C. van Rijen, Rainer Surges, Erwin A. van Vliet, Helmut W. Kessels, Jan A. Gorter, Diede W. M. Broekaart, Angelika Mühlebner, Martin Schidlowski, Theodor Rüber, Bastian David, Gabriele Ruffolo, Liesbeth François, Cellular and Computational Neuroscience (SILS, FNWI), Graduate School, APH - Aging & Later Life, APH - Mental Health, ANS - Cellular & Molecular Mechanisms, Pathology, Medical Biology, ANS - Neurodegeneration, and AGEM - Amsterdam Gastroenterology Endocrinology Metabolism
- Subjects
Male ,Astrocytes ,Glutathione metabolism ,Iron ,Status epilepticus ,Temporal lobe epilepsy with hippocampal sclerosis ,0301 basic medicine ,Cell Culture Techniques ,Hippocampus ,metabolism [Hippocampus] ,Hippocampal formation ,physiology [Oxidative Stress] ,Epilepsy ,0302 clinical medicine ,etiology [Iron Metabolism Disorders] ,pathology [Astrocytes] ,metabolism [Iron] ,Aged, 80 and over ,metabolism [Astrocytes] ,biology ,Middle Aged ,metabolism [Status Epilepticus] ,medicine.anatomical_structure ,pathology [Status Epilepticus] ,Female ,metabolism [Epilepsy, Temporal Lobe] ,medicine.symptom ,Astrocyte ,Adult ,medicine.medical_specialty ,pathology [Epilepsy, Temporal Lobe] ,complications [Status Epilepticus] ,Pathology and Forensic Medicine ,Temporal lobe ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Internal medicine ,medicine ,Animals ,Humans ,ddc:610 ,Aged ,Original Paper ,Hippocampal sclerosis ,business.industry ,complications [Epilepsy, Temporal Lobe] ,medicine.disease ,Iron Metabolism Disorders ,Rats ,Ferritin ,Oxidative Stress ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Epilepsy, Temporal Lobe ,Case-Control Studies ,pathology [Iron Metabolism Disorders] ,biology.protein ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
Neuronal dysfunction due to iron accumulation in conjunction with reactive oxygen species (ROS) could represent an important, yet underappreciated, component of the epileptogenic process. However, to date, alterations in iron metabolism in the epileptogenic brain have not been addressed in detail. Iron-related neuropathology and antioxidant metabolic processes were investigated in resected brain tissue from patients with temporal lobe epilepsy and hippocampal sclerosis (TLE-HS), post-mortem brain tissue from patients who died after status epilepticus (SE) as well as brain tissue from the electrically induced SE rat model of TLE. Magnetic susceptibility of the presumed seizure-onset zone from three patients with focal epilepsy was compared during and after seizure activity. Finally, the cellular effects of iron overload were studied in vitro using an acute mouse hippocampal slice preparation and cultured human fetal astrocytes. While iron-accumulating neurons had a pyknotic morphology, astrocytes appeared to acquire iron-sequestrating capacity as indicated by prominent ferritin expression and iron retention in the hippocampus of patients with SE or TLE. Interictal to postictal comparison revealed increased magnetic susceptibility in the seizure-onset zone of epilepsy patients. Post-SE rats had consistently higher hippocampal iron levels during the acute and chronic phase (when spontaneous recurrent seizures are evident). In vitro, in acute slices that were exposed to iron, neurons readily took up iron, which was exacerbated by induced epileptiform activity. Human astrocyte cultures challenged with iron and ROS increased their antioxidant and iron-binding capacity, but simultaneously developed a pro-inflammatory phenotype upon chronic exposure. These data suggest that seizure-mediated, chronic neuronal iron uptake might play a role in neuronal dysfunction/loss in TLE-HS. On the other hand, astrocytes sequester iron, specifically in chronic epilepsy. This function might transform astrocytes into a highly resistant, pro-inflammatory phenotype potentially contributing to pro-epileptogenic inflammatory processes. Supplementary Information The online version contains supplementary material available at 10.1007/s00401-021-02348-6.
- Published
- 2021
- Full Text
- View/download PDF
15. HnRNP K mislocalisation is a novel protein pathology of frontotemporal lobar degeneration and ageing and leads to cryptic splicing
- Author
-
Pietro Fratta, Dipanjan Bhattacharya, Towfique Raj, Sara Cappelli, Jack Humphrey, Anna-Leigh Brown, Tammaryn Lashley, Marco Foiani, Yi Hua Low, Sandrine C. Foti, Yasmine T. Asi, Emanuele Buratti, Ariana Gatt, and Alexander Bampton
- Subjects
0301 basic medicine ,Adult ,Male ,Aging ,viruses ,RNA Splicing ,genetic processes ,Context (language use) ,Biology ,Heterogeneous ribonucleoprotein particle ,Frontotemporal lobar degeneration ,environment and public health ,Pathology and Forensic Medicine ,Heterogeneous-Nuclear Ribonucleoprotein K ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Exon ,0302 clinical medicine ,Stress granule ,medicine ,Humans ,Aged ,Aged, 80 and over ,Original Paper ,hnRNP K ,RNA ,Cryptic exons ,Middle Aged ,medicine.disease ,Cell biology ,Ageing ,030104 developmental biology ,Case-Control Studies ,RNA splicing ,health occupations ,Female ,Neurology (clinical) ,030217 neurology & neurosurgery ,Frontotemporal dementia - Abstract
Heterogeneous nuclear ribonucleoproteins (HnRNPs) are a group of ubiquitously expressed RNA-binding proteins implicated in the regulation of all aspects of nucleic acid metabolism. HnRNP K is a member of this highly versatile hnRNP family. Pathological redistribution of hnRNP K to the cytoplasm has been linked to the pathogenesis of several malignancies but, until now, has been underexplored in the context of neurodegenerative disease. Here we show hnRNP K mislocalisation in pyramidal neurons of the frontal cortex to be a novel neuropathological feature that is associated with both frontotemporal lobar degeneration and ageing. HnRNP K mislocalisation is mutually exclusive to TDP-43 and tau pathological inclusions in neurons and was not observed to colocalise with mitochondrial, autophagosomal or stress granule markers. De-repression of cryptic exons in RNA targets following TDP-43 nuclear depletion is an emerging mechanism of potential neurotoxicity in frontotemporal lobar degeneration and the mechanistically overlapping disorder amyotrophic lateral sclerosis. We silenced hnRNP K in neuronal cells to identify the transcriptomic consequences of hnRNP K nuclear depletion. Intriguingly, by performing RNA-seq analysis we find that depletion of hnRNP K induces 101 novel cryptic exon events. We validated cryptic exon inclusion in an SH-SY5Y hnRNP K knockdown and in FTLD brain exhibiting hnRNP K nuclear depletion. We, therefore, present evidence for hnRNP K mislocalisation to be associated with FTLD and for this to induce widespread changes in splicing. Supplementary Information The online version contains supplementary material available at 10.1007/s00401-021-02340-0.
- Published
- 2021
16. Structure of Tau filaments in Prion protein amyloidoses
- Author
-
G.I. Hallinan, Ruben Vidal, Anllely Fernandez, Manali Ghosh, Holly J. Garringer, Frank S. Vago, Rejaul Hoq, Bernardino Ghetti, and Wen Jiang
- Subjects
Amyloid ,Prions ,Tau protein ,Nonsense mutation ,Plaque, Amyloid ,tau Proteins ,Prion Proteins ,Pathology and Forensic Medicine ,PRNP ,Cellular and Molecular Neuroscience ,PrP-CAA ,Alzheimer Disease ,mental disorders ,medicine ,Gerstmann-Straussler-Scheinker Disease ,Humans ,GSS ,Neurodegeneration ,Cryo-EM ,Original Paper ,biology ,Chemistry ,APrP ,Brain ,Neurofibrillary Tangles ,Amyloidosis ,medicine.disease ,Molecular biology ,Corticobasal Degeneration ,Chronic traumatic encephalopathy ,Phenotype ,biology.protein ,Neurology (clinical) ,Cerebral amyloid angiopathy ,Tau ,Alzheimer's disease - Abstract
In human neurodegenerative diseases associated with the intracellular aggregation of Tau protein, the ordered cores of Tau filaments adopt distinct folds. Here, we analyze Tau filaments isolated from the brain of individuals affected by Prion-Protein cerebral amyloid angiopathy (PrP-CAA) with a nonsense mutation in the PRNP gene that leads to early termination of translation of PrP (Q160Ter or Q160X), and Gerstmann–Sträussler–Scheinker (GSS) disease, with a missense mutation in the PRNP gene that leads to an amino acid substitution at residue 198 (F198S) of PrP. The clinical and neuropathologic phenotypes associated with these two mutations in PRNP are different; however, the neuropathologic analyses of these two genetic variants have consistently shown the presence of numerous neurofibrillary tangles (NFTs) made of filamentous Tau aggregates in neurons. We report that Tau filaments in PrP-CAA (Q160X) and GSS (F198S) are composed of 3-repeat and 4-repeat Tau isoforms, having a striking similarity to NFTs in Alzheimer disease (AD). In PrP-CAA (Q160X), Tau filaments are made of both paired helical filaments (PHFs) and straight filaments (SFs), while in GSS (F198S), only PHFs were found. Mass spectrometry analyses of Tau filaments extracted from PrP-CAA (Q160X) and GSS (F198S) brains show the presence of post-translational modifications that are comparable to those seen in Tau aggregates from AD. Cryo-EM analysis reveals that the atomic models of the Tau filaments obtained from PrP-CAA (Q160X) and GSS (F198S) are identical to those of the Tau filaments from AD, and are therefore distinct from those of Pick disease, chronic traumatic encephalopathy, and corticobasal degeneration. Our data support the hypothesis that in the presence of extracellular amyloid deposits and regardless of the primary amino acid sequence of the amyloid protein, similar molecular mechanisms are at play in the formation of identical Tau filaments.
- Published
- 2021
- Full Text
- View/download PDF
17. Inhibition of nuclear export restores nuclear localization and residual tumor suppressor function of truncated SMARCB1/INI1 protein in a molecular subset of atypical teratoid/rhabdoid tumors
- Author
-
Roy W. R. Dudley, Reiner Siebert, Christian Thomas, Karolina Nemes, Francesca Zin, Michael C. Frühwald, Tenzin Gayden, Rajiv Pathak, Marcel Kool, Steffen Albrecht, Florian Oyen, Pascal Johann, Martin Hasselblatt, Susanne Bens, Nada Jabado, Uwe Kordes, Werner Paulus, Jason Karamchandani, and Ganjam V. Kalpana
- Subjects
Male ,Cytoplasmic ,INI1 ,Neoplasm, Residual ,Tumor suppressor gene ,Mutant ,Malignant rhabdoid tumor ,Active Transport, Cell Nucleus ,SMARCB1 ,Selinexor ,Biology ,medicine.disease_cause ,Pathology and Forensic Medicine ,Atypical teratoid/rhabdoid tumor ,Central Nervous System Neoplasms ,Cellular and Molecular Neuroscience ,medicine ,Humans ,Genes, Tumor Suppressor ,ddc:610 ,Nuclear export signal ,Rhabdoid Tumor ,Original Paper ,Mutation ,Teratoma ,Infant ,SMARCB1 Protein ,medicine.disease ,Neoplasms, Neuroepithelial ,BAF47 ,Cytoplasm ,Child, Preschool ,Atypical teratoid rhabdoid tumor ,Cancer research ,Female ,Neurology (clinical) ,Nuclear localization sequence - Abstract
Loss of nuclear SMARCB1 (INI1/hSNF5/BAF47) protein expression due to biallelic mutations of the SMARCB1 tumor suppressor gene is a hallmark of atypical teratoid/rhabdoid tumors (ATRT), but the presence of cytoplasmic SMARCB1 protein in these tumors has not yet been described. In a series of 102 primary ATRT, distinct cytoplasmic SMARCB1 staining on immunohistochemistry was encountered in 19 cases (19%) and was highly over-represented in cases showing pathogenic sequence variants leading to truncation or mutation of the C-terminal part of SMARCB1 (15/19 vs. 4/83; Chi-square: 56.04, p = 1.0E−10) and, related to this, in tumors of the molecular subgroup ATRT-TYR (16/36 vs. 3/66; Chi-square: 24.47, p = 7.6E−7). Previous reports have indicated that while SMARCB1 lacks a bona fide nuclear localization signal, it harbors a masked nuclear export signal (NES) and that truncation of the C-terminal region results in unmasking of this NES leading to cytoplasmic localization. To determine if cytoplasmic localization found in ATRT is due to unmasking of NES, we generated GFP fusions of one of the SMARCB1 truncating mutations (p.Q318X) found in the tumors along with a p.L266A mutation, which was shown to disrupt the interaction of SMARCB1-NES with exportin-1. We found that while the GFP-SMARCB1(Q318X) mutant localized to the cytoplasm, the double mutant GFP-SMARCB1(Q318X;L266A) localized to the nucleus, confirming NES requirement for cytoplasmic localization. Furthermore, cytoplasmic SMARCB1(Q318X) was unable to cause senescence as determined by morphological observations and by senescence-associated β-galactosidase assay, while nuclear SMARCB1(Q318X;L266A) mutant regained this function. Selinexor, a selective exportin-1 inhibitor, was effective in inhibiting the nuclear export of SMARCB1(Q318X) and caused rapid cell death in rhabdoid tumor cells. In conclusion, inhibition of nuclear export restores nuclear localization and residual tumor suppressor function of truncated SMARCB1. Therapies aimed at preventing nuclear export of mutant SMARCB1 protein may represent a promising targeted therapy in ATRT harboring truncating C-terminal SMARCB1 mutations.
- Published
- 2021
- Full Text
- View/download PDF
18. Multiple system atrophy-associated oligodendroglial protein p25α stimulates formation of novel α-synuclein strain with enhanced neurodegenerative potential
- Author
-
Prakruti Rabadia, Benoit I. Giasson, Sissel Ida Schmidt, Emil Gregersen, Daniel E. Otzen, Nicolas Mendez, Katarina Willén, Marina Romero-Ramos, Cristine Betzer, Zachary A. Sorrentino, Nelson Ferreira, Mingdong Dong, Morten Meyer, Kaare Bjerregaard-Andersen, Clara Perez-Gozalbo, Ümit Akbey, Mohammad Shahnawaz, Jie Wang, Marjo Beltoja, Jan Stanislaw Nowak, Lasse Reimer, Ersoy Cholak, Poul Henning Jensen, Hjalte Gram, Claudio Soto, and Madhu Nagaraj
- Subjects
Multiple system atrophy (MSA) ,Synucleinopathies ,Protein Conformation ,Transgene ,animal diseases ,Strains ,Mice, Transgenic ,Nerve Tissue Proteins ,Protein aggregation ,Pathology and Forensic Medicine ,Cell Line ,Cellular and Molecular Neuroscience ,Mice ,In vivo ,medicine ,Animals ,Humans ,heterocyclic compounds ,Tubulin polymerisation-promoting protein (TPPP) ,Proteostasis Deficiencies ,Tropism ,Inclusion Bodies ,Original Paper ,Strain (chemistry) ,Chemistry ,Neurodegenerative Diseases ,Multiple System Atrophy ,medicine.disease ,Phenotype ,Cell biology ,nervous system diseases ,P25α ,Substantia Nigra ,Oligodendroglia ,nervous system ,Α-Synuclein ,health occupations ,alpha-Synuclein ,Neurology (clinical) ,Intracellular - Abstract
Pathology consisting of intracellular aggregates of alpha-Synuclein (α-Syn) spread through the nervous system in a variety of neurodegenerative disorders including Parkinson’s disease, dementia with Lewy bodies, and multiple system atrophy. The discovery of structurally distinct α-Syn polymorphs, so-called strains, supports a hypothesis where strain-specific structures are templated into aggregates formed by native α-Syn. These distinct strains are hypothesised to dictate the spreading of pathology in the tissue and the cellular impact of the aggregates, thereby contributing to the variety of clinical phenotypes. Here, we present evidence of a novel α-Syn strain induced by the multiple system atrophy-associated oligodendroglial protein p25α. Using an array of biophysical, biochemical, cellular, and in vivo analyses, we demonstrate that compared to α-Syn alone, a substoichiometric concentration of p25α redirects α-Syn aggregation into a unique α-Syn/p25α strain with a different structure and enhanced in vivo prodegenerative properties. The α-Syn/p25α strain induced larger inclusions in human dopaminergic neurons. In vivo, intramuscular injection of preformed fibrils (PFF) of the α-Syn/p25α strain compared to α-Syn PFF resulted in a shortened life span and a distinct anatomical distribution of inclusion pathology in the brain of a human A53T transgenic (line M83) mouse. Investigation of α-Syn aggregates in brain stem extracts of end-stage mice demonstrated that the more aggressive phenotype of the α-Syn/p25α strain was associated with an increased load of α-Syn aggregates based on a Förster resonance energy transfer immunoassay and a reduced α-Syn aggregate seeding activity based on a protein misfolding cyclic amplification assay. When injected unilaterally into the striata of wild-type mice, the α-Syn/p25α strain resulted in a more-pronounced motoric phenotype than α-Syn PFF and exhibited a “tropism” for nigro-striatal neurons compared to α-Syn PFF. Overall, our data support a hypothesis whereby oligodendroglial p25α is responsible for generating a highly prodegenerative α-Syn strain in multiple system atrophy. Supplementary Information The online version contains supplementary material available at 10.1007/s00401-021-02316-0.
- Published
- 2021
- Full Text
- View/download PDF
19. Rare germline variants in the E-cadherin gene CDH1 are associated with the risk of brain tumors of neuroepithelial and epithelial origin
- Author
-
Robert Hüneburg, Helge Martens, Martin Stangel, Andreas Erbersdobler, Wiebke Ewert, Matthias Preller, Stephan Wolf, Ruthild G. Weber, Bettina Wiese, Amir Samii, Natalie Elyan, Christopher Previti, Joachim K. Krauss, Stefan Aretz, Rouzbeh Banan, Ulrike Beyer, Bujung Hong, Christine A. M. Weber, Jan Hinrich Bräsen, Frank Brand, Alisa Förster, Jessica Kronenberg, and Christian Hartmann
- Subjects
Adenoma ,Familial glioma ,Oligodendroglioma ,Population ,medicine.disease_cause ,Germline ,Pathology and Forensic Medicine ,CDH1 ,Rats, Sprague-Dawley ,Cellular and Molecular Neuroscience ,Antigens, CD ,Glioma ,medicine ,Animals ,Humans ,ddc:610 ,Gene Knock-In Techniques ,education ,Protein Kinase Inhibitors ,Original Paper ,Whole-genome sequencing ,education.field_of_study ,Aniline Compounds ,Whole Genome Sequencing ,biology ,Brain Neoplasms ,Carcinoma ,Wnt signaling pathway ,Genetic Variation ,E-cadherin ,Cancer ,DNA, Neoplasm ,β-catenin ,Cadherins ,medicine.disease ,Neoplasms, Neuroepithelial ,Rats ,HEK293 Cells ,Purines ,Cancer research ,biology.protein ,Neurology (clinical) ,Carcinogenesis ,Antibody Diversity - Abstract
The genetic basis of brain tumor development is poorly understood. Here, leukocyte DNA of 21 patients from 15 families with ≥ 2 glioma cases each was analyzed by whole-genome or targeted sequencing. As a result, we identified two families with rare germline variants, p.(A592T) or p.(A817V), in the E-cadherin gene CDH1 that co-segregate with the tumor phenotype, consisting primarily of oligodendrogliomas, WHO grade II/III, IDH-mutant, 1p/19q-codeleted (ODs). Rare CDH1 variants, previously shown to predispose to gastric and breast cancer, were significantly overrepresented in these glioma families (13.3%) versus controls (1.7%). In 68 individuals from 28 gastric cancer families with pathogenic CDH1 germline variants, brain tumors, including a pituitary adenoma, were observed in three cases (4.4%), a significantly higher prevalence than in the general population (0.2%). Furthermore, rare CDH1 variants were identified in tumor DNA of 6/99 (6%) ODs. CDH1 expression was detected in undifferentiated and differentiating oligodendroglial cells isolated from rat brain. Functional studies using CRISPR/Cas9-mediated knock-in or stably transfected cell models demonstrated that the identified CDH1 germline variants affect cell membrane expression, cell migration and aggregation. E-cadherin ectodomain containing variant p.(A592T) had an increased intramolecular flexibility in a molecular dynamics simulation model. E-cadherin harboring intracellular variant p.(A817V) showed reduced β-catenin binding resulting in increased cytosolic and nuclear β-catenin levels reverted by treatment with the MAPK interacting serine/threonine kinase 1 inhibitor CGP 57380. Our data provide evidence for a role of deactivating CDH1 variants in the risk and tumorigenesis of neuroepithelial and epithelial brain tumors, particularly ODs, possibly via WNT/β-catenin signaling.
- Published
- 2021
- Full Text
- View/download PDF
20. TREM2 expression in the brain and biological fluids in prion diseases
- Author
-
Valerie L. Sim, José Eriton Gomes da Cunha, Niels Kruse, Óscar López-Pérez, Katrin Thüne, Enric Vidal, Peter Hermann, Inga Zerr, Miguel Calero, Henrik Zetterberg, Daniela Diaz-Lucena, Matthias Schmitz, Anna Villar-Piqué, Franc Llorens, Hailey Pineau, Alba Marín-Moreno, Raquel Sánchez-Valle, Joachim Riggert, José Antonio del Río, Kaj Blennow, Pol Andrés-Benito, Juan María Torres, Isidre Ferrer, Brit Mollenhauer, Anna Ladogana, Juan Carlos Espinosa, Generalitat de Catalunya, Instituto de Salud Carlos III, Fundació La Marató de TV3, European Commission, Swedish Research Council, Ministero della Salute, Alzheimer Society of Canada, Ministerio de Ciencia, Innovación y Universidades (España), Diaz-Lucena, Daniela, Kruse, Niels, Thüne, Katrin, Villar-Piqué, Anna, da Cunha, Jose Eriton Gomes, López-Pérez, Óscar, Andrés-Benito, Pol, Ladogana, Anna, Calero, Miguel, Vidal, Enric, Pineau, Hailey, Sim, Valerie, Zetterberg, Henrik, Blennow, Kaj, Del Río, Jose Antonio, Marín-Moreno, Alba, Espinosa, Juan Carlos, Torres, Juan María, Sánchez-Valle, Raquel, Mollenhauer, Brit, Ferrer, Isidre, Zerr, Inga, Producció Animal, Sanitat Animal, Government of Catalonia (España), Fundación La Marató TV3, Unión Europea. Fondo Europeo de Desarrollo Regional (FEDER/ERDF), Unión Europea. Comisión Europea. H2020, European Research Council, Alzheimers Drug Discovery Foundation, UK Dementia Research Institute, Stichting Alzheimer Onderzoek, Hjärnfonden (Suecia), Swedish government, European Union Joint Programme for Neurodegenerative Disorders, Ministero della Salute (Italia), Alberta Synergies in Alzheimer’s and Related Disorders, Alzheimer Society of Alberta and Northwest Territories, and Centro de Investigación Biomédica en Red - CIBERNED (Enfermedades Neurodegenerativas)
- Subjects
ADAM10 ,metabolism [Microglia] ,ADAM10 Protein ,Mice ,Plasma ,Cerebrospinal fluid ,genetics [Membrane Glycoproteins] ,TREM2 ,genetics [Receptors, Immunologic] ,Medicine ,Cerebrospinal fuid ,Receptors, Immunologic ,Receptor ,Membrane Glycoproteins ,Microglia ,Brain ,metabolism [Receptors, Immunologic] ,medicine.anatomical_structure ,pathology [Prion Diseases] ,metabolism [ADAM10 Protein] ,Malalties per prions ,cerebrospinal fluid [Membrane Glycoproteins] ,metabolism [Prion Diseases] ,metabolism [Alzheimer Disease] ,metabolism [Biomarkers] ,Prion diseases ,blood [Receptors, Immunologic] ,Prion Proteins ,Pathology and Forensic Medicine ,PRNP ,Cellular and Molecular Neuroscience ,metabolism [Prion Proteins] ,Alzheimer Disease ,blood [Membrane Glycoproteins] ,genetics [Prion Diseases] ,mental disorders ,Animals ,Humans ,Malaltia de Creutzfeldt-Jakob ,ddc:610 ,Fatal familial insomnia ,Original Paper ,business.industry ,Multiple sclerosis ,Líquid cefalorraquidi ,medicine.disease ,Creutzfeldt-Jakob disease ,nervous system diseases ,Disease Models, Animal ,cerebrospinal fluid [ADAM10 Protein] ,metabolism [Brain] ,Immunology ,blood [ADAM10 Protein] ,Neurology (clinical) ,business ,metabolism [Membrane Glycoproteins] ,Biomarkers - Abstract
19 Pág. Centro de Investigación en Sanidad Animal (CISA), Triggering receptor expressed on myeloid cells 2 (TREM2) is an innate immune cell surface receptor that regulates microglial function and is involved in the pathophysiology of several neurodegenerative diseases. Its soluble form (sTREM2) results from shedding of the TREM2 ectodomain. The role of TREM2 in prion diseases, a group of rapidly progressive dementias remains to be elucidated. In the present study, we analysed the expression of TREM2 and its main sheddase ADAM10 in the brain of sporadic Creutzfeldt-Jakob disease (sCJD) patients and evaluated the role of CSF and plasma sTREM2 as a potential diagnostic marker of prion disease. Our data indicate that, compared to controls, TREM2 is increased in sCJD patient brains at the mRNA and protein levels in a regional and subtype dependent fashion, and expressed in a subpopulation of microglia. In contrast, ADAM10 is increased at the protein, but not the mRNA level, with a restricted neuronal expression. Elevated CSF sTREM2 is found in sCJD, genetic CJD with mutations E200K and V210I in the prion protein gene (PRNP), and iatrogenic CJD, as compared to healthy controls (HC) (AUC = 0.78-0.90) and neurological controls (AUC = 0.73-0.85), while CSF sTREM2 is unchanged in fatal familial insomnia. sTREM2 in the CSF of cases with Alzheimer's disease, and multiple sclerosis was not significantly altered in our series. CSF sTREM2 concentrations in sCJD are PRNP codon 129 and subtype-related, correlate with CSF 14-3-3 positivity, total-tau and YKL-40, and increase with disease progression. In plasma, sTREM2 is increased in sCJD compared with HC (AUC = 0.80), displaying positive correlations with plasma total-tau, neurofilament light, and YKL-40. We conclude that comparative study of TREM2 in brain and biological fluids of prion diseases reveals TREM2 to be altered in human prion diseases with a potential value in target engagement, patient stratification, and disease monitoring., We thank the HUB-ICO-IDIBELL-Biobank and the CERCA Programme of the Generalitat de Catalunya for institutional support. This study was funded by the Instituto Carlos III (Grants Number CP16/00041 and PI19/00144) to FL. This project was also funded by la Fundació La Marató de TV3 (Grants No. 201821-30, 201821-31and 201821-32 to FL, JCE and EV, respectively) and by the Fondo Europeo de Desarrollo Regional (FEDER) through the Interreg V-A España-Francia-Andorra (POCTEFA 2014–2020) programme (at 65%) to IF. AVP is supported by the Beatriu de Pinós programme (2018-BP-00129) from the Ministry of Business and nowledge of the Government of Catalonia, and cofunded by the EU Horizon 2020 programme under an MSCA grant agreement (801370). HZ is a Wallenberg Scholar supported by grants from the Swedish Research Council (#2018-02532), the European Research Council (#681712), Swedish State Support for Clinical Research (#ALFGBG-720931), the Alzheimer Drug Discovery Foundation (ADDF), USA (#201809-2016862), and the UK Dementia Research Institute at UCL. KB is supported by the Swedish Research Council (#2017-00915), the Swedish State Support for Clinical Research (ADDF), USA (#RDAPB-201809-2016615), the Swedish Alzheimer Foundation (#AF-742881), Hjärnfonden, Sweden (#FO2017-0243), the Swedish state under the agreement between the Swedish government and the County Councils, the ALF-agreement (#ALFGBG-715986), and the European Union Joint Programme for Neurodegenerative Disorders (JPND2019-466-236). AL is supported by the Ministero della Salute, Italy, for the national surveillance of Creutzfeldt-Jakob disease. This research was also supported in part by the Alberta Synergies in Alzheimer’s and Related Disorders (SynAD) programme which is funded by the Alzheimer Society of Alberta and Northwest Territories through their Hope for Tomorrow programme and the University Hospital Foundation. SynAD operates in partnership with the Neuroscience and Mental Health Institute at the University of Alberta. JADR was supported by grants from the Spanish Ministry of Science, Innovation and Universities (MICINN/FEDER) (RTI2018-099773-B-I00), the CERCA Programme, and by the Commission for Universities and Research of the Department of Innovation, Universities, and Enterprise of the Generalitat de Catalunya (SGR2017-648) and CIBERNED (CMED2018-2)
- Published
- 2021
- Full Text
- View/download PDF
21. Age-associated insolubility of parkin in human midbrain is linked to redox balance and sequestration of reactive dopamine metabolites
- Author
-
Qiubo Jiang, Brian O'Nuallain, Clemens R. Scherzer, Eve C. Tsai, Jennifer A. Chan, Peggy Taylor, John Woulfe, An Tran, Ming Jin, Steve M. Callaghan, Gary S. Shaw, Masashi Takanashi, Bojan Shutinoski, Mei Zhang, Jacqueline M. Tokarew, Jasmine M. Khan, Nobutaka Hattori, Daniel N. El-Kodsi, Luigi Zecca, Alexandre Prat, Andrew B. West, Andy C. H. Ng, Xiajun Dong, Juan Li, Travis K. Fehr, Liqun Wang, Nathalie A. Lengacher, Angela P. Nguyen, David S. Park, Doo Soon Im, Julianna J. Tomlinson, Gergely Tóth, Michael G. Schlossmacher, Kathryn R. Barber, Lawrence G. Puente, Arne Holmgren, Stephanie Zandee, and Rajib Sengupta
- Subjects
Adult ,Male ,Aging ,Adolescent ,Dopamine ,Ubiquitin-Protein Ligases ,Substantia nigra ,Oxidative phosphorylation ,Parkinsonism ,medicine.disease_cause ,Parkin ,Neuromelanin ,Anti-oxidant ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,Mice ,Young Adult ,Mesencephalon ,Dopaminergic Cell ,PRKN/PARK2 gene ,medicine ,Animals ,Humans ,Young-onset Parkinson disease ,Child ,Aged ,Aged, 80 and over ,Original Paper ,Redox chemistry ,Chemistry ,Neurodegeneration ,Middle Aged ,medicine.disease ,Cell biology ,nervous system diseases ,Mice, Inbred C57BL ,Child, Preschool ,Nerve Degeneration ,Dopamine metabolism ,Female ,Neurology (clinical) ,Oxidation-Reduction ,Oxidative stress ,medicine.drug - Abstract
The mechanisms by which parkin protects the adult human brain from Parkinson disease remain incompletely understood. We hypothesized that parkin cysteines participate in redox reactions and that these are reflected in its posttranslational modifications. We found that in post mortem human brain, including in the Substantia nigra, parkin is largely insoluble after age 40 years; this transition is linked to its oxidation, such as at residues Cys95 and Cys253. In mice, oxidative stress induces posttranslational modifications of parkin cysteines that lower its solubility in vivo. Similarly, oxidation of recombinant parkin by hydrogen peroxide (H2O2) promotes its insolubility and aggregate formation, and in exchange leads to the reduction of H2O2. This thiol-based redox activity is diminished by parkin point mutants, e.g., p.C431F and p.G328E. In prkn-null mice, H2O2 levels are increased under oxidative stress conditions, such as acutely by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxin exposure or chronically due to a second, genetic hit; H2O2 levels are also significantly increased in parkin-deficient human brain. In dopamine toxicity studies, wild-type parkin, but not disease-linked mutants, protects human dopaminergic cells, in part through lowering H2O2. Parkin also neutralizes reactive, electrophilic dopamine metabolites via adduct formation, which occurs foremost at the primate-specific residue Cys95. Further, wild-type but not p.C95A-mutant parkin augments melanin formation in vitro. By probing sections of adult, human midbrain from control individuals with epitope-mapped, monoclonal antibodies, we found specific and robust parkin reactivity that co-localizes with neuromelanin pigment, frequently within LAMP-3/CD63+ lysosomes. We conclude that oxidative modifications of parkin cysteines are associated with protective outcomes, which include the reduction of H2O2, conjugation of reactive dopamine metabolites, sequestration of radicals within insoluble aggregates, and increased melanin formation. The loss of these complementary redox effects may augment oxidative stress during ageing in dopamine-producing cells of mutant PRKN allele carriers, thereby enhancing the risk of Parkinson’s-linked neurodegeneration.
- Published
- 2021
22. Latent trait modeling of tau neuropathology in progressive supranuclear palsy
- Author
-
Melissa E. Murray, Owen A. Ross, Nilufer Ertekin-Taner, Xue Wang, Joseph S. Reddy, Rosa Rademakers, Ryan J. Uitti, Monica Casey Castanedes, Alexandra I. Soto-Beasley, Daniel J. Serie, Mariet Allen, Matt Baker, Julia E. Crook, Dennis W. Dickson, Gerard D. Schellenberg, Zbigniew K. Wszolek, Naomi Kouri, Neill R. Graff-Radford, and Minerva M. Carrasquillo
- Subjects
0301 basic medicine ,Male ,Single-nucleotide polymorphism ,Genome-wide association study ,tau Proteins ,Neuropathology ,Quantitative trait locus ,Biology ,Pathology and Forensic Medicine ,Progressive supranuclear palsy ,MOBP ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,medicine ,MAPT ,SNP ,Humans ,Aged ,Genetics ,Aged, 80 and over ,Original Paper ,Middle Aged ,medicine.disease ,eye diseases ,030104 developmental biology ,Expression quantitative trait loci ,Female ,Neurology (clinical) ,Tauopathy ,Human medicine ,Supranuclear Palsy, Progressive ,Tau ,Latent traits ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
Progressive supranuclear palsy (PSP) is the second most common neurodegenerative Parkinsonian disorder after Parkinson’s disease, and is characterized as a primary tauopathy. Leveraging the considerable clinical and neuropathologic heterogeneity associated with PSP, we measured tau neuropathology as quantitative traits to perform a genome-wide association study (GWAS) within PSP to identify genes and biological pathways that underlie the PSP disease process. In 882 PSP cases, semi-quantitative scores for phosphorylated tau-immunoreactive coiled bodies (CBs), neurofibrillary tangles (NFTs), tufted astrocytes (TAs), and tau threads were documented from 18 brain regions, and converted to latent trait (LT) variables using the R ltm package. LT analysis utilizes a multivariate regression model that links categorical responses to unobserved covariates allowing for a reduction of dimensionality, generating a single, continuous variable to account for the multiple lesions and brain regions assessed. We first tested for association with PSP LTs and the top PSP GWAS susceptibility loci. Significant SNP/LT associations were identified at rs242557 (MAPT H1c sub-haplotype) with hindbrain CBs and rs1768208 (MOBP) with forebrain tau threads. Digital microscopy was employed to quantify phosphorylated tau burden in midbrain tectum and red nucleus in 795 PSP cases and tau burdens were used as quantitative phenotypes in GWAS. Top associations were identified at rs1768208 with midbrain tectum and red nucleus tau burden. Additionally, we performed a PSP LT GWAS on an initial cohort, a follow-up SNP panel (37 SNPs, P –5) in an extended cohort, and a combined analysis. Top SNP/LT associations were identified at SNPs in or near SPTBN5/EHD4, SEC13/ATP2B2, EPHB1/PPP2R3A, TBC1D8, IFNGR1/OLIG3, ST6GAL1, HK1, CALB1, and SGCZ. Finally, testing for SNP/transcript associations using whole transcriptome and whole genome data identified significant expression quantitative trait loci at rs3088159/SPTBN5/EHD4 and rs154239/GHRL. Modeling tau neuropathology heterogeneity using LTs as quantitative phenotypes in a GWAS may provide substantial insight into biological pathways involved in PSP by affecting regional tau burden.
- Published
- 2021
23. Clear cell meningiomas are defined by a highly distinct DNA methylation profile and mutations in SMARCE1
- Author
-
Matija Snuderl, Zied Abdullaev, Christel Herold-Mende, Ralf Ketter, Uta Schick, Zane Jaunmuktane, David T.W. Jones, Christian Mawrin, Daniel Schrimpf, Leonille Schweizer, Christina Blume, Miriam Ratliff, Arnault Tauziède-Espariat, Pascale Varlet, Arie Perry, Felix Sahm, Damian Stichel, Walter Stummer, Martin Hasselblatt, Jürgen Hench, Stefan M. Pfister, Pieter Wesseling, Guido Reifenberger, Jens Schittenhelm, Helin Dogan, Andreas von Deimling, David W. Ellison, Christian Hartmann, Philipp Sievers, Melike Pekmezci, Wolfgang Wick, David E. Reuss, Stephan Frank, Martin Sill, Sebastian Brandner, Stéphanie Puget, Benno Küsters, Kenneth Aldape, Andreas Unterberg, CCA - Imaging and biomarkers, CCA - Cancer biology and immunology, and Pathology
- Subjects
Male ,Chromosomal Proteins, Non-Histone ,DNA Mutational Analysis ,Population ,Brain tumor ,Biology ,Epigenesis, Genetic ,DNA methylation profile ,Pathology and Forensic Medicine ,Cohort Studies ,Meningioma ,Young Adult ,Cellular and Molecular Neuroscience ,Clear Cell Meningioma ,medicine ,otorhinolaryngologic diseases ,Humans ,Epigenetics ,Child ,education ,neoplasms ,Clear cell ,Original Paper ,education.field_of_study ,Brain Neoplasms ,DNA, Neoplasm ,DNA Methylation ,medicine.disease ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,Immunohistochemistry ,SMARCE1 ,nervous system diseases ,DNA-Binding Proteins ,Treatment Outcome ,Mutation ,DNA methylation ,Disease Progression ,Cancer research ,Female ,Neurology (clinical) ,Neoplasm Recurrence, Local ,Genome-Wide Association Study - Abstract
Clear cell meningioma represents an uncommon variant of meningioma that typically affects children and young adults. Although an enrichment of loss-of-function mutations in the SMARCE1 gene has been reported for this subtype, comprehensive molecular investigations are lacking. Here we describe a molecularly distinct subset of tumors (n = 31), initially identified through genome-wide DNA methylation screening among a cohort of 3093 meningiomas, of which most were diagnosed histologically as clear cell meningioma. This cohort was further supplemented by an additional 11 histologically diagnosed clear cell meningiomas for analysis (n = 42). Targeted DNA sequencing revealed SMARCE1 mutations in 33/34 analyzed samples, accompanied by a nuclear loss of expression determined via immunohistochemistry and a decreased SMARCE1 transcript expression in the tumor cells. Analysis of time to progression or recurrence of patients within the clear cell meningioma group (n = 14) in comparison to those with meningioma WHO grade 2 (n = 220) revealed a similar outcome and support the assignment of WHO grade 2 to these tumors. Our findings indicate the existence of a highly distinct epigenetic signature of clear cell meningiomas, separate from all other variants of meningiomas, with recurrent mutations in the SMARCE1 gene. This suggests that these tumors may arise from a different precursor cell population than the broad spectrum of the other meningioma subtypes.
- Published
- 2021
- Full Text
- View/download PDF
24. Distinct amyloid-β and tau-associated microglia profiles in Alzheimer’s disease
- Author
-
Nieske Brouwer, Thomas Möller, Jan Mulder, Maya E. Woodbury, Emma Gerrits, Peter Paul De Deyn, Knut Biber, Wilfred F. A. den Dunnen, Markus P. Kummer, Yannick Vermeiren, Erik Boddeke, Mirjam Lambourne, Susanne M. Kooistra, Bart J. L. Eggen, Molecular Neuroscience and Ageing Research (MOLAR), Translational Immunology Groningen (TRIGR), and Restoring Organ Function by Means of Regenerative Medicine (REGENERATE)
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Amyloid ,Central nervous system ,Population ,tau Proteins ,Biology ,Pathology and Forensic Medicine ,s disease ,Amyloid-β ,Cellular and Molecular Neuroscience ,Alzheimer Disease ,Single-nucleus RNA sequencing ,mental disorders ,medicine ,Extracellular ,Humans ,Amyloid-β ,education ,Alzheimer’ ,Aged ,Aged, 80 and over ,education.field_of_study ,Original Paper ,Amyloid beta-Peptides ,Microglia ,Brain ,Phenotype ,Pathophysiology ,Nutritional Biology ,medicine.anatomical_structure ,nervous system ,Female ,Neurology (clinical) ,Human medicine ,Tau ,Alzheimer’s disease ,Homeostasis - Abstract
Alzheimer’s disease (AD) is the most prevalent form of dementia and is characterized by abnormal extracellular aggregates of amyloid-β and intraneuronal hyperphosphorylated tau tangles and neuropil threads. Microglia, the tissue-resident macrophages of the central nervous system (CNS), are important for CNS homeostasis and implicated in AD pathology. In amyloid mouse models, a phagocytic/activated microglia phenotype has been identified. How increasing levels of amyloid-β and tau pathology affect human microglia transcriptional profiles is unknown. Here, we performed snRNAseq on 482,472 nuclei from non-demented control brains and AD brains containing only amyloid-β plaques or both amyloid-β plaques and tau pathology. Within the microglia population, distinct expression profiles were identified of which two were AD pathology-associated. The phagocytic/activated AD1-microglia population abundance strongly correlated with tissue amyloid-β load and localized to amyloid-β plaques. The AD2-microglia abundance strongly correlated with tissue phospho-tau load and these microglia were more abundant in samples with overt tau pathology. This full characterization of human disease-associated microglia phenotypes provides new insights in the pathophysiological role of microglia in AD and offers new targets for microglia-state-specific therapeutic strategies. Supplementary Information The online version contains supplementary material available at 10.1007/s00401-021-02263-w.
- Published
- 2021
- Full Text
- View/download PDF
25. Developmental malformations in Huntington disease: neuropathologic evidence of focal neuronal migration defects in a subset of adult brains
- Author
-
Karen Marder, Mark F. Mehler, Marc K. Rosenblum, Richard A. Hickman, Phyllis L. Faust, and J. P. Vonsattel
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,Huntingtin ,Hamartoma ,Neurogenesis ,Autopsy ,Disease ,Development ,Biology ,Nervous System Malformations ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,Germline mutation ,Cell Movement ,medicine ,Humans ,Malformation ,Aged ,Retrospective Studies ,Neurons ,Original Paper ,Brain ,Middle Aged ,medicine.disease ,Huntington Disease ,Heterotopia (medicine) ,Cohort ,Female ,Neurology (clinical) ,Heterotopia ,Trinucleotide repeat expansion - Abstract
Neuropathologic hallmarks of Huntington Disease (HD) include the progressive neurodegeneration of the striatum and the presence of Huntingtin (HTT) aggregates that result from abnormal polyQ expansion of the HTT gene. Whether the pathogenic trinucleotide repeat expansion of the HTT gene causes neurodevelopmental abnormalities has garnered attention in both murine and human studies; however, documentation of discrete malformations in autopsy brains of HD individuals has yet to be described. We retrospectively searched the New York Brain Bank (discovery cohort) and an independent cohort (validation cohort) to determine whether developmental malformations are more frequently detected in HD versus non-HD brains and to document their neuropathologic features. One-hundred and thirty HD and 1600 non-HD whole brains were included in the discovery cohort and 720 HD and 1989 non-HD half brains were assessed in the validation cohort. Cases with developmental malformations were found at 6.4–8.2 times greater frequency in HD than in non-HD brains (discovery cohort: OR 8.68, 95% CI 3.48–21.63, P=4.8 × 10-5; validation cohort: OR 6.50, 95% CI 1.83–23.17, P=0.0050). Periventricular nodular heterotopias (PNH) were the most frequent malformations and contained HTT and p62 aggregates analogous to the cortex, whereas cortical malformations with immature neuronal populations did not harbor such inclusions. HD individuals with malformations had heterozygous HTT CAG expansions between 40 and 52 repeats, were more frequently women, and all were asymmetric and focal, aside from one midline hypothalamic hamartoma. Using two independent brain bank cohorts, this large neuropathologic series demonstrates an increased occurrence of developmental malformations in HD brains. Since pathogenic HTT gene expansion is associated with genomic instability, one possible explanation is that neuronal precursors are more susceptible to somatic mutation of genes involved in cortical migration. Our findings further support emerging evidence that pathogenic trinucleotide repeat expansions of the HTT gene may impact neurodevelopment.
- Published
- 2021
- Full Text
- View/download PDF
26. Neuropathological consensus criteria for the evaluation of Lewy pathology in post-mortem brains: a multi-centre study
- Author
-
Tuomo Polvikoski, Laura Parkkinen, Lea T. Grinberg, Peter T. Nelson, Steve M. Gentleman, Johannes Attems, John Q. Trojanowski, Jon B. Toledo, Edward B. Lee, Ellen Gelpi, Kirsty E. McAleese, Colin Smith, Gabor G. Kovacs, Seth Love, Manuela Neumann, Ian G. McKeith, Lauren Walker, Glenda M. Halliday, Beata Sikorska, Kurt A. Jellinger, Tibor Hortobágyi, Dietmar Rudolf Thal, and Parkinson's UK
- Subjects
Male ,INVOLVEMENT ,Aging ,ALPHA-SYNUCLEIN ,Consensus criteria ,Neurodegenerative ,Audiology ,Alzheimer's Disease ,GUIDELINES ,PARKINSONS-DISEASE ,pathology [Brain] ,80 and over ,Pathology ,2.1 Biological and endogenous factors ,Cluster Analysis ,Aetiology ,Multi centre ,ALZHEIMERS ,Observer Variation ,Brain Mapping ,Parkinson's Disease ,CLINICAL DIAGNOSTIC-CRITERIA ,DEMENTIA ,Brain ,Parkinson Disease ,Neurological ,Female ,Autopsy ,Alzheimer's disease ,diagnosis [Lewy Body Disease] ,Life Sciences & Biomedicine ,medicine.medical_specialty ,Consensus ,Clinical Sciences ,Clinical Neurology ,Lewy body disease ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,Alzheimer Disease ,classification [Lewy Body Disease] ,Lewy pathology ,Acquired Cognitive Impairment ,MANAGEMENT ,medicine ,Humans ,Dementia ,ddc:610 ,Staging system ,Aged ,pathology [Lewy Bodies] ,Original Paper ,Science & Technology ,Neurology & Neurosurgery ,Lewy body ,business.industry ,Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,pathology [Lewy Body Disease] ,Reproducibility of Results ,1103 Clinical Sciences ,COGNITIVE IMPAIRMENT ,medicine.disease ,Diagnostic neuropathology ,Brain Disorders ,Lewy Bodies ,Neurosciences & Neurology ,Neurology (clinical) ,1109 Neurosciences ,business ,SYSTEM - Abstract
Currently, the neuropathological diagnosis of Lewy body disease (LBD) may be stated according to several staging systems, which include the Braak Lewy body stages (Braak), the consensus criteria by McKeith and colleagues (McKeith), the modified McKeith system by Leverenz and colleagues (Leverenz), and the Unified Staging System by Beach and colleagues (Beach). All of these systems use semi-quantitative scoring (4- or 5-tier scales) of Lewy pathology (LP; i.e., Lewy bodies and Lewy neurites) in defined cortical and subcortical areas. While these systems are widely used, some suffer from low inter-rater reliability and/or an inability to unequivocally classify all cases with LP. To address these limitations, we devised a new system, the LP consensus criteria (LPC), which is based on the McKeith system, but applies a dichotomous approach for the scoring of LP (i.e., “absent” vs. “present”) and includes amygdala-predominant and olfactory-only stages. α-Synuclein-stained slides from brainstem, limbic system, neocortex, and olfactory bulb from a total of 34 cases with LP provided by the Newcastle Brain Tissue Resource (NBTR) and the University of Pennsylvania brain bank (UPBB) were scanned and assessed by 16 raters, who provided diagnostic categories for each case according to Braak, McKeith, Leverenz, Beach, and LPC systems. In addition, using LP scores available from neuropathological reports of LP cases from UPBB (n = 202) and NBTR (n = 134), JT (UPBB) and JA (NBTR) assigned categories according to all staging systems to these cases. McKeith, Leverenz, and LPC systems reached good (Krippendorff’s α ≈ 0.6), while both Braak and Beach systems had lower (Krippendorff’s α ≈ 0.4) inter-rater reliability, respectively. Using the LPC system, all cases could be unequivocally classified by the majority of raters, which was also seen for 97.1% when the Beach system was used. However, a considerable proportion of cases could not be classified when using Leverenz (11.8%), McKeith (26.5%), or Braak (29.4%) systems. The category of neocortical LP according to the LPC system was associated with a 5.9 OR (p p = 0.0001) in the 202 UPBB cases. We established that the LPC system has good reproducibility and allows classification of all cases into distinct categories. We expect that it will be reliable and useful in routine diagnostic practice and, therefore, suggest that it should be the standard future approach for the basic post-mortem evaluation of LP.
- Published
- 2021
- Full Text
- View/download PDF
27. Mitochondrial bioenergetic deficits in C9orf72 amyotrophic lateral sclerosis motor neurons cause dysfunctional axonal homeostasis
- Author
-
David Story, Karen Burr, Roderick N. Carter, Arpan R Mehta, Bhuvaneish T. Selvaraj, Siddharthan Chandran, Colin Smith, Jenna M. Gregory, Giles E. Hardingham, Nicholas M. Morton, Don J. Mahad, Owen Dando, Karina McDade, and Jyoti Nanda
- Subjects
Adult ,Male ,Motor neuron ,Mitochondrial DNA ,Induced Pluripotent Stem Cells ,Gene Dosage ,Mitochondrion ,Biology ,Axon ,Pathology and Forensic Medicine ,Electron Transport ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,C9orf72 ,medicine ,Homeostasis ,Humans ,Neurodegeneration ,Amyotrophic lateral sclerosis ,Aged ,030304 developmental biology ,Motor Neurons ,Original Paper ,0303 health sciences ,C9orf72 Protein ,Amyotrophic Lateral Sclerosis ,Energy metabolism ,Middle Aged ,medicine.disease ,Axons ,Mitochondria ,Cell biology ,Posterior Horn Cells ,medicine.anatomical_structure ,Gene Expression Regulation ,nervous system ,Mitochondrial biogenesis ,Female ,Neurology (clinical) ,Frontotemporal dementia ,030217 neurology & neurosurgery - Abstract
Axonal dysfunction is a common phenotype in neurodegenerative disorders, including in amyotrophic lateral sclerosis (ALS), where the key pathological cell-type, the motor neuron (MN), has an axon extending up to a metre long. The maintenance of axonal function is a highly energy-demanding process, raising the question of whether MN cellular energetics is perturbed in ALS, and whether its recovery promotes axonal rescue. To address this, we undertook cellular and molecular interrogation of multiple patient-derived induced pluripotent stem cell lines and patient autopsy samples harbouring the most common ALS causing mutation, C9orf72. Using paired mutant and isogenic expansion-corrected controls, we show that C9orf72 MNs have shorter axons, impaired fast axonal transport of mitochondrial cargo, and altered mitochondrial bioenergetic function. RNAseq revealed reduced gene expression of mitochondrially encoded electron transport chain transcripts, with neuropathological analysis of C9orf72-ALS post-mortem tissue importantly confirming selective dysregulation of the mitochondrially encoded transcripts in ventral horn spinal MNs, but not in corresponding dorsal horn sensory neurons, with findings reflected at the protein level. Mitochondrial DNA copy number was unaltered, both in vitro and in human post-mortem tissue. Genetic manipulation of mitochondrial biogenesis in C9orf72 MNs corrected the bioenergetic deficit and also rescued the axonal length and transport phenotypes. Collectively, our data show that loss of mitochondrial function is a key mediator of axonal dysfunction in C9orf72-ALS, and that boosting MN bioenergetics is sufficient to restore axonal homeostasis, opening new potential therapeutic strategies for ALS that target mitochondrial function.
- Published
- 2021
- Full Text
- View/download PDF
28. Supratentorial ependymoma in childhood: more than just RELA or YAP
- Author
-
Stephan Frank, Andreas Waha, Jörg Faber, Verena Dreschmann, Laszlo Solymosi, Martin Ebinger, Valentina Zschernack, Marie A. Neu, Martin Mynarek, Triantafyllia Brozou, Evelyn Dörner, Bernhard Erdlenbruch, Alexander Claviez, Torsten Pietsch, Felipe Andreiuolo, Stefan Rutkowski, Stefan S. Bielack, Jürgen Hench, Christian Vokuhl, Stephanie T Jünger, Michael C. Frühwald, Annika Stock, and Maria Luisa Garrè
- Subjects
Ependymoma ,Male ,medicine.medical_specialty ,Pathology ,Adolescent ,Brain tumor ,Copy number analysis ,Methylation profiling ,Biology ,Supratentorial ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,medicine ,Humans ,ddc:610 ,Tanycytic ,Child ,Epigenomics ,Adaptor Proteins, Signal Transducing ,Pediatric ,Original Paper ,Chromothripsis ,C11orf95 fusion ,Cytogenetics ,Transcription Factor RelA ,Infant ,Supratentorial Neoplasms ,YAP-Signaling Proteins ,Methylation ,medicine.disease ,Childhood ,Child, Preschool ,Female ,Neurology (clinical) ,Chromosome 22 ,Transcription Factors - Abstract
Two distinct genetically defined entities of ependymoma arising in the supratentorial compartment are characterized by the presence of either a C11orf95-RELA or a YAP-MAMLD1 fusion, respectively. There is growing evidence that supratentorial ependymomas without these genetic features exist. In this study, we report on 18 pediatric non-RELA/non-YAP supratentorial ependymomas that were systematically characterized by means of their histology, immunophenotype, genetics, and epigenomics. Comprehensive molecular analyses included high-resolution copy number analysis, methylation profiling, analysis of fusion transcripts by Nanostring technology, and RNA sequencing. Based upon histological and immunohistochemical features two main patterns were identified—RELA-like (n = 9) and tanycytic ependymomas (n = 6). In the RELA-like group histologically assigned to WHO grade III and resembling RELA-fused ependymomas, tumors lacked nuclear expression of p65-RelA as a surrogate marker for a pathological activation of the NF-κB pathway. Three tumors showed alternative C11orf95 fusions to MAML2 or NCOA1. A methylation-based brain tumor classifier assigned two RELA-like tumors to the methylation class “EP, RELA-fusion”; the others demonstrated no significant similarity score. Of the tanycytic group, 5/6 tumors were assigned a WHO grade II. No gene fusions were detected. Methylation profiling did not show any association with an established methylation class. We additionally identified two astroblastoma-like tumors that both presented with chromothripsis of chromosome 22 but lacked MN1 breaks according to FISH analysis. They revealed novel fusion events involving genes in chromosome 22. One further tumor with polyploid cytogenetics was interpreted as PFB ependymoma by the brain tumor methylation classifier but had no relation to the posterior fossa. Clinical follow-up was available for 16/18 patients. Patients with tanycytic and astroblastoma-like tumors had no relapse, while 2 patients with RELA-like ependymomas died. Our data indicate that in addition to ependymomas discovered so far, at least two more supratentorial ependymoma types (RELA-like and tanycytic) exist.
- Published
- 2021
29. Exosomes induce endolysosomal permeabilization as a gateway by which exosomal tau seeds escape into the cytosol
- Author
-
Jürgen Götz, Juan Carlos Polanco, Adam Briner, Gabriel Rhys Hand, and Chuanzhou Li
- Subjects
Microtubule-associated protein tau ,Endosome ,Tau protein ,Mice, Transgenic ,tau Proteins ,Spreading ,Endosomes ,Protein aggregation ,Exosomes ,Exosome ,Permeability ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,Mice ,Cytosol ,Lysosome ,mental disorders ,medicine ,Autophagy ,Animals ,Humans ,Proteostasis Deficiencies ,Original Paper ,biology ,Chemistry ,HEK 293 cells ,Lentivirus ,rab7 GTP-Binding Proteins ,Microvesicles ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,HEK293 Cells ,Tauopathies ,rab GTP-Binding Proteins ,biology.protein ,Neurology (clinical) ,Lysosomes ,Alzheimer’s disease ,seeding - Abstract
The microtubule-associated protein tau has a critical role in Alzheimer’s disease and other tauopathies. A proposed pathomechanism in the progression of tauopathies is the trans-synaptic spreading of tau seeds, with a role for exosomes which are secretory nanovesicles generated by late endosomes. Our previous work demonstrated that brain-derived exosomes isolated from tau transgenic rTg4510 mice encapsulate tau seeds with the ability to induce tau aggregation in recipient cells. We had also shown that exosomes can hijack the endosomal pathway to spread through interconnected neurons. Here, we reveal how tau seeds contained within internalized exosomes exploit mechanisms of lysosomal degradation to escape the endosome and induce tau aggregation in the cytosol of HEK293T-derived ‘tau biosensor cells’. We found that the majority of the exosome-containing endosomes fused with lysosomes to form endolysosomes. Exosomes induced their permeabilization, irrespective of the presence of tau seeds, or whether the exosomal preparations originated from mouse brains or HEK293T cells. We also found that permeabilization is a conserved mechanism, operating in both non-neuronal tau biosensor cells and primary neurons. However, permeabilization of endolysosomes only occurred in a small fraction of cells, which supports the notion that permeabilization occurs by a thresholded mechanism. Interestingly, tau aggregation was only induced in cells that exhibited permeabilization, presenting this as an escape route of exosomal tau seeds into the cytosol. Overexpression of RAB7, which is required for the formation of endolysosomes, strongly increased tau aggregation. Conversely, inhibition of lysosomal function with alkalinizing agents, or by knocking-down RAB7, decreased tau aggregation. Together, we conclude that the enzymatic activities of lysosomes permeabilize exosomal and endosomal membranes, thereby facilitating access of exosomal tau seeds to cytosolic tau to induce its aggregation. Our data underscore the importance of endosomal membrane integrity in mechanisms of cellular invasion by misfolded proteins that are resistant to lysosomal degradation. Supplementary Information The online version contains supplementary material available at 10.1007/s00401-020-02254-3.
- Published
- 2021
30. The proteome of granulovacuolar degeneration and neurofibrillary tangles in Alzheimer’s disease
- Author
-
Jeroen J.M. Hoozemans, Annemieke J. M. Rozemuller, Conny Leistner, August B. Smit, Fenna Marbus, Ka Wan Li, Regina M Peferoen-Baert, Débora Pita-Illobre, David Hondius, Iryna Paliukhovich, Frank Koopmans, Pathology, Amsterdam Neuroscience - Neuroinfection & -inflammation, Amsterdam Neuroscience - Neurodegeneration, Molecular and Cellular Neurobiology, Amsterdam Neuroscience - Cellular & Molecular Mechanisms, AIMMS, and Center for Neurogenomics and Cognitive Research
- Subjects
0301 basic medicine ,Neurofibrillary tangles ,Proteomics ,Male ,Proteome ,Alzheimer´s disease ,Cellular homeostasis ,Biology ,Pathology and Forensic Medicine ,Tangle ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Microtubule ,Alzheimer Disease ,Humans ,Granulovacuolar degeneration ,Neuropathology ,Laser capture microdissection ,Aged ,Aged, 80 and over ,Inclusion Bodies ,Neurons ,Original Paper ,Middle Aged ,Cell biology ,030104 developmental biology ,Proteostasis ,Nerve Degeneration ,Vacuoles ,Female ,Neurology (clinical) ,Casein kinase 1 ,Tau ,030217 neurology & neurosurgery - Abstract
Granulovacuolar degeneration (GVD) is a common feature in Alzheimer’s disease (AD). The occurrence of GVD is closely associated with that of neurofibrillary tangles (NFTs) and GVD is even considered to be a pre-NFT stage in the disease process of AD. Currently, the composition of GVD bodies, the mechanisms associated with GVD and how GVD exactly relates to NFTs is not well understood. By combining immunohistochemistry (IHC) and laser microdissection (LMD) we isolated neurons with GVD and those bearing tangles separately from human post-mortem AD hippocampus (n = 12) using their typical markers casein kinase (CK)1δ and phosphorylated tau (AT8). Control neurons were isolated from cognitively healthy cases (n = 12). 3000 neurons per sample were used for proteome analysis by label free LC–MS/MS. In total 2596 proteins were quantified across samples and a significant change in abundance of 115 proteins in GVD and 197 in tangle bearing neurons was observed compared to control neurons. With IHC the presence of PPIA, TOMM34, HSP70, CHMP1A, TPPP and VXN was confirmed in GVD containing neurons. We found multiple proteins localizing specifically to the GVD bodies, with VXN and TOMM34 being the most prominent new protein markers for GVD bodies. In general, protein groups related to protein folding, proteasomal function, the endolysosomal pathway, microtubule and cytoskeletal related function, RNA processing and glycolysis were found to be changed in GVD neurons. In addition to these protein groups, tangle bearing neurons show a decrease in ribosomal proteins, as well as in various proteins related to protein folding. This study, for the first time, provides a comprehensive human based quantitative assessment of protein abundances in GVD and tangle bearing neurons. In line with previous functional data showing that tau pathology induces GVD, our data support the model that GVD is part of a pre-NFT stage representing a phase in which proteostasis and cellular homeostasis is disrupted. Elucidating the molecular mechanisms and cellular processes affected in GVD and its relation to the presence of tau pathology is highly relevant for the identification of new drug targets for therapy. Supplementary Information The online version contains supplementary material available at 10.1007/s00401-020-02261-4.
- Published
- 2021
- Full Text
- View/download PDF
31. In vitro amplification of pathogenic tau conserves disease-specific bioactive characteristics
- Author
-
Jennifer D. McBride, Bin Zhang, Kurt R. Brunden, Lakshmi Changolkar, Virginia M.-Y. Lee, Jeffrey J. Nirschl, Kevt’her Hoxha, John Q. Trojanowski, Hong Xu, Garrett S. Gibbons, Gerard D. Schellenberg, Mia O’Reilly, Soo-Jung Kim, Dawn M. Riddle, and Anna Stieber
- Subjects
0301 basic medicine ,Disease specific ,in vitro seeding ,Tau protein ,Primary Cell Culture ,tau Proteins ,Fibril ,Pathology and Forensic Medicine ,law.invention ,Progressive supranuclear palsy ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,0302 clinical medicine ,law ,Alzheimer Disease ,mental disorders ,medicine ,tau strains ,Corticobasal degeneration ,Animals ,Cells, Cultured ,Conserved Sequence ,Original Paper ,biology ,tauopathy ,tau spreading ,Gene Amplification ,food and beverages ,Brain ,Neurodegenerative Diseases ,Neurofibrillary Tangles ,medicine.disease ,Immunohistochemistry ,In vitro ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Tauopathies ,Recombinant DNA ,biology.protein ,Neurology (clinical) ,Tauopathy ,Supranuclear Palsy, Progressive ,030217 neurology & neurosurgery - Abstract
The microtubule-associated protein tau (tau) forms hyperphosphorylated aggregates in the brains of tauopathy patients that can be pathologically and biochemically defined as distinct tau strains. Recent studies show that these tau strains exhibit strain-specific biological activities, also referred to as pathogenicities, in the tau spreading models. Currently, the specific pathogenicity of human-derived tau strains cannot be fully recapitulated by synthetic tau preformed fibrils (pffs), which are generated from recombinant tau protein. Reproducing disease-relevant tau pathology in cell and animal models necessitates the use of human brain-derived tau seeds. However, the availability of human-derived tau is extremely limited. Generation of tau variants that can mimic the pathogenicity of human-derived tau seeds would significantly extend the scale of experimental design within the field of tauopathy research. Previous studies have demonstrated that in vitro seeding reactions can amplify the beta-sheet structure of tau protein from a minute quantity of human-derived tau. However, whether the strain-specific pathogenicities of the original, human-derived tau seeds are conserved in the amplified tau strains has yet to be experimentally validated. Here, we used biochemically enriched brain-derived tau seeds from Alzheimer’s disease (AD), corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP) patient brains with a modified seeding protocol to template the recruitment of recombinant 2N4R (T40) tau in vitro. We quantitatively interrogated efficacy of the amplification reactions and the pathogenic fidelity of the amplified material to the original tau seeds using recently developed sporadic tau spreading models. Our data suggest that different tau strains can be faithfully amplified in vitro from tau isolated from different tauopathy brains and that the amplified tau variants retain their strain-dependent pathogenic characteristics. Supplementary Information The online version contains supplementary material available at 10.1007/s00401-020-02253-4.
- Published
- 2021
32. Atypical teratoid/rhabdoid tumors (ATRTs) with SMARCA4 mutation are molecularly distinct from SMARCB1-deficient cases
- Author
-
Annie Huang, Julien Masliah-Planchon, Ben Ho, Anne Bendel, Santhosh A. Upadhyaya, Sepehr Safaei, David Creytens, Marcel Kool, Uwe Kordes, Ulrich Schüller, Daniela Indenbirken, Michael C. Frühwald, Piyush Joshi, William D. Foulkes, Mamy Andrianteranagna, Michael Bockmayr, Pascal Johann, Martin Hasselblatt, Dörthe Holdhof, Jonathan W. Bush, Michael Spohn, and Franck Bourdeaut
- Subjects
HYPOMETHYLATION ,CHILDREN ,VARIANTS ,medicine.disease_cause ,ATRT ,Central Nervous System Neoplasms ,Transcriptome ,BRG1 ,SMARCA4 ,Medicine and Health Sciences ,Age of Onset ,SMARCB1 ,Child ,Mutation ,DNA methylation ,Teratoma ,Nuclear Proteins ,RNA sequencing ,RHABDOID TUMORS ,SMARCB1 Protein ,Middle Aged ,Child, Preschool ,Adult ,Adolescent ,Clinical Neurology ,HYPERCALCEMIC TYPE ,Biology ,Pathology and Forensic Medicine ,Young Adult ,Cellular and Molecular Neuroscience ,Germline mutation ,Rhabdoid ,SMALL-CELL-CARCINOMA ,medicine ,Humans ,SUBGROUPS ,ddc:610 ,Gene ,Rhabdoid Tumor ,Medulloblastoma ,Original Paper ,COMPLEX ,Gene Expression Profiling ,DNA Helicases ,Computational Biology ,Biology and Life Sciences ,medicine.disease ,Survival Analysis ,OVARY ,Cancer research ,Neurology (clinical) ,Transcription Factors - Abstract
Atypical teratoid/rhabdoid tumors (ATRTs) are very aggressive childhood malignancies of the central nervous system. The underlying genetic cause are inactivating bi-allelic mutations in SMARCB1 or (rarely) in SMARCA4. ATRT-SMARCA4 have been associated with a higher frequency of germline mutations, younger age, and an inferior prognosis in comparison to SMARCB1 mutated cases. Based on their DNA methylation profiles and transcriptomics, SMARCB1 mutated ATRTs have been divided into three distinct molecular subgroups: ATRT-TYR, ATRT-SHH, and ATRT-MYC. These subgroups differ in terms of age at diagnosis, tumor location, type of SMARCB1 alterations, and overall survival. ATRT-SMARCA4 are, however, less well understood, and it remains unknown, whether they belong to one of the described ATRT subgroups. Here, we examined 14 ATRT-SMARCA4 by global DNA methylation analyses. We show that they form a separate group segregating from SMARCB1 mutated ATRTs and from other SMARCA4-deficient tumors like small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) or SMARCA4 mutated extra-cranial malignant rhabdoid tumors. In contrast, medulloblastoma (MB) samples with heterozygous SMARCA4 mutations do not group separately, but with established MB subgroups. RNA sequencing of ATRT-SMARCA4 confirmed the clustering results based on DNA methylation profiling and displayed an absence of typical signature genes upregulated in SMARCB1 deleted ATRT. In summary, our results suggest that, in line with previous clinical observations, ATRT-SMARCA4 should be regarded as a distinct molecular subgroup.
- Published
- 2020
- Full Text
- View/download PDF
33. Lewy pathology of the esophagus correlates with the progression of Lewy body disease: a Japanese cohort study of autopsy cases
- Author
-
Yasuhiro Sakashita, Masako Ikemura, Ito Kawakami, Renpei Sengoku, Mikihiro Yamazaki, Tomio Arai, Shinya Tanaka, Yuko Saito, Shinji Ito, Atsuko Motoda, Tomoyasu Matsubara, Zen-ichi Tanei, and Shigeo Murayama
- Subjects
Adult ,Central Nervous System ,Lewy Body Disease ,Male ,Pathology ,medicine.medical_specialty ,Parkinson's disease ,Myenteric Plexus ,Autopsy ,Disease ,Pathology and Forensic Medicine ,Cohort Studies ,Cellular and Molecular Neuroscience ,Esophagus ,Japan ,Peripheral Nervous System ,medicine ,Prevalence ,Humans ,Pure autonomic failure ,Aged ,Biological Specimen Banks ,α-Synuclein ,Aged, 80 and over ,Plexus ,Original Paper ,Dementia with Lewy bodies ,business.industry ,Age Factors ,Middle Aged ,medicine.disease ,Immunohistochemistry ,medicine.anatomical_structure ,Peripheral nervous system ,Parkinson’s disease ,alpha-Synuclein ,Female ,Lewy Bodies ,Neurology (clinical) ,Enteric nervous system ,business - Abstract
Lewy body disease (LBD) is a spectrum of progressive neurodegenerative disorders characterized by the wide distribution of Lewy bodies and neurites in the central and peripheral nervous system (CNS, PNS). Clinical diagnoses include Parkinson’s disease (PD), dementia with Lewy bodies, or pure autonomic failure. All types of LBD are accompanied by non-motor symptoms (NMSs) including gastrointestinal dysfunctions such as constipation. Its relationship to Lewy body-related α-synucleinopathy (Lewy pathology) of the enteric nervous system (ENS) is attracting attention because it can precede the motor symptoms. To clarify the role of ENS Lewy pathology in disease progression, we performed a clinicopathological study using the Brain Bank for Aging Research in Japan. Five-hundred and eighteen cases were enrolled in the study. Lewy pathology of the CNS and PNS, including the lower esophagus as a representative of the ENS, was examined via autopsy findings. Results showed that one-third of older people (178 cases, 34%) exhibited Lewy pathology, of which 78 cases (43.8%) exhibited the pathology in the esophagus. In the esophageal wall, Auerbach’s plexus (41.6%) was most susceptible to the pathology, followed by the adventitia (33.1%) and Meissner’s plexus (14.6%). Lewy pathology of the esophagus was significantly associated with autonomic failures such as constipation (p
- Published
- 2020
34. Archeological neuroimmunology: resurrection of a pathogenic immune response from a historical case sheds light on human autoimmune encephalomyelitis and multiple sclerosis
- Author
-
Eduardo Beltrán, Manuela Paunovic, David Gebert, Emine Cesur, Markus Jeitler, Romana Höftberger, Joachim Malotka, Simone Mader, Naoto Kawakami, Edgar Meinl, Monika Bradl, Klaus Dornmair, and Hans Lassmann
- Subjects
Original Paper ,Cellular and Molecular Neuroscience ,Human autoimmune encephalitis ,Next generation sequencing ,Myelin oligodendrocytes glycoprotein ,Neurology (clinical) ,Multiple sclerosis (MS) ,Pathology and Forensic Medicine - Abstract
Aim of our study was to identify the target auto-antigen in the central nervous system recognized by the immune system of a unique patient, who died more than 60 years ago from a disease with pathological changes closely resembling multiple sclerosis (MS), following a misguided immunization with lyophilized calf brain tissue. Total mRNA was isolated from formaldehyde fixed and paraffin embedded archival brain tissue containing chronic active inflammatory demyelinating lesions with inflammatory infiltrates rich in B-lymphocytes and plasma cells. Analysis of the transcriptome by next generation sequencing and reconstruction of the dominant antibody by bioinformatic tools revealed the presence of one strongly expanded B-cell clone, producing an autoantibody against a conformational epitope of myelin oligodendrocytes glycoprotein (MOG), similar to that recognized by the well characterized monoclonal anti-MOG antibody 8-18C5. The reconstructed antibody induced demyelination after systemic or intrathecal injection into animals with T-cell mediated encephalomyelitis. Our study suggests that immunization with bovine brain tissue in humans may—in a small subset of patients—induce a disease with an intermediate clinical and pathological presentation between MS and MOG-antibody associated inflammatory demyelinating disease (MOGAD). Electronic supplementary material The online version of this article (10.1007/s00401-020-02239-2) contains supplementary material, which is available to authorized users.
- Published
- 2020
- Full Text
- View/download PDF
35. Epigenomic, genomic, and transcriptomic landscape of schwannomatosis
- Author
-
Kimberly Laskie Ostrow, Laura Papi, Irene Paganini, Kenneth Aldape, Vikas Patil, David A. Largaespada, Jaishri O. Blakeley, Olivia Singh, Prisni Rath, Yogi Sundaravadanam, Yasin Mamatjan, Anat Stemmer-Rachamimov, Roberta Sestini, Suganth Suppiah, Farshad Nassiri, Sheila Mansouri, Francesca Gensini, Trevor J. Pugh, Jeff C. Liu, Sameer Agnihotri, Marcela Maria Ferrer, Shirin Karimi, Scott R. Plotkin, and Gelareh Zadeh
- Subjects
0301 basic medicine ,Skin Neoplasms ,Neurofibromatoses ,Peripheral nerve sheath tumors ,Pain ,Genomics ,Biology ,Nerve Sheath Neoplasms ,Pathology and Forensic Medicine ,Epigenesis, Genetic ,Loss of heterozygosity ,Cohort Studies ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Germline mutation ,medicine ,Humans ,Genetic Predisposition to Disease ,Epigenetics ,Schwannomatosis ,Gene ,Germ-Line Mutation ,Epigenomics ,Genetics ,Neurofibromin 2 ,Original Paper ,Correction ,High-Temperature Requirement A Serine Peptidase 1 ,DNA Methylation ,medicine.disease ,MAPK ,Adaptor Proteins, Vesicular Transport ,030104 developmental biology ,DNA methylation ,Neurology (clinical) ,Gene Fusion ,Mitogen-Activated Protein Kinases ,LZTR1 ,Transcriptome ,030217 neurology & neurosurgery ,Neurilemmoma ,Transcription Factors - Abstract
Schwannomatosis (SWNTS) is a genetic cancer predisposition syndrome that manifests as multiple and often painful neuronal tumors called schwannomas (SWNs). While germline mutations in SMARCB1 or LZTR1, plus somatic mutations in NF2 and loss of heterozygosity in chromosome 22q have been identified in a subset of patients, little is known about the epigenomic and genomic alterations that drive SWNTS-related SWNs (SWNTS-SWNs) in a majority of the cases. We performed multiplatform genomic analysis and established the molecular signature of SWNTS-SWNs. We show that SWNTS-SWNs harbor distinct genomic features relative to the histologically identical non-syndromic sporadic SWNs (NS-SWNS). We demonstrate the existence of four distinct DNA methylation subgroups of SWNTS-SWNs that are associated with specific transcriptional programs and tumor location. We show several novel recurrent non-22q deletions and structural rearrangements. We detected the SH3PXD2A-HTRA1 gene fusion in SWNTS-SWNs, with predominance in LZTR1-mutant tumors. In addition, we identified specific genetic, epigenetic, and actionable transcriptional programs associated with painful SWNTS-SWNs including PIGF, VEGF, MEK, and MTOR pathways, which may be harnessed for management of this syndrome.
- Published
- 2020
36. Accumulation of amyloid precursor protein C-terminal fragments triggers mitochondrial structure, function, and mitophagy defects in Alzheimer’s disease models and human brains
- Author
-
Julie Cazareth, Raphaëlle Pardossi-Piquard, Arnaud Mary, Inger Lauritzen, Renaud Bussiere, Alexandre Bourgeois, Véronique Paquis-Flucklinger, Sandra Lacas-Gervais, Paula Fernanda Kinoshita, Konstantina Fragaki, Frédéric Checler, Loan Vaillant-Beuchot, Rosanna Mary, Mounia Chami, Valérie Buée-Scherrer, Sophie Pagnotta, Cécile Martin, Fanny Eysert, Charlotte Bauer, Céline Badot, Luc Buée, Institut de pharmacologie moléculaire et cellulaire (IPMC), Centre National de la Recherche Scientifique (CNRS)-Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA), and Excellence Laboratory LabEx DISTALZ
- Subjects
DYNAMICS ,[SDV]Life Sciences [q-bio] ,Mitochondrion ,Parkin ,Amyloid beta-Protein Precursor ,Mice ,0302 clinical medicine ,Mitophagy ,Pathology ,Amyloid precursor protein ,Aspartic Acid Endopeptidases ,ER MEMBRANES ,IN-VIVO ,C99 ,Aged, 80 and over ,Membrane Potential, Mitochondrial ,0303 health sciences ,TRANSGENIC MODEL ,biology ,Chemistry ,Brain ,MOUSE MODEL ,Alzheimer's disease ,Cell biology ,Mitochondria ,Female ,Autopsy ,Life Sciences & Biomedicine ,Alzheimer’s disease ,Genetically modified mouse ,LYSOSOMAL DYSFUNCTION ,Amyloid ,Amyloid beta ,Clinical Neurology ,PINK1 ,INTRANEURONAL ACCUMULATION ,Pathology and Forensic Medicine ,Cell Line ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Alzheimer Disease ,Animals ,Humans ,A-BETA ,APP-CTFs ,030304 developmental biology ,Aged ,Original Paper ,Science & Technology ,Neurology & Neurosurgery ,Neurosciences ,1103 Clinical Sciences ,Peptide Fragments ,C83 ,biology.protein ,BETA-CTF FRAGMENT ,Neurosciences & Neurology ,Neurology (clinical) ,Amyloid Precursor Protein Secretases ,APP ,1109 Neurosciences ,Reactive Oxygen Species ,030217 neurology & neurosurgery - Abstract
Several lines of recent evidence indicate that the amyloid precursor protein-derived C-terminal fragments (APP-CTFs) could correspond to an etiological trigger of Alzheimer’s disease (AD) pathology. Altered mitochondrial homeostasis is considered an early event in AD development. However, the specific contribution of APP-CTFs to mitochondrial structure, function, and mitophagy defects remains to be established. Here, we demonstrate in neuroblastoma SH-SY5Y cells expressing either APP Swedish mutations, or the β-secretase-derived APP-CTF fragment (C99) combined with β- and γ-secretase inhibition, that APP-CTFs accumulation independently of Aβ triggers excessive mitochondrial morphology alteration (i.e., size alteration and cristae disorganization) associated with enhanced mitochondrial reactive oxygen species production. APP-CTFs accumulation also elicit basal mitophagy failure illustrated by enhanced conversion of LC3, accumulation of LC3-I and/or LC3-II, non-degradation of SQSTM1/p62, inconsistent Parkin and PINK1 recruitment to mitochondria, enhanced levels of membrane and matrix mitochondrial proteins, and deficient fusion of mitochondria with lysosomes. We confirm the contribution of APP-CTFs accumulation to morphological mitochondria alteration and impaired basal mitophagy in vivo in young 3xTgAD transgenic mice treated with γ-secretase inhibitor as well as in adeno-associated-virus-C99 injected mice. Comparison of aged 2xTgAD and 3xTgAD mice indicates that, besides APP-CTFs, an additional contribution of Aβ to late-stage mitophagy activation occurs. Importantly, we report on mitochondrial accumulation of APP-CTFs in human post-mortem sporadic AD brains correlating with mitophagy failure molecular signature. Since defective mitochondria homeostasis plays a pivotal role in AD pathogenesis, targeting mitochondrial dysfunctions and/or mitophagy by counteracting early APP-CTFs accumulation may represent relevant therapeutic interventions in AD. Electronic supplementary material The online version of this article (10.1007/s00401-020-02234-7) contains supplementary material, which is available to authorized users.
- Published
- 2020
- Full Text
- View/download PDF
37. Molecular characterization of CNS paragangliomas identifies cauda equina paragangliomas as a distinct tumor entity
- Author
-
Alexander Younsi, David E. Reuss, Johannes Kahn, David Capper, Anne Schöler, Georgios Ntoulias, Daniel Teichmann, Hee-Yeong Kim, Dag Moskopp, Diaa Al Safatli, Abigail K. Suwala, Jan Walter, Sven-Axel May, Peter Vajkoczy, David Kaul, Bettina Knie, Frank L. Heppner, Wolfgang Hartmann, Andreas Unterberg, Martin Hasselblatt, Leonille Schweizer, Damian Stichel, Felix Thierfelder, Christian Hartmann, Andreas Jödicke, Arend Koch, Martin Misch, Ruben Jödicke, Annika K. Wefers, Patrick Soschinski, Andreas von Deimling, Michael Bockmayr, Lars Wessels, and Christian Thomas
- Subjects
Male ,musculoskeletal diseases ,Pathology ,medicine.medical_specialty ,genetics [Central Nervous System Neoplasms] ,genetics [Paraganglioma] ,SDHB ,diagnosis [Neoplasm Recurrence, Local] ,Neuroendocrine tumors ,Biology ,Cauda equina ,Pathology and Forensic Medicine ,Central Nervous System Neoplasms ,Diagnosis, Differential ,Paraganglioma ,Head and neck ,Cellular and Molecular Neuroscience ,Cytokeratin ,genetics [Neuroendocrine Tumors] ,Germline mutation ,pathology [Cauda Equina] ,pathology [Neuroendocrine Tumors] ,GATA3 ,medicine ,Humans ,ddc:610 ,pathology [Neoplasm Recurrence, Local] ,diagnosis [Neuroendocrine Tumors] ,Germ-Line Mutation ,Exome sequencing ,genetics [Germ-Line Mutation] ,Original Paper ,DNA methylation ,Adrenal gland ,pathology [Central Nervous System Neoplasms] ,Prognosis ,medicine.disease ,genetics [Neoplasm Recurrence, Local] ,pathology [Paraganglioma] ,Neuroendocrine Tumors ,medicine.anatomical_structure ,Female ,Neurology (clinical) ,Neoplasm Recurrence, Local ,600 Technik, Medizin, angewandte Wissenschaften::610 Medizin und Gesundheit::610 Medizin und Gesundheit - Abstract
Paragangliomas/pheochromocytomas are rare neuroendocrine tumors that arise from the adrenal gland or ganglia at various sites throughout the body. They display a remarkable diversity of driver alterations and are associated with germline mutations in up to 40% of the cases. Comprehensive molecular profiling of abdomino-thoracic paragangliomas revealed four molecularly defined and clinically relevant subtypes. Paragangliomas of the cauda equina region are considered to belong to one of the defined molecular subtypes, but a systematic molecular analysis has not yet been performed. In this study, we analyzed genome-wide DNA methylation profiles of 57 cauda equina paragangliomas and show that these tumors are epigenetically distinct from non-spinal paragangliomas and other tumors. In contrast to paragangliomas of other sites, chromosomal imbalances are widely lacking in cauda equina paragangliomas. Furthermore, RNA and DNA exome sequencing revealed that frequent genetic alterations found in non-spinal paragangliomas—including the prognostically relevant SDH mutations—are absent in cauda equina paragangliomas. Histologically, cauda equina paragangliomas show frequently gangliocytic differentiation and strong immunoreactivity to pan-cytokeratin and cytokeratin 18, which is not common in paragangliomas of other sites. None of our cases had a familial paraganglioma syndrome. Tumors rarely recurred (9%) or presented with multiple lesions within the spinal compartment (7%), but did not metastasize outside the CNS. In summary, we show that cauda equina paragangliomas represent a distinct, sporadic tumor entity defined by a unique clinical and morpho-molecular profile. Electronic supplementary material The online version of this article (10.1007/s00401-020-02218-7) contains supplementary material, which is available to authorized users.
- Published
- 2020
- Full Text
- View/download PDF
38. Altered DNA methylation profiles in blood from patients with sporadic Creutzfeldt–Jakob disease
- Author
-
John Collinge, Rachel Raybould, Adam Smith, Tony Belicard, Luke C. Dabin, Jonathan M. Schott, Emmanuelle Viré, Rebecca G. Smith, Fernando Guntoro, Simon Mead, Tracy Campbell, Peter Sarkies, and Katie Lunnon
- Subjects
Adult ,Male ,Prion disease ,Disease ,Creutzfeldt-Jakob Syndrome ,Prion Diseases ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,AIM2 ,Alzheimer Disease ,medicine ,Humans ,Genetic Predisposition to Disease ,Epigenetics ,Neurodegeneration ,Disease duration ,Aged ,Aged, 80 and over ,Original Paper ,DNA methylation ,business.industry ,Brain ,Methylation ,Middle Aged ,medicine.disease ,Blood ,sCJD ,Case-Control Studies ,Immunology ,Shaker Superfamily of Potassium Channels ,Pyrosequencing ,Female ,Neurology (clinical) ,FKBP5 ,business - Abstract
Prion diseases are fatal and transmissible neurodegenerative disorders caused by the misfolding and aggregation of prion protein. Although recent studies have implicated epigenetic variation in common neurodegenerative disorders, no study has yet explored their role in human prion diseases. Here we profiled genome-wide blood DNA methylation in the most common human prion disease, sporadic Creutzfeldt–Jakob disease (sCJD). Our case–control study (n = 219), when accounting for differences in cell type composition between individuals, identified 38 probes at genome-wide significance (p –7). Nine of these sites were taken forward in a replication study, performed in an independent case–control (n = 186) cohort using pyrosequencing. Sites in or close to FKBP5, AIM2 (2 probes), UHRF1, KCNAB2 successfully replicated. The blood-based DNA methylation signal was tissue- and disease-specific, in that the replicated probe signals were unchanged in case–control studies using sCJD frontal-cortex (n = 84), blood samples from patients with Alzheimer’s disease, and from inherited and acquired prion diseases. Machine learning algorithms using blood DNA methylation array profiles accurately distinguished sCJD patients and controls. Finally, we identified sites whose methylation levels associated with prolonged survival in sCJD patients. Altogether, this study has identified a peripheral DNA methylation signature of sCJD with a variety of potential biomarker applications.
- Published
- 2020
- Full Text
- View/download PDF
39. C11orf95-RELA reprograms 3D epigenome in supratentorial ependymoma
- Author
-
Nathaniel Jillette, Xiao-Nan Li, Albert W. Cheng, Jacqueline Jufen Zhu, and Ching C. Lau
- Subjects
Ependymoma ,Oncogene Proteins, Fusion ,Biology ,medicine.disease_cause ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,medicine ,Humans ,Transcription factor ,C11orf95-RELA ,Epigenomics ,Original Paper ,Brain Neoplasms ,Transcription Factor RelA ,Proteins ,Supratentorial Neoplasms ,Correction ,Epigenome ,medicine.disease ,Supratentorial ependymoma ,Fusion protein ,3D genome ,Chromatin ,Cancer research ,Neurology (clinical) ,Carcinogenesis ,Reprogramming ,Signal Transduction - Abstract
Supratentorial ependymoma (ST-EPN) is a type of malignant brain tumor mainly seen in children. Since 2014, it has been known that an intrachromosomal fusion C11orf95-RELA is an oncogenic driver in ST-EPN [Parker et al. Nature 506:451–455 (2014); Pietsch et al. Acta Neuropathol 127:609–611 (2014)] but the molecular mechanisms of oncogenesis are unclear. Here we show that the C11orf95 component of the fusion protein dictates DNA binding activity while the RELA component is required for driving the expression of ependymoma-associated genes. Epigenomic characterizations using ChIP-seq and HiChIP approaches reveal that C11orf95-RELA modulates chromatin states and mediates chromatin interactions, leading to transcriptional reprogramming in ependymoma cells. Our findings provide important characterization of the molecular underpinning of C11orf95-RELA fusion and shed light on potential therapeutic targets for C11orf95-RELA subtype ependymoma. Electronic supplementary material The online version of this article (10.1007/s00401-020-02225-8) contains supplementary material, which is available to authorized users.
- Published
- 2020
40. Antibody against TDP-43 phosphorylated at serine 375 suggests conformational differences of TDP-43 aggregates among FTLD–TDP subtypes
- Author
-
Francesca Paron, Petra Frick, Emanuele Buratti, Manuela Neumann, Jonas Kosten, and Ian R. A. Mackenzie
- Subjects
Male ,metabolism [Inclusion Bodies] ,Phosphorylation-specific antibody ,TDP-43 ,Protein Conformation ,chemistry [Inclusion Bodies] ,medicine.disease_cause ,Serine ,pathology [Inclusion Bodies] ,0302 clinical medicine ,C9orf72 ,Conformation-specific ,Phosphorylation ,Inclusion Bodies ,Aged, 80 and over ,0303 health sciences ,Mutation ,Frontotemporal lobar degeneration ,Middle Aged ,Phenotype ,DNA-Binding Proteins ,metabolism [Frontotemporal Dementia] ,Immunohistochemistry ,Female ,metabolism [DNA-Binding Proteins] ,Frontotemporal dementia ,Biology ,Protein Aggregation, Pathological ,Pathology and Forensic Medicine ,Protein Aggregates ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,metabolism [Protein Aggregation, Pathological] ,mental disorders ,medicine ,Humans ,ddc:610 ,pathology [Amyotrophic Lateral Sclerosis] ,Aged ,030304 developmental biology ,Antiserum ,Original Paper ,metabolism [Amyotrophic Lateral Sclerosis] ,Amyotrophic Lateral Sclerosis ,Correction ,nutritional and metabolic diseases ,chemistry [DNA-Binding Proteins] ,medicine.disease ,Molecular biology ,nervous system diseases ,TDP-43 strains ,pathology [Frontotemporal Dementia] ,Neurology (clinical) ,ALS ,030217 neurology & neurosurgery - Abstract
Aggregation of hyperphosphorylated TDP-43 is the hallmark pathological feature of the most common molecular form of frontotemporal lobar degeneration (FTLD–TDP) and in the vast majority of cases with amyotrophic lateral sclerosis (ALS–TDP). However, most of the specific phosphorylation sites remain to be determined, and their relevance regarding pathogenicity and clinical and pathological phenotypic diversity in FTLD–TDP and ALS–TDP remains to be identified. Here, we generated a novel antibody raised against TDP-43 phosphorylated at serine 375 (pTDP-43S375) located in the low-complexity domain, and used it to investigate the presence of S375 phosphorylation in a series (n = 44) of FTLD–TDP and ALS–TDP cases. Immunoblot analysis demonstrated phosphorylation of S375 to be a consistent feature of pathological TDP-43 species, including full-length and C-terminal fragments, in all FTLD–TDP subtypes examined (A–C) and in ALS–TDP. Of particular interest, however, detailed immunohistochemical analysis showed striking differences in the immunoreactivity profile of inclusions with the pTDP-43S375 antiserum among pathological subtypes. TDP-43 pathology of ALS–TDP, FTLD–TDP type B (including cases with the C9orf72 mutation), and FTLD–TDP type C all showed strong pTDP-43S375 immunoreactivity that was similar in amount and morphology to that seen with an antibody against TDP-43 phosphorylated at S409/410 used as the gold standard. In stark contrast, TDP-43 pathology in sporadic and genetic forms of FTLD–TDP type A (including cases with GRN and C9orf72 mutations) was found to be almost completely negative by pTDP-43S375 immunohistochemistry. These data suggest a subtype-specific, conformation-dependent binding of pTDP-43S375 antiserum to TDP-43 aggregates, consistent with the idea of distinct structural TDP-43 conformers (i.e., TDP-43 strains) as the molecular basis for the phenotypic diversity in TDP-43 proteinopathies.
- Published
- 2020
- Full Text
- View/download PDF
41. TREM2 activation on microglia promotes myelin debris clearance and remyelination in a model of multiple sclerosis
- Author
-
Danilo Licastro, Melissa Manis, Bryan Bollman, Fabia Filipello, Michael E. Buckland, Laura Piccio, Carlos Cruchaga, David M. Holtzman, Kathie A. Mihindukulasuriya, Adiljan Ibrahim, Bruno A. Benitez, Francesca Cignarella, Robert Mikesell, Alberto Locca, Claudia Cantoni, Ilaria Tassi, Arnon Rosenthal, Oscar Harari, Li Deng, and Tina Schwabe
- Subjects
Adult ,Male ,Multiple Sclerosis ,CNS demyelination ,Central nervous system ,Biology ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,Myelin ,Mice ,Phagocytosis ,medicine ,Animals ,Humans ,Remyelination ,Receptors, Immunologic ,Myelin Sheath ,Aged ,Mice, Knockout ,Original Paper ,Membrane Glycoproteins ,Microglia ,TREM2 ,Multiple sclerosis ,Middle Aged ,medicine.disease ,Oligodendrocyte ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,nervous system ,Female ,Neurology (clinical) - Abstract
Multiple sclerosis (MS) is an inflammatory, demyelinating, and neurodegenerative disease of the central nervous system (CNS) triggered by autoimmune mechanisms. Microglia are critical for the clearance of myelin debris in areas of demyelination, a key step to allow remyelination. TREM2 is expressed by microglia and promotes microglial survival, proliferation, and phagocytic activity. Herein we demonstrate that TREM2 was highly expressed on myelin-laden phagocytes in active demyelinating lesions in the CNS of subjects with MS. In gene expression studies, macrophages from subjects with TREM2 genetic deficiency displayed a defect in phagocytic pathways. Treatment with a new TREM2 agonistic antibody promoted the clearance of myelin debris in the cuprizone model of CNS demyelination. Effects included enhancement of myelin uptake and degradation, resulting in accelerated myelin debris removal by microglia. Most importantly, antibody-dependent TREM2 activation on microglia increased density of oligodendrocyte precursors in areas of demyelination, as well as the formation of mature oligodendrocytes thus enhancing remyelination and axonal integrity. These results are relevant as they propose TREM2 on microglia as a potential new target to promote remyelination. Electronic supplementary material The online version of this article (10.1007/s00401-020-02193-z) contains supplementary material, which is available to authorized users.
- Published
- 2020
42. Post-mortem analyses of PiB and flutemetamol in diffuse and cored amyloid-β plaques in Alzheimer’s disease
- Author
-
William E. Klunk, Gill Farrar, Milos D. Ikonomovic, Chester A. Mathis, Christopher Buckley, Julia Kofler, and Eric E. Abrahamson
- Subjects
0301 basic medicine ,Male ,Pathology ,medicine.medical_specialty ,Amyloid ,Positron emission tomography ,Plaque, Amyloid ,Fibril ,Pathology and Forensic Medicine ,law.invention ,Striatum ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,0302 clinical medicine ,Confocal microscopy ,law ,Alzheimer Disease ,Flutemetamol ,Fluorescence microscope ,medicine ,Humans ,Pittsburgh compound B ,Benzothiazoles ,Aged ,Fluorescence microscopy ,Aged, 80 and over ,Original Paper ,Aniline Compounds ,medicine.diagnostic_test ,Chemistry ,Histology ,Middle Aged ,Thiazoles ,030104 developmental biology ,Positron-Emission Tomography ,Immunohistochemistry ,Female ,Neurology (clinical) ,Autopsy ,Radiopharmaceuticals ,030217 neurology & neurosurgery - Abstract
Specificity and sensitivity of positron emission tomography (PET) radiopharmaceuticals targeting fibrillar amyloid-β (Aβ) deposits is high for detection of neuritic Aβ plaques, a mature form of Aβ deposits which often have dense Aβ core (i.e., cored plaques). However, imaging-to-autopsy validation studies of amyloid PET radioligands have identified several false positive cases all of which had mainly diffuse Aβ plaques (i.e., plaques without neuritic pathology or dense amyloid core), and high amyloid PET signal was reported in the striatum where diffuse plaques predominate in Alzheimer’s disease (AD). Relative contributions of different plaque types to amyloid PET signal is unclear, particularly in neocortical areas where they are intermixed in AD. In vitro binding assay and autoradiography were performed using [3H]flutemetamol and [3H]Pittsburgh Compound-B (PiB) in frozen brain homogenates from 30 autopsy cases including sporadic AD and non-AD controls with a range of brain Aβ burden and plaque density. Fixed tissue sections of frontal cortex and caudate from 10 of the AD cases were processed for microscopy using fluorescent derivatives of flutemetamol (cyano-flutemetamol) and PiB (cyano-PiB) and compared to Aβ immunohistochemistry and pan-amyloid (X-34) histology. Using epifluorescence microscopy, percent area coverage and fluorescence output values of cyano-PiB- and cyano-flutemetamol-labeled plaques in two-dimensional microscopic fields were then calculated and combined to obtain integrated density measurements. Using confocal microscopy, we analysed total fluorescence output of the entire three-dimensional volume of individual cored plaques and diffuse plaques labeled with cyano-flutemetamol or cyano-PiB. [3H]Flutemetamol and [3H]PiB binding values in tissue homogenates correlated strongly and their binding pattern in tissue sections, as seen on autoradiograms, overlapped the pattern of Aβ-immunoreactive plaques on directly adjacent sections. Cyano-flutemetamol and cyano-PiB fluorescence was prominent in cored plaques and less so in diffuse plaques. Across brain regions and cases, percent area coverage of cyano-flutemetamol-labeled plaques correlated strongly with cyano-PiB-labeled and Aβ-immunoreactive plaques. For both ligands, plaque burden, calculated as percent area coverage of all Aβ plaque types, was similar in frontal cortex and caudate regions, while integrated density values were significantly greater in frontal cortex, which contained both cored plaques and diffuse plaques, compared to the caudate, which contained only diffuse plaques. Three-dimensional analysis of individual plaques labeled with either ligand showed that total fluorescence output of a single cored plaque was equivalent to total fluorescence output of approximately three diffuse plaques of similar volume. Our results indicate that [18F]flutemetamol and [11C]PiB PET signal is influenced by both diffuse plaques and cored plaques, and therefore is likely a function of plaque size and density of Aβ fibrils in plaques. Brain areas with large volumes/frequencies of diffuse plaques could yield [18F]flutemetamol and [11C]PiB PET retention levels comparable to brain regions with a lower volume/frequency of cored plaques.
- Published
- 2020
43. Interleukin-1 promotes autoimmune neuroinflammation by suppressing endothelial heme oxygenase-1 at the blood–brain barrier
- Author
-
Florian C. Kurschus, Tobias Bopp, Ingo Bechman, Ari Waisman, Harald Binder, Silvia Cardoso, Subhashini Bolisetty, Lisa Johann, Judith Hauptmann, Federico Marini, Elisa Colombo, Ilgiz A. Mufazalov, Markus Schwaninger, Miguel P. Soares, Judith Strauß, Sonja Moos, Anupam Agarwal, Fred Lühder, Matthias Klein, Maja Kitic, Florian Wanke, Khalad Karram, Martin Krueger, and Tommy Regen
- Subjects
0301 basic medicine ,Encephalomyelitis, Autoimmune, Experimental ,Experimental autoimmune encephalomyelitis (EAE) ,Autoimmunity ,Blood–brain barrier ,Pathology and Forensic Medicine ,Proinflammatory cytokine ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Chemokine receptor ,0302 clinical medicine ,medicine ,Animals ,Neuroinflammation ,Inflammation ,Original Paper ,Microglia ,Chemistry ,Experimental autoimmune encephalomyelitis ,Endothelial Cells ,Interleukin ,medicine.disease ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,Heme oxygenase ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Blood-Brain Barrier ,Heme oxygenase-1 (HO-1) ,Neurology (clinical) ,Heme Oxygenase-1 ,030217 neurology & neurosurgery ,Interleukin-1 ,Signal Transduction - Abstract
The proinflammatory cytokine interleukin 1 (IL-1) is crucially involved in the pathogenesis of multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). Herein, we studied the role of IL-1 signaling in blood–brain barrier (BBB) endothelial cells (ECs), astrocytes and microglia for EAE development, using mice with the conditional deletion of its signaling receptor IL-1R1. We found that IL-1 signaling in microglia and astrocytes is redundant for the development of EAE, whereas the IL-1R1 deletion in BBB-ECs markedly ameliorated disease severity. IL-1 signaling in BBB-ECs upregulated the expression of the adhesion molecules Vcam-1, Icam-1 and the chemokine receptor Darc, all of which have been previously shown to promote CNS-specific inflammation. In contrast, IL-1R1 signaling suppressed the expression of the stress-responsive heme catabolizing enzyme heme oxygenase-1 (HO-1) in BBB-ECs, promoting disease progression via a mechanism associated with deregulated expression of the IL-1-responsive genes Vcam1, Icam1 and Ackr1 (Darc). Mechanistically, our data emphasize a functional crosstalk of BBB-EC IL-1 signaling and HO-1, controlling the transcription of downstream proinflammatory genes promoting the pathogenesis of autoimmune neuroinflammation. Electronic supplementary material The online version of this article (10.1007/s00401-020-02187-x) contains supplementary material, which is available to authorized users.
- Published
- 2020
- Full Text
- View/download PDF
44. Large-scale pathway specific polygenic risk and transcriptomic community network analysis identifies novel functional pathways in Parkinson disease
- Author
-
Anthony R. Soltis, Ziv Gan-Or, Clifton L. Dalgard, Debra Ehrlich, Leonard H, Sara Saez-Atienzar, Cornelis Blauwendraat, Ali Torkamani, J. R. Gibbs, Sonja W. Scholz, Bryan J. Traynor, Clemens R. Scherzer, Jonggeol Jeff Kim, Jinhui Ding, Mark R. Cookson, Juan A. Botía, Matt Bookman, Andrew B. Singleton, Mike A. Nalls, Sara Bandres-Ciga, Monica Diez-Fairen, Hirotaka Iwaki, Lasse Pihlstrøm, Alastair J. Noyce, Dena G. Hernandez, Mina Ryten, Mary B. Makarious, and Faraz Faghri
- Subjects
Original Paper ,Polygenic risk ,Transcriptome community maps ,Context (language use) ,Computational biology ,Disease ,Quantitative trait locus ,Biology ,Chromatin remodeling ,Pathology and Forensic Medicine ,Parkinson disease ,Transcriptome ,Cellular and Molecular Neuroscience ,Mendelian randomization ,Neurology (clinical) ,Allele ,Signal transduction ,Gene - Abstract
Polygenic inheritance plays a central role in Parkinson disease (PD). A priority in elucidating PD etiology lies in defining the biological basis of genetic risk. Unraveling how risk leads to disruption will yield disease-modifying therapeutic targets that may be effective. Here, we utilized a high-throughput and hypothesis-free approach to determine biological processes underlying PD using the largest currently available cohorts of genetic and gene expression data from International Parkinson’s Disease Genetics Consortium (IPDGC) and the Accelerating Medicines Partnership-Parkinson’s disease initiative (AMP-PD), among other sources. We applied large-scale gene-set specific polygenic risk score (PRS) analyses to assess the role of common variation on PD risk focusing on publicly annotated gene sets representative of curated pathways. We nominated specific molecular sub-processes underlying protein misfolding and aggregation, post-translational protein modification, immune response, membrane and intracellular trafficking, lipid and vitamin metabolism, synaptic transmission, endosomal–lysosomal dysfunction, chromatin remodeling and apoptosis mediated by caspases among the main contributors to PD etiology. We assessed the impact of rare variation on PD risk in an independent cohort of whole-genome sequencing data and found evidence for a burden of rare damaging alleles in a range of processes, including neuronal transmission-related pathways and immune response. We explored enrichment linked to expression cell specificity patterns using single-cell gene expression data and demonstrated a significant risk pattern for dopaminergic neurons, serotonergic neurons, hypothalamic GABAergic neurons, and neural progenitors. Subsequently, we created a novel way of building de novo pathways by constructing a network expression community map using transcriptomic data derived from the blood of PD patients, which revealed functional enrichment in inflammatory signaling pathways, cell death machinery related processes, and dysregulation of mitochondrial homeostasis. Our analyses highlight several specific promising pathways and genes for functional prioritization and provide a cellular context in which such work should be done. Electronic supplementary material The online version of this article (10.1007/s00401-020-02181-3) contains supplementary material, which is available to authorized users.
- Published
- 2020
- Full Text
- View/download PDF
45. Prion propagation estimated from brain diffusion MRI is subtype dependent in sporadic Creutzfeldt–Jakob disease
- Author
-
Sara Garbarino, Janis Blevins, Alberto Bizzi, Mark L. Cohen, Lawrence B. Schonberger, Riccardo Pascuzzo, Pierluigi Gambetti, Alexandra L. Young, Neil P. Oxtoby, Brian S. Appleby, Gianmarco Castelli, and Daniel C. Alexander
- Subjects
Adult ,Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Prion disease ,Creutzfeldt–Jakob disease ,Precuneus ,Prion propagation ,Spongiform degeneration ,Creutzfeldt-Jakob Syndrome ,Prion Proteins ,Pathology and Forensic Medicine ,Lesion ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Magnetic resonance imaging ,0302 clinical medicine ,medicine ,Humans ,Aged ,Original Paper ,Disease progression ,Neocortex ,medicine.diagnostic_test ,business.industry ,Sporadic Creutzfeldt-Jakob disease ,Middle Aged ,Diffusion Magnetic Resonance Imaging ,Early Diagnosis ,030104 developmental biology ,medicine.anatomical_structure ,Female ,Epicentre ,Neurology (clinical) ,Abnormality ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Diffusion MRI - Abstract
Sporadic Creutzfeldt–Jakob disease (sCJD) is a transmissible brain proteinopathy. Five main clinicopathological subtypes (sCJD-MM(V)1, -MM(V)2C, -MV2K, -VV1, and -VV2) are currently distinguished. Histopathological evidence suggests that the localisation of prion aggregates and spongiform lesions varies among subtypes. Establishing whether there is an initial site with detectable imaging abnormalities (epicentre) and an order of lesion propagation would be informative for disease early diagnosis, patient staging, management and recruitment in clinical trials. Diffusion magnetic resonance imaging (MRI) is the most-used and most-sensitive test to detect spongiform degeneration. This study was designed to identify, in vivo and for the first time, subtype-dependent epicentre and lesion propagation in the brain using diffusion-weighted images (DWI), in the largest known cross-sectional dataset of autopsy-proven subjects with sCJD. We estimate lesion propagation by cross-sectional DWI using event-based modelling, a well-established data-driven technique. DWI abnormalities of 594 autopsy-diagnosed subjects (448 patients with sCJD) were scored in 12 brain regions by 1 neuroradiologist blind to the diagnosis. We used the event-based model to reconstruct sequential orderings of lesion propagation in each of five pure subtypes. Follow-up data from 151 patients validated the estimated sequences. Results showed that epicentre and ordering of lesion propagation are subtype specific. The two most common subtypes (-MM1 and -VV2) showed opposite ordering of DWI abnormality appearance: from the neocortex to subcortical regions, and vice versa, respectively. The precuneus was the most likely epicentre also in -MM2 and -VV1 although at variance with -MM1, abnormal signal was also detected early in cingulate and insular cortices. The caudal-rostral sequence of lesion propagation that characterises -VV2 was replicated in -MV2K. Combined, these data-driven models provide unprecedented dynamic insights into subtype-specific epicentre at onset and propagation of the pathologic process, which may also enhance early diagnosis and enable disease staging in sCJD.
- Published
- 2020
- Full Text
- View/download PDF
46. HIF-1α is involved in blood–brain barrier dysfunction and paracellular migration of bacteria in pneumococcal meningitis
- Author
-
Sven Hammerschmidt, Karl H. Plate, Roland Nau, Kavi Devraj, Daniel Spitzer, Sylvaine Guérit, Jadranka Macas, Stefan Günther, Maryam I Khel, Camelia M. Monoranu, Michel Mittelbronn, Anne K. Braczynski, Roxana Heidemann, Omolara O. Ogunshola, Gayatri Devraj, Jana Seele, Volkhard A. J. Kempf, Uwe Ködel, Wibke Ballhorn, University of Zurich, Devraj, Kavi, and Kempf, Volkhard A J
- Subjects
Male ,Vascular Endothelial Growth Factor A ,2804 Cellular and Molecular Neuroscience ,medicine.disease_cause ,Dexamethasone ,Mice ,chemistry.chemical_compound ,Blood–brain barrier (BBB) ,Aged, 80 and over ,Meningitis, Pneumococcal ,Middle Aged ,10081 Institute of Veterinary Physiology ,VEGF ,Vascular endothelial growth factor ,2728 Neurology (clinical) ,Streptococcus pneumoniae ,medicine.anatomical_structure ,Blood-Brain Barrier ,Female ,medicine.symptom ,Meningitis ,medicine.drug ,Adult ,Paracellular transmigration ,Endothelium ,Clinical Neurology ,HIF-1α ,Brain damage ,Blood–brain barrier ,Permeability ,Pathology and Forensic Medicine ,Cerebral edema ,Cellular and Molecular Neuroscience ,medicine ,HIF ,Animals ,Humans ,Aged ,Original Paper ,business.industry ,Transendothelial and Transepithelial Migration ,1α ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,S. pneumoniae ,2734 Pathology and Forensic Medicine ,Mice, Inbred C57BL ,chemistry ,Cancer research ,570 Life sciences ,biology ,Neurology (clinical) ,business - Abstract
Bacterial meningitis is a deadly disease most commonly caused by Streptococcus pneumoniae, leading to severe neurological sequelae including cerebral edema, seizures, stroke, and mortality when untreated. Meningitis is initiated by the transfer of S. pneumoniae from blood to the brain across the blood–cerebrospinal fluid barrier or the blood–brain barrier (BBB). The underlying mechanisms are still poorly understood. Current treatment strategies include adjuvant dexamethasone for inflammation and cerebral edema, followed by antibiotics. The success of dexamethasone is however inconclusive, necessitating new therapies for controlling edema, the primary reason for neurological complications. Since we have previously shown a general activation of hypoxia inducible factor (HIF-1α) in bacterial infections, we hypothesized that HIF-1α, via induction of vascular endothelial growth factor (VEGF) is involved in transmigration of pathogens across the BBB. In human, murine meningitis brain samples, HIF-1α activation was observed by immunohistochemistry. S. pneumoniae infection in brain endothelial cells (EC) resulted in in vitro upregulation of HIF-1α/VEGF (Western blotting/qRT-PCR) associated with increased paracellular permeability (fluorometry, impedance measurements). This was supported by bacterial localization at cell–cell junctions in vitro and in vivo in brain ECs from mouse and humans (confocal, super-resolution, electron microscopy, live-cell imaging). Hematogenously infected mice showed increased permeability, S. pneumoniae deposition in the brain, along with upregulation of genes in the HIF-1α/VEGF pathway (RNA sequencing of brain microvessels). Inhibition of HIF-1α with echinomycin, siRNA in bEnd5 cells or using primary brain ECs from HIF-1α knock-out mice revealed reduced endothelial permeability and transmigration of S. pneumoniae. Therapeutic rescue using the HIF-1α inhibitor echinomycin resulted in increased survival and improvement of BBB function in S. pneumoniae-infected mice. We thus demonstrate paracellular migration of bacteria across BBB and a critical role for HIF-1α/VEGF therein and hence propose targeting this pathway to prevent BBB dysfunction and ensuing brain damage in infections.
- Published
- 2020
- Full Text
- View/download PDF
47. Ultrasensitive RT-QuIC assay with high sensitivity and specificity for Lewy body-associated synucleinopathies
- Author
-
Simone Baiardi, Niccolò Candelise, Anna Ladogana, Sabina Capellari, Byron Caughey, Elena Antelmi, Giovanna Calandra-Buonaura, Piero Parchi, Giuseppe Plazzi, Giulia Giannini, Andrew G. Hughson, Marcello Rossi, Christina D. Orrù, Angela Mammana, Pietro Cortelli, Rossi M., Candelise N., Baiardi S., Capellari S., Giannini G., Orru C.D., Antelmi E., Mammana A., Hughson A.G., Calandra-Buonaura G., Ladogana A., Plazzi G., Cortelli P., Caughey B., and Parchi P.
- Subjects
Lewy Body Disease ,Pathology ,medicine.medical_specialty ,Parkinson's disease ,Synucleinopathies ,diagnosis ,prion disease ,Prion disease ,multiple system atrophy ,Sensitivity and Specificity ,REM sleep behavior disorder ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,Biomarker ,Diagnosis ,Multiple system atrophy ,Parkinson’s disease ,α-Synuclein ,mental disorders ,medicine ,Humans ,Dementia ,Cognitive decline ,Original Paper ,Lewy body ,business.industry ,Dementia with Lewy bodies ,Parkinsonism ,Correction ,medicine.disease ,nervous system diseases ,Spectrometry, Fluorescence ,alpha-Synuclein ,biomarker ,Neurology (clinical) ,business ,Diagnosi - Abstract
The clinical diagnosis of synucleinopathies, including Parkinson’s disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA), is challenging, especially at an early disease stage, due to the heterogeneous and often non-specific clinical manifestations. The discovery of reliable specific markers for synucleinopathies would consequently be of great aid to the diagnosis and management of these disorders. Real-Time Quaking-Induced Conversion (RT-QuIC) is an ultrasensitive technique that has been previously used to detect self-templating amyloidogenic proteins in the cerebrospinal fluid (CSF) and other biospecimens in prion disease and synucleinopathies. Using a wild-type recombinant α-synuclein as a substrate, we applied RT-QuIC to a large cohort of 439 CSF samples from clinically well-characterized, or post-mortem verified patients with parkinsonism or dementia. Of significance, we also studied patients with isolated REM sleep behavior disorder (iRBD) (n = 18) and pure autonomic failure (PAF) (n = 28), representing clinical syndromes that are often caused by a synucleinopathy, and may precede the appearance of parkinsonism or cognitive decline. The results show that our RT-QuIC assay can accurately detect α-synuclein seeding activity across the spectrum of Lewy Body (LB)-related disorders (LBD), including DLB, PD, iRBD, and PAF, with an overall sensitivity of 95.3%. In contrast, all but two patients with MSA showed no α-synuclein seeding activity in the applied experimental setting. The analysis of the fluorescence response reflecting the amount of α-synuclein seeds revealed no significant differences between the clinical syndromes associated with LB pathology. Finally, the assay demonstrated 98% specificity in a neuropathological cohort of 101 cases lacking LB pathology. In conclusion, α-synuclein RT-QuIC provides an accurate marker of synucleinopathies linked to LB pathology and may have a pivotal role in the early discrimination and management of affected patients. The finding of no α-synuclein seeding activity in MSA seems to support the current view that MSA and LBD are associated with different conformational strains of α-synuclein. Electronic supplementary material The online version of this article (10.1007/s00401-020-02160-8) contains supplementary material, which is available to authorized users.
- Published
- 2020
- Full Text
- View/download PDF
48. PTEN activation contributes to neuronal and synaptic engulfment by microglia in tauopathy
- Author
-
Jürgen Götz, Bradley T. Hyman, Harrison Tudor Evans, Rachel E. Bennett, Sevannah A. Ellis, Joseph Benetatos, and Liviu-Gabriel Bodea
- Subjects
Male ,Genetically modified mouse ,PTEN ,Phosphatase ,Pathology and Forensic Medicine ,Mice ,Cellular and Molecular Neuroscience ,Phagocytosis ,mental disorders ,medicine ,Animals ,Humans ,Tensin ,Neurodegeneration ,Aged ,Aged, 80 and over ,Neurons ,Original Paper ,biology ,Microglia ,Chemistry ,PTEN Phosphohydrolase ,Frontotemporal lobar degeneration ,Middle Aged ,medicine.disease ,Synapse ,Cell biology ,medicine.anatomical_structure ,Tauopathies ,Synapses ,biology.protein ,Female ,Neurology (clinical) ,Tauopathy ,Tau - Abstract
Phosphatase and tensin homolog (PTEN) regulates synaptic density in development; however, whether PTEN also regulates synapse loss in a neurodegenerative disorder such as frontotemporal lobar degeneration with Tau deposition (FTLD-Tau) has not been explored. Here, we found that pathological Tau promotes early activation of PTEN, which precedes apoptotic caspase-3 cleavage in the rTg4510 mouse model of FTLD-Tau. We further demonstrate increased synaptic and neuronal exposure of the apoptotic signal phosphatidylserine that tags neuronal structures for microglial uptake, thereby linking PTEN activation to synaptic and neuronal structure elimination. By applying pharmacological inhibition of PTEN's protein phosphatase activity, we observed that microglial uptake can be decreased in Tau transgenic mice. Finally, we reveal a dichotomous relationship between PTEN activation and age in FTLD-Tau patients and healthy controls. Together, our findings suggest that in tauopathy, PTEN has a role in the synaptotoxicity of pathological Tau and promotes microglial removal of affected neuronal structures. Electronic supplementary material The online version of this article (10.1007/s00401-020-02151-9) contains supplementary material, which is available to authorized users.
- Published
- 2020
- Full Text
- View/download PDF
49. Diverse, evolving conformer populations drive distinct phenotypes in frontotemporal lobar degeneration caused by the same MAPT-P301L mutation
- Author
-
Shelaine C. Fleck, Holger Wille, Serene Wohlgemuth, Erik Gomez-Cardona, Sergi Borrego-Écija, Laura Molina-Porcel, Zhuang Zhuang Han, Olivier Julien, Jiri G. Safar, Ghazaleh Eskandari-Sedighi, Nathalie Daude, Ellen Gelpi, Chae Kim, David Westaway, Tracy Haldiman, Jing Yang, and Sang-Gyun Kang
- Subjects
0301 basic medicine ,Genetically modified mouse ,Male ,Focal pathology ,Aging/P301L mutation ,Tau protein ,Seeding ,tau Proteins ,Biology ,medicine.disease_cause ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,0302 clinical medicine ,Germline mutation ,Transgenic mouse ,mental disorders ,medicine ,Animals ,Humans ,Conformation ,Aged ,Genetics ,Mutation ,Original Paper ,Brain ,Correction ,Frontotemporal lobar degeneration ,Human brain ,Middle Aged ,medicine.disease ,Phenotype ,Tauopathy ,030104 developmental biology ,medicine.anatomical_structure ,Tauopathies ,Strain ensembles ,biology.protein ,Female ,Neurology (clinical) ,Frontotemporal Lobar Degeneration ,030217 neurology & neurosurgery - Abstract
Tau protein accumulation is a common denominator of major dementias, but this process is inhomogeneous, even when triggered by the same germline mutation. We considered stochastic misfolding of human tau conformers followed by templated conversion of native monomers as an underlying mechanism and derived sensitive conformational assays to test this concept. Assessments of brains from aged TgTauP301L transgenic mice revealed a prodromal state and three distinct signatures for misfolded tau. Frontotemporal lobar degeneration (FTLD)-MAPT-P301L patients with different clinical phenotypes also displayed three signatures, two resembling those found in TgTauP301L mice. As physicochemical and cell bioassays confirmed diverse tau strains in the mouse and human brain series, we conclude that evolution of diverse tau conformers is intrinsic to the pathogenesis of this uni-allelic form of tauopathy. In turn, effective therapeutic interventions in FTLD will need to address evolving repertoires of misfolded tau species rather than singular, static molecular targets. Electronic supplementary material The online version of this article (10.1007/s00401-020-02148-4) contains supplementary material, which is available to authorized users.
- Published
- 2020
50. Identification of early pericyte loss and vascular amyloidosis in Alzheimer’s disease retina
- Author
-
Julia Sheyn, Andrei A. Kramerov, Alexander V. Ljubimov, Anthony Rodriguez, Yosef Koronyo, Nazanin Mirzaei, Oana M. Dumitrascu, Ernesto Barron, Giovanna C. Regis, Dieu-Trang Fuchs, David R. Hinton, Maya Koronyo-Hamaoui, Keith L. Black, Carol A. Miller, Altan Rentsendorj, and Haoshen Shi
- Subjects
0301 basic medicine ,Male ,Pathology ,medicine.medical_specialty ,Amyloid ,Vascular damage ,Retina ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Amyloid beta-Protein Precursor ,0302 clinical medicine ,Cognition ,Alzheimer Disease ,medicine ,Humans ,Cognitive decline ,Neurodegeneration ,Retinopathy ,Aged ,Aged, 80 and over ,Original Paper ,Amyloid beta-Peptides ,business.industry ,Amyloidosis ,Brain ,Retinal ,medicine.disease ,Cerebral Amyloid Angiopathy ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Blood-Brain Barrier ,Female ,Neurology (clinical) ,Pericyte ,Cerebral amyloid angiopathy ,business ,Pericytes ,Alzheimer’s disease ,030217 neurology & neurosurgery - Abstract
Pericyte loss and deficient vascular platelet-derived growth factor receptor-β (PDGFRβ) signaling are prominent features of the blood–brain barrier breakdown described in Alzheimer’s disease (AD) that can predict cognitive decline yet have never been studied in the retina. Recent reports using noninvasive retinal amyloid imaging, optical coherence tomography angiography, and histological examinations support the existence of vascular-structural abnormalities and vascular amyloid β-protein (Aβ) deposits in retinas of AD patients. However, the cellular and molecular mechanisms of such retinal vascular pathology were not previously explored. Here, by modifying a method of enzymatically clearing non-vascular retinal tissue and fluorescent immunolabeling of the isolated blood vessel network, we identified substantial pericyte loss together with significant Aβ deposition in retinal microvasculature and pericytes in AD. Evaluation of postmortem retinas from a cohort of 56 human donors revealed an early and progressive decrease in vascular PDGFRβ in mild cognitive impairment (MCI) and AD compared to cognitively normal controls. Retinal PDGFRβ loss significantly associated with increased retinal vascular Aβ40 and Aβ42 burden. Decreased vascular LRP-1 and early apoptosis of pericytes in AD retina were also detected. Mapping of PDGFRβ and Aβ40 levels in pre-defined retinal subregions indicated that certain geometrical and cellular layers are more susceptible to AD pathology. Further, correlations were identified between retinal vascular abnormalities and cerebral Aβ burden, cerebral amyloid angiopathy (CAA), and clinical status. Overall, the identification of pericyte and PDGFRβ loss accompanying increased vascular amyloidosis in Alzheimer’s retina implies compromised blood–retinal barrier integrity and provides new targets for AD diagnosis and therapy. Electronic supplementary material The online version of this article (10.1007/s00401-020-02134-w) contains supplementary material, which is available to authorized users.
- Published
- 2020
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.