Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 408
Filter
1.
Brain Behav ; 14(6): e3552, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38953731

ABSTRACT

INTRODUCTION: Hepcidin is a peptide associated with controlling the distribution of iron in tissues. Growing interest is linked with its impact on neurodegenerative diseases, as disruption of the iron regulation may be considered an initiatory element of pathological protein accumulation. The possible impact of hepcidin was not previously sufficiently explored in progressive supranuclear palsy (PSP). METHODS: Twelve patients with PSP-Richardson's syndrome (PSP-RS), 12 with PSP-Parkinsonism Predominant (PSP-P), and 12 controls were examined using Unified Parkinson's Disease Rating Scale-III part (UPDRS-III) in OFF stage and analyzed in the context of hepcidin levels in the serum. RESULTS: The work revealed increased levels of hepcidin in PSP-RS when compared to PSP-P and controls. Moreover, hepcidin was found to be negatively correlated with UPDRS-III results in PSP-RS, whereas positively in PSP-P. CONCLUSION: The work may suggest a possible impact of hepcidin in PSP, possibly differing depending on its subtype.


Subject(s)
Hepcidins , Supranuclear Palsy, Progressive , Aged , Female , Humans , Male , Middle Aged , Hepcidins/blood , Severity of Illness Index , Supranuclear Palsy, Progressive/blood , Supranuclear Palsy, Progressive/metabolism
2.
Brain ; 147(7): 2428-2439, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38842726

ABSTRACT

Four-repeat (4R) tauopathies are neurodegenerative diseases characterized by cerebral accumulation of 4R tau pathology. The most prominent 4R tauopathies are progressive supranuclear palsy (PSP) and corticobasal degeneration characterized by subcortical tau accumulation and cortical neuronal dysfunction, as shown by PET-assessed hypoperfusion and glucose hypometabolism. Yet, there is a spatial mismatch between subcortical tau deposition patterns and cortical neuronal dysfunction, and it is unclear how these two pathological brain changes are interrelated. Here, we hypothesized that subcortical tau pathology induces remote neuronal dysfunction in functionally connected cortical regions to test a pathophysiological model that mechanistically links subcortical tau accumulation to cortical neuronal dysfunction in 4R tauopathies. We included 51 Aß-negative patients with clinically diagnosed PSP variants (n = 26) or corticobasal syndrome (n = 25) who underwent structural MRI and 18F-PI-2620 tau-PET. 18F-PI-2620 tau-PET was recorded using a dynamic one-stop-shop acquisition protocol to determine an early 0.5-2.5 min post tracer-injection perfusion window for assessing cortical neuronal dysfunction, as well as a 20-40 min post tracer-injection window to determine 4R-tau load. Perfusion-PET (i.e. early window) was assessed in 200 cortical regions, and tau-PET was assessed in 32 subcortical regions of established functional brain atlases. We determined tau epicentres as subcortical regions with the highest 18F-PI-2620 tau-PET signal and assessed the connectivity of tau epicentres to cortical regions of interest using a resting-state functional MRI-based functional connectivity template derived from 69 healthy elderly controls from the ADNI cohort. Using linear regression, we assessed whether: (i) higher subcortical tau-PET was associated with reduced cortical perfusion; and (ii) cortical perfusion reductions were observed preferentially in regions closely connected to subcortical tau epicentres. As hypothesized, higher subcortical tau-PET was associated with overall lower cortical perfusion, which remained consistent when controlling for cortical tau-PET. Using group-average and subject-level PET data, we found that the seed-based connectivity pattern of subcortical tau epicentres aligned with cortical perfusion patterns, where cortical regions that were more closely connected to the tau epicentre showed lower perfusion. Together, subcortical tau-accumulation is associated with remote perfusion reductions indicative of neuronal dysfunction in functionally connected cortical regions in 4R-tauopathies. This suggests that subcortical tau pathology may induce cortical dysfunction, which may contribute to clinical disease manifestation and clinical heterogeneity.


Subject(s)
Cerebral Cortex , Positron-Emission Tomography , Supranuclear Palsy, Progressive , Tauopathies , tau Proteins , Humans , Male , Female , Positron-Emission Tomography/methods , Aged , Tauopathies/diagnostic imaging , Tauopathies/metabolism , Tauopathies/pathology , tau Proteins/metabolism , Middle Aged , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Supranuclear Palsy, Progressive/diagnostic imaging , Supranuclear Palsy, Progressive/metabolism , Supranuclear Palsy, Progressive/pathology , Supranuclear Palsy, Progressive/physiopathology , Magnetic Resonance Imaging/methods
3.
Ann Neurol ; 96(2): 247-261, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38771066

ABSTRACT

OBJECTIVE: Although astrocytic pathology is a pathological hallmark of progressive supranuclear palsy (PSP), its pathophysiological role remains unclear. This study aimed to assess astrocyte reactivity in vivo in patients with PSP. Furthermore, we investigated alterations in brain lactate levels and their relationship with astrocyte reactivity. METHODS: We included 30 patients with PSP-Richardson syndrome and 30 healthy controls; in patients, tau deposition was confirmed through 18F-florzolotau positron emission tomography. Myo-inositol, an astroglial marker, and lactate were quantified in the anterior cingulate cortex through magnetic resonance spectroscopy. We measured plasma biomarkers, including glial fibrillary acidic protein as another astrocytic marker. The anterior cingulate cortex was histologically assessed in postmortem samples of another 3 patients with PSP with comparable disease durations. RESULTS: The levels of myo-inositol and plasma glial fibrillary acidic protein were significantly higher in patients than those in healthy controls (p < 0.05); these increases were significantly associated with PSP rating scale and cognitive function scores (p < 0.05). The lactate level was high in patients, and correlated significantly with high myo-inositol levels. Histological analysis of the anterior cingulate cortex in patients revealed reactive astrocytes, despite mild tau deposition, and no marked synaptic loss. INTERPRETATION: We discovered high levels of astrocyte biomarkers in patients with PSP, suggesting astrocyte reactivity. The association between myo-inositol and lactate levels suggests a link between reactive astrocytes and brain energy metabolism changes. Our results indicate that astrocyte reactivity in the anterior cingulate cortex precedes pronounced tau pathology and neurodegenerative processes in that region, and affects brain function in PSP. ANN NEUROL 2024;96:247-261.


Subject(s)
Astrocytes , Glial Fibrillary Acidic Protein , Gyrus Cinguli , Inositol , Lactic Acid , Supranuclear Palsy, Progressive , Humans , Supranuclear Palsy, Progressive/metabolism , Supranuclear Palsy, Progressive/diagnostic imaging , Supranuclear Palsy, Progressive/pathology , Astrocytes/metabolism , Astrocytes/pathology , Male , Female , Aged , Middle Aged , Glial Fibrillary Acidic Protein/metabolism , Glial Fibrillary Acidic Protein/blood , Lactic Acid/blood , Lactic Acid/metabolism , Inositol/metabolism , Gyrus Cinguli/metabolism , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/pathology , Biomarkers/blood , tau Proteins/metabolism , Positron-Emission Tomography
4.
Neurology ; 102(11): e209453, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38759132

ABSTRACT

BACKGROUND AND OBJECTIVES: Degeneration of the presynaptic nigrostriatal dopaminergic system is one of the main biological features of Parkinson disease (PD), multiple system atrophy (MSA), progressive supranuclear palsy (PSP), and corticobasal degeneration (CBD), which can be measured using single-photon emission CT imaging for diagnostic purposes. Despite its widespread use in clinical practice and research, the diagnostic properties of presynaptic nigrostriatal dopaminergic (DAT) imaging in parkinsonism have never been evaluated against the diagnostic gold standard of neuropathology. The aim of this study was to evaluate the diagnostic parameters of DAT imaging compared with pathologic diagnosis in patients with parkinsonism. METHODS: Retrospective cohort study of patients with DAT imaging for the investigation of a clinically uncertain parkinsonism with brain donation between 2010 and 2021 to the Queen Square Brain Bank (London). Patients with DAT imaging for investigation of pure ataxia or dementia syndromes without parkinsonism were excluded. Those with a pathologic diagnosis of PD, MSA, PSP, or CBD were considered presynaptic dopaminergic parkinsonism, and other pathologies were considered postsynaptic for the analysis. DAT imaging was performed in routine clinical practice and visually classified by hospital nuclear medicine specialists as normal or abnormal. The results were correlated with neuropathologic diagnosis to calculate diagnostic accuracy parameters for the diagnosis of presynaptic dopaminergic parkinsonism. RESULTS: All of 47 patients with PD, 41 of 42 with MSA, 68 of 73 with PSP, and 6 of 10 with CBD (sensitivity 100%, 97.6%, 93.2%, and 60%, respectively) had abnormal presynaptic dopaminergic imaging. Eight of 17 patients with presumed postsynaptic parkinsonism had abnormal scans (specificity 52.9%). DISCUSSION: DAT imaging has very high sensitivity and negative predictive value for the diagnosis of presynaptic dopaminergic parkinsonism, particularly for PD. However, patients with CBD, and to a lesser extent PSP (of various phenotypes) and MSA (with predominant ataxia), can show normal DAT imaging. A range of other neurodegenerative disorders may have abnormal DAT scans with low specificity in the differential diagnosis of parkinsonism. DAT imaging is a useful diagnostic tool in the differential diagnosis of parkinsonism, although clinicians should be aware of its diagnostic properties and limitations. CLASSIFICATION OF EVIDENCE: This study provides Class II evidence that DAT imaging does not accurately distinguish between presynaptic dopaminergic parkinsonism and non-presynaptic dopaminergic parkinsonism.


Subject(s)
Dopamine Plasma Membrane Transport Proteins , Multiple System Atrophy , Parkinsonian Disorders , Tomography, Emission-Computed, Single-Photon , Humans , Female , Aged , Male , Retrospective Studies , Dopamine Plasma Membrane Transport Proteins/metabolism , Parkinsonian Disorders/diagnostic imaging , Parkinsonian Disorders/pathology , Parkinsonian Disorders/metabolism , Tomography, Emission-Computed, Single-Photon/methods , Middle Aged , Multiple System Atrophy/diagnostic imaging , Multiple System Atrophy/pathology , Multiple System Atrophy/metabolism , Supranuclear Palsy, Progressive/diagnostic imaging , Supranuclear Palsy, Progressive/pathology , Supranuclear Palsy, Progressive/metabolism , Aged, 80 and over , Parkinson Disease/diagnostic imaging , Parkinson Disease/metabolism , Parkinson Disease/pathology , Cohort Studies , Corticobasal Degeneration/diagnostic imaging , Corticobasal Degeneration/metabolism , Dopamine/metabolism , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology , Sensitivity and Specificity , Dopaminergic Imaging
5.
J Neurol ; 271(7): 4267-4280, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38632125

ABSTRACT

OBJECTIVE: Frontal hypometabolism on FDG-PET is observed in progressive supranuclear palsy (PSP), although it is unclear whether it is a feature of all PSP clinical variants and hence whether it is a useful diagnostic feature. We aimed to compare the frequency, severity, and pattern of frontal hypometabolism across PSP variants and determine whether frontal hypometabolism is related to clinical dysfunction. METHODS: Frontal hypometabolism in prefrontal, premotor, and sensorimotor cortices was visually graded on a 0-3 scale using CortexID Z-score images in 137 PSP patients. Frontal asymmetry was recorded. Severity scores were used to categorize patients as premotor-predominant, prefrontal-predominant, sensorimotor-predominant, mixed-predominance, or no regional predominance. Frontal ratings were compared across PSP clinical variants, and Spearman correlations were used to assess relationships with the Frontal Assessment Battery (FAB). RESULTS: 97% showed evidence of frontal hypometabolism which was most common (100%) in the speech-language (PSP-SL), corticobasal (PSP-CBS), and frontal (PSP-F) variants and least common in the progressive gait freezing (PSP-PGF) variant (73%). PSP-SL and PSP-CBS showed more severe hypometabolism than Richardson's syndrome (PSP-RS), Parkinsonism (PSP-P), and PSP-PGF. A premotor-predominant pattern was most common in PSP-SL and PSP-CBS, with more mixed patterns in the other variants. Hypometabolism was most commonly asymmetric in PSP-SL, PSP-P, PSP-F and PSP-CBS. Worse hypometabolism in nearly all frontal regions correlated with worse scores on the FAB. CONCLUSIONS: Frontal hypometabolism is a common finding in PSP, although it varies in severity and pattern across PSP variants and will likely be the most diagnostically useful in PSP-SL and PSP-CBS.


Subject(s)
Frontal Lobe , Positron-Emission Tomography , Supranuclear Palsy, Progressive , Humans , Supranuclear Palsy, Progressive/diagnostic imaging , Supranuclear Palsy, Progressive/metabolism , Female , Male , Aged , Middle Aged , Frontal Lobe/diagnostic imaging , Frontal Lobe/metabolism , Fluorodeoxyglucose F18 , Severity of Illness Index , Aged, 80 and over
6.
Cell ; 187(10): 2446-2464.e22, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38582079

ABSTRACT

Tauopathies are age-associated neurodegenerative diseases whose mechanistic underpinnings remain elusive, partially due to a lack of appropriate human models. Here, we engineered human induced pluripotent stem cell (hiPSC)-derived neuronal lines to express 4R Tau and 4R Tau carrying the P301S MAPT mutation when differentiated into neurons. 4R-P301S neurons display progressive Tau inclusions upon seeding with Tau fibrils and recapitulate features of tauopathy phenotypes including shared transcriptomic signatures, autophagic body accumulation, and reduced neuronal activity. A CRISPRi screen of genes associated with Tau pathobiology identified over 500 genetic modifiers of seeding-induced Tau propagation, including retromer VPS29 and genes in the UFMylation cascade. In progressive supranuclear palsy (PSP) and Alzheimer's Disease (AD) brains, the UFMylation cascade is altered in neurofibrillary-tangle-bearing neurons. Inhibiting the UFMylation cascade in vitro and in vivo suppressed seeding-induced Tau propagation. This model provides a robust platform to identify novel therapeutic strategies for 4R tauopathy.


Subject(s)
Induced Pluripotent Stem Cells , Neurons , Tauopathies , tau Proteins , Humans , Induced Pluripotent Stem Cells/metabolism , tau Proteins/metabolism , Tauopathies/metabolism , Tauopathies/pathology , Neurons/metabolism , Neurons/pathology , Animals , Mice , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Brain/metabolism , Brain/pathology , Supranuclear Palsy, Progressive/metabolism , Supranuclear Palsy, Progressive/pathology , Supranuclear Palsy, Progressive/genetics , Cell Differentiation , Mutation , Autophagy
7.
Clin Nucl Med ; 49(5): 387-396, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38465965

ABSTRACT

BACKGROUND: Progressive supranuclear palsy (PSP) is a tauopathy that involves subcortical regions but also extends to cortical areas. The clinical impact of different tau protein sites and their influence on glymphatic dysfunction have not been investigated. PATIENTS AND METHODS: Participants (n = 55; 65.6 ± 7.1 years; 29 women) with PSP (n = 32) and age-matched normal controls (NCs; n = 23) underwent 18 F-Florzolotau tau PET, MRI, PSP Rating Scale (PSPRS), and Mini-Mental State Examination. Cerebellar gray matter (GM) and parametric estimation of reference signal intensity were used as references for tau burden measured by SUV ratios. Glymphatic activity was measured by diffusion tensor image analysis along the perivascular space (DTI-ALPS). RESULTS: Parametric estimation of reference signal intensity is a better reference than cerebellar GM to distinguish tau burden between PSP and NCs. PSP patients showed higher cortical and subcortical tau SUV ratios than NCs ( P < 0.001 and <0.001). Cortical and subcortical tau deposition correlated with PSPRS, UPDRS, and Mini-Mental State Examination scores (all P 's < 0.05). Cortical tau deposition was further associated with the DTI-ALPS index and frontal-temporal-parietal GM atrophy. The DTI-ALPS indexes showed a significantly negative correlation with the PSPRS total scores ( P < 0.01). Finally, parietal and occipital lobe tau depositions showed mediating effects between the DTI-ALPS index and PSPRS score. CONCLUSIONS: Cortical tau deposition is associated with glymphatic dysfunction and plays a role in mediating glymphatic dysfunction and clinical severity. Our results provide a possible explanation for the worsening of clinical severity in patients with PSP.


Subject(s)
Supranuclear Palsy, Progressive , tau Proteins , Humans , Female , tau Proteins/metabolism , Supranuclear Palsy, Progressive/metabolism , Magnetic Resonance Imaging , Image Processing, Computer-Assisted
8.
Mov Disord ; 39(5): 847-854, 2024 May.
Article in English | MEDLINE | ID: mdl-38477228

ABSTRACT

BACKGROUND: As a biomarker targeting vesicular monoamine transporter 2 (VMAT2), 18F-9-fluoropropyldihydrotetrabenazine (18F-FP-DTBZ) positron emission tomography (PET) is highly accurate in diagnosing Parkinson's disease (PD) and assessing its severity. However, evidence is insufficient in patients with progressive supranuclear palsy (PSP). OBJECTIVE: We evaluated the striatal and extrastriatal monoaminergic disruption of PSP and differences in patterns between patients with PSP, PD, and healthy controls (HCs) using 18F-FP-DTBZ PET, as well as its correlations with the clinical characteristics of PSP. METHODS: We recruited 58 patients with PSP, 23 age- and duration-matched patients with PD, as well as 17 HCs. Patients were scanned using 18F-FP-DTBZ PET/computed tomography, and images were spatially normalized and analyzed based on the volume of interest. RESULTS: VMAT2 binding differed significantly in the striatum and substantia nigra among the groups (P < 0.001). A more severe disruption in the caudate was noted in the PSP group (P < 0.001) than in the PD group. However, no differences were found in the nucleus accumbens, hippocampus, amygdala, or raphe between the PD and PSP groups. Within the PSP group, striatal VMAT2 binding was significantly associated with the fall/postural stability subscore of the PSP Rating Scale, especially in the putamen. Furthermore, VMAT2 binding was correlated with Mini-Mental State Examination or Montreal Cognitive Assessment in the hippocampus. CONCLUSIONS: Caudate disruptions showed prominent differences among the groups. VAMT2 binding in the striatum and hippocampus reflects the severity of fall/postural stability and cognition, respectively. © 2024 International Parkinson and Movement Disorder Society.


Subject(s)
Corpus Striatum , Parkinson Disease , Supranuclear Palsy, Progressive , Vesicular Monoamine Transport Proteins , Humans , Supranuclear Palsy, Progressive/diagnostic imaging , Supranuclear Palsy, Progressive/metabolism , Male , Female , Aged , Middle Aged , Vesicular Monoamine Transport Proteins/metabolism , Corpus Striatum/metabolism , Corpus Striatum/diagnostic imaging , Parkinson Disease/metabolism , Parkinson Disease/diagnostic imaging , Positron-Emission Tomography/methods , Tetrabenazine/analogs & derivatives , Substantia Nigra/diagnostic imaging , Substantia Nigra/metabolism , Substantia Nigra/pathology , Positron Emission Tomography Computed Tomography/methods
10.
Brain Pathol ; 34(2): e13238, 2024 03.
Article in English | MEDLINE | ID: mdl-38214380

ABSTRACT

With the new era of disease-modifying therapies for neurodegenerative diseases, a novel approach for the molecular classification of neurodegenerative diseases is needed. In this research letter, there is a summary of the advances made in Alzheimer's disease, Lewy body disorders, and progressive supranuclear palsy toward this classification.


Subject(s)
Alzheimer Disease , Lewy Body Disease , Neurodegenerative Diseases , Supranuclear Palsy, Progressive , Humans , tau Proteins/metabolism , alpha-Synuclein/metabolism , Supranuclear Palsy, Progressive/metabolism , Phenotype
11.
Cells ; 12(24)2023 12 06.
Article in English | MEDLINE | ID: mdl-38132096

ABSTRACT

Progressive supranuclear palsy (PSP) is a neurodegenerative disease characterized by four-repeat tau deposition in various cell types and anatomical regions, and can manifest as several clinical phenotypes, including the most common phenotype, Richardson's syndrome. The limited availability of biomarkers for PSP relates to the overlap of clinical features with other neurodegenerative disorders, but identification of a growing number of biomarkers from imaging is underway. One way to increase the reliability of imaging biomarkers is to combine different modalities for multimodal imaging. This review aimed to provide an overview of the current state of PSP hybrid imaging by combinations of positron emission tomography (PET) and magnetic resonance imaging (MRI). Specifically, combined PET and MRI studies in PSP highlight the potential of [18F]AV-1451 to detect tau, but also the challenge in differentiating PSP from other neurodegenerative diseases. Studies over the last years showed a reduced synaptic density in [11C]UCB-J PET, linked [11C]PK11195 and [18F]AV-1451 markers to disease progression, and suggested the potential role of [18F]RO948 PET for identifying tau pathology in subcortical regions. The integration of quantitative global and regional gray matter analysis by MRI may further guide the assessment of reduced cortical thickness or volume alterations, and diffusion MRI could provide insight into microstructural changes and structural connectivity in PSP. Challenges in radiopharmaceutical biomarkers and hybrid imaging require further research targeting markers for comprehensive PSP diagnosis.


Subject(s)
Neurodegenerative Diseases , Supranuclear Palsy, Progressive , Humans , Supranuclear Palsy, Progressive/diagnostic imaging , Supranuclear Palsy, Progressive/metabolism , Radiopharmaceuticals , Reproducibility of Results , Multimodal Imaging , Biomarkers/analysis
12.
Proc Natl Acad Sci U S A ; 120(41): e2300258120, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37801475

ABSTRACT

Despite much effort, antibody therapies for Alzheimer's disease (AD) have shown limited efficacy. Challenges to the rational design of effective antibodies include the difficulty of achieving specific affinity to critical targets, poor expression, and antibody aggregation caused by buried charges and unstructured loops. To overcome these challenges, we grafted previously determined sequences of fibril-capping amyloid inhibitors onto a camel heavy chain antibody scaffold. These sequences were designed to cap fibrils of tau, known to form the neurofibrillary tangles of AD, thereby preventing fibril elongation. The nanobodies grafted with capping inhibitors blocked tau aggregation in biosensor cells seeded with postmortem brain extracts from AD and progressive supranuclear palsy (PSP) patients. The tau capping nanobody inhibitors also blocked seeding by recombinant tau oligomers. Another challenge to the design of effective antibodies is their poor blood-brain barrier (BBB) penetration. In this study, we also designed a bispecific nanobody composed of a nanobody that targets a receptor on the BBB and a tau capping nanobody inhibitor, conjoined by a flexible linker. We provide evidence that the bispecific nanobody improved BBB penetration over the tau capping inhibitor alone after intravenous administration in mice. Our results suggest that the design of synthetic antibodies that target sequences that drive protein aggregation may be a promising approach to inhibit the prion-like seeding of tau and other proteins involved in AD and related proteinopathies.


Subject(s)
Alzheimer Disease , Single-Domain Antibodies , Supranuclear Palsy, Progressive , Humans , Animals , Mice , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , tau Proteins/metabolism , Single-Domain Antibodies/pharmacology , Single-Domain Antibodies/metabolism , Neurofibrillary Tangles/metabolism , Supranuclear Palsy, Progressive/metabolism , Antibodies/metabolism , Brain/metabolism
13.
J Alzheimers Dis ; 96(1): 57-64, 2023.
Article in English | MEDLINE | ID: mdl-37742642

ABSTRACT

The first primary age-related tauopathy (PART) genome-wide association study confirmed significant associations of Alzheimer's disease (AD) and progressive supranuclear palsy (PSP) genetic variants with PART, and highlighted a novel genetic variant rs56405341. Here, we perform a comprehensive analysis of rs56405341. We found that rs56405341 was significantly associated with C4orf33 mRNA expression, but not JADE1 mRNA expression in multiple brain tissues. C4orf33 was mainly expressed in cerebellar hemisphere and cerebellum, and JADE1 was mainly expressed in thyroid, and coronary artery. Meanwhile, we found significantly downregulated C4orf33 expression both AD and PSP compared with normal controls, respectively.


Subject(s)
Alzheimer Disease , Supranuclear Palsy, Progressive , Tauopathies , Humans , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Supranuclear Palsy, Progressive/genetics , Supranuclear Palsy, Progressive/metabolism , Genome-Wide Association Study , RNA, Messenger
14.
J Med Chem ; 66(15): 10628-10638, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37487189

ABSTRACT

A chemical fingerprint search identified Z3777013540 (1-(5-(6-fluoro-1H-indol-2-yl)pyrimidin-2-yl)piperidin-4-ol; 1) as a potential 4R-tau binding ligand. Binding assays in post-mortem Alzheimer's disease (AD), progressive supranuclear palsy (PSP), and corticobasal degeneration (CBD) brain with [3H]1 provided KD (nM) values in AD = 4.0, PSP = 5.1, and CBD = 4.5. In vivo positron emission tomography (PET) imaging in rats with [18F]1 demonstrated good brain penetration and rapid clearance from normal brain tissues. A subsequent molecular similarity search using 1 as the query revealed an additional promising compound, Z4169252340 (4-(5-(6-fluoro-1H-indol-2-yl)pyrimidin-2-yl)morpholine; 21). Binding assays with [3H]21 provided KD (nM) values in AD = 1.2, PSP = 1.6, and CBD = 1.7 and lower affinities for binding aggregated α-synuclein and amyloid-beta. PET imaging in rats with [18F]21 demonstrated a higher brain penetration than [18F]1 and rapid clearance from normal brain tissues. We anticipate that 1 and 21 will be useful for the identification of other potent novel 4R-tau radiotracers.


Subject(s)
Alzheimer Disease , Supranuclear Palsy, Progressive , Tauopathies , Animals , Rats , tau Proteins/metabolism , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Supranuclear Palsy, Progressive/metabolism , Positron-Emission Tomography/methods , Brain/diagnostic imaging , Brain/metabolism
15.
Neurology ; 101(3): e289-e299, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37268436

ABSTRACT

BACKGROUND AND OBJECTIVES: Corticobasal syndrome (CBS) is a clinical phenotype characterized by asymmetric parkinsonism, rigidity, myoclonus, and apraxia. Originally believed secondary to corticobasal degeneration (CBD), mounting clinicopathologic studies have revealed heterogenous neuropathologies. The objectives of this study were to determine the pathologic heterogeneity of CBS, the clinicoradiologic findings associated with different underlying pathologies causing CBS, and the positive predictive value (PPV) of current diagnostic criteria for CBD among patients with a CBS. METHODS: Clinical data, brain MRI, and neuropathologic data of patients followed at Mayo Clinic and diagnosed with CBS antemortem were reviewed according to neuropathology category at autopsy. RESULTS: The cohort consisted of 113 patients with CBS, 61 (54%) female patients. Mean ± SD disease duration was 7 ± 3.7 years; mean ± SD age at death was 70.5 ± 9.1 years. The primary neuropathologic diagnoses were 43 (38%) CBD, 27 (24%) progressive supranuclear palsy (PSP), 17 (15%) Alzheimer disease (AD), 10 (9%) frontotemporal lobar degeneration (FTLD) with TAR DNA-binding protein 43 (TDP) inclusions, 7 (6%) diffuse Lewy body disease (DLBD)/AD, and 9 (8%) with other diagnoses. Patients with CBS-AD or CBS-DLBD/AD were youngest at death (median [interquartile range]: 64 [13], 64 [11] years) while CBS-PSP were oldest (77 [12.5] years, p = 0.024). Patients with CBS-DLBD/AD had the longest disease duration (9 [6] years), while CBS-other had the shortest (3 [4.25] years, p = 0.04). Posterior cortical signs and myoclonus were more characteristic of patients with CBS-AD and patients with CBS-DLBD/AD. Patients with CBS-DLBD/AD displayed more features of Lewy body dementia. Voxel-based morphometry revealed widespread cortical gray matter loss characteristic of CBS-AD, while CBS-CBD and CBS-PSP predominantly involved premotor regions with greater amount of white matter loss. Patients with CBS-DLBD/AD showed atrophy in a focal parieto-occipital region, and patients with CBS-FTLD-TDP had predominant prefrontal cortical loss. Patients with CBS-PSP had the lowest midbrain/pons ratio (p = 0.012). Of 67 cases meeting clinical criteria for possible CBD at presentation, 27 were pathology-proven CBD, yielding a PPV of 40%. DISCUSSION: A variety of neurodegenerative disorders can be identified in patients with CBS, but clinical and regional imaging differences aid in predicting underlying neuropathology. PPV analysis of the current CBD diagnostic criteria revealed suboptimal performance. Biomarkers adequately sensitive and specific for CBD are needed.


Subject(s)
Alzheimer Disease , Corticobasal Degeneration , Lewy Body Disease , Myoclonus , Supranuclear Palsy, Progressive , Female , Male , Humans , Myoclonus/complications , Supranuclear Palsy, Progressive/metabolism , Alzheimer Disease/complications , Magnetic Resonance Imaging , Lewy Body Disease/diagnostic imaging , Lewy Body Disease/complications
16.
Expert Opin Ther Targets ; 27(4-5): 269-279, 2023.
Article in English | MEDLINE | ID: mdl-37140480

ABSTRACT

INTRODUCTION: Tau has commanded much attention as a potential therapeutic target in neurodegenerative diseases. Tau pathology is a hallmark of primary tauopathies, such as progressive supranuclear palsy (PSP), corticobasal syndrome (CBS), and subtypes of frontotemporal dementia (FTD), as well as secondary tauopathies, such as Alzheimer's disease (AD). The development of tau therapeutics must reconcile with the structural complexity of the tau proteome, as well as an incomplete understanding of the role of tau in both physiology and disease. AREAS COVERED: This review offers a current perspective on tau biology, discusses key barriers to the development of effective tau-based therapeutics, and promotes the idea that pathogenic (as opposed to merely pathological) tau should be at the center of drug development efforts. EXPERT OPINION: An efficacious tau therapeutic will exhibit several primary features: 1) selectivity for pathogenic tau versus other tau species; 2) blood-brain barrier and cell membrane permeability, enabling access to intracellular tau in disease-relevant brain regions; and 3) minimal toxicity. Oligomeric tau is proposed as a major pathogenic form of tau and a compelling drug target in tauopathies.


Subject(s)
Alzheimer Disease , Supranuclear Palsy, Progressive , Tauopathies , Humans , Brain/metabolism , Supranuclear Palsy, Progressive/metabolism , Supranuclear Palsy, Progressive/pathology , tau Proteins/metabolism
17.
Alzheimers Dement ; 19(10): 4609-4618, 2023 10.
Article in English | MEDLINE | ID: mdl-36946611

ABSTRACT

INTRODUCTION: Secernin-1 (SCRN1) is a neuronal protein that co-localizes with neurofibrillary tangles in Alzheimer's disease (AD), but not with tau inclusions in corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), or Pick's disease. METHODS: We measured SCRN1 concentration in cerebrospinal fluid (CSF) using a novel mass spectrometric parallel reaction monitoring method in three clinical cohorts comprising patients with neurochemically characterized AD (n = 25) and controls (n = 28), clinically diagnosed Parkinson's disease (PD; n = 38), multiple system atrophy (MSA; n = 31), PSP (n = 20), CBD (n = 8), healthy controls (n = 37), and neuropathology-confirmed AD (n = 47). RESULTS: CSF SCRN1 was significantly increased in AD (P < 0.01, fold change = 1.4) compared to controls (receiver operating characteristic area under the curve = 0.78) but not in CBD, PSP, PD, or MSA. CSF SCRN1 positively correlated with CSF total tau (R = 0.78, P = 1.1 × 10-13 ), phosphorylated tau181 (R = 0.64, P = 3.2 × 10-8 ), and Braak stage and negatively correlated with Mini-Mental State Examination score. DISCUSSION: CSF SCRN1 is a candidate biomarker of AD, reflecting tau pathology. HIGHLIGHTS: We developed a parallel reaction monitoring assay to measure secernin-1 (SCRN1) in cerebrospinal fluid (CSF). CSF SCRN1 was increased in Alzheimer's disease compared to healthy controls. CSF SCRN1 remained unchanged in Parkinson's disease, multiple system atrophy, progressive supranuclear palsy, or corticobasal degeneration compared to controls. CSF SCRN1 correlated strongly with CSF phosphorylated tau and total tau. CSF SCRN1 increased across Braak stages and negatively correlated with Mini-Mental State Examination score.


Subject(s)
Alzheimer Disease , Nerve Tissue Proteins , tau Proteins , Humans , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Biomarkers/cerebrospinal fluid , Biomarkers/metabolism , Corticobasal Degeneration/cerebrospinal fluid , Corticobasal Degeneration/metabolism , Corticobasal Degeneration/pathology , Multiple System Atrophy/cerebrospinal fluid , Multiple System Atrophy/metabolism , Multiple System Atrophy/pathology , Nerve Tissue Proteins/cerebrospinal fluid , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Parkinson Disease/cerebrospinal fluid , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Supranuclear Palsy, Progressive/cerebrospinal fluid , Supranuclear Palsy, Progressive/genetics , Supranuclear Palsy, Progressive/metabolism , Supranuclear Palsy, Progressive/pathology , tau Proteins/cerebrospinal fluid , tau Proteins/metabolism
18.
J Neurochem ; 165(4): 563-586, 2023 05.
Article in English | MEDLINE | ID: mdl-36847488

ABSTRACT

Progressive supranuclear palsy (PSP) is a late-onset neurodegenerative disease defined pathologically by the presence of insoluble phosphorylated-Tau (p-Tau) in neurons and glia. Identifying co-aggregating proteins within p-Tau inclusions may reveal important insights into processes affected by the aggregation of Tau. We used a proteomic approach, which combines antibody-mediated biotinylation and mass spectrometry (MS) to identify proteins proximal to p-Tau in PSP. Using this proof-of-concept workflow for identifying interacting proteins of interest, we characterized proteins proximal to p-Tau in PSP cases, identifying >84% of previously identified interaction partners of Tau and known modifiers of Tau aggregation, while 19 novel proteins not previously found associated with Tau were identified. Furthermore, our data also identified confidently assigned phosphorylation sites that have been previously reported on p-Tau. Additionally, using ingenuity pathway analysis (IPA) and human RNA-seq datasets, we identified proteins previously associated with neurological disorders and pathways involved in protein degradation, stress responses, cytoskeletal dynamics, metabolism, and neurotransmission. Together, our study demonstrates the utility of biotinylation by antibody recognition (BAR) approach to answer a fundamental question to rapidly identify proteins in proximity to p-Tau from post-mortem tissue. The application of this workflow opens up the opportunity to identify novel protein targets to give us insight into the biological process at the onset and progression of tauopathies.


Subject(s)
Neurodegenerative Diseases , Supranuclear Palsy, Progressive , Tauopathies , Humans , tau Proteins/metabolism , Supranuclear Palsy, Progressive/metabolism , Proteolysis , Proteomics , Tauopathies/metabolism , Synaptic Transmission
19.
Handb Clin Neurol ; 193: 23-32, 2023.
Article in English | MEDLINE | ID: mdl-36803814

ABSTRACT

The toxic proteinopathy paradigm has defined neurodegenerative disorders for over a century. This gain-of-function (GOF) framework posited that proteins become toxic when turned into amyloids (pathology), predicting that lowering its levels would translate into clinical benefits. Genetic observations used to support a GOF framework are equally compatible with a loss-of-function (LOF) framework, as the soluble pool of proteins rendered unstable by these mutations (e.g., APP in Alzheimer's disease, SNCA in Parkinson's disease) aggregate, becoming depleted. In this review, we highlight misconceptions that have prevented LOF from gaining currency. Some of these misconceptions include no phenotype in knock-out animals (there is neurodegenerative phenotype in knock-out animals) and high levels of proteins in patients (patients have lower levels of the proteins involved in neurodegeneration than healthy age-matched controls). We also expose the internal contradictions within the GOF framework, namely that (1) pathology can have both pathogenic and protective roles; (2) the neuropathology gold standard for diagnosis can be present in normal individuals and absent in those affected; (3) oligomers are the toxic species even if they are ephemeral and decrease over time. We therefore advocate for a paradigm shift from proteinopathy (GOF) to proteinopenia (LOF) based on the universal depletion of soluble functional proteins in neurodegenerative diseases (low amyloid-ß 42 in Alzheimer's disease, low α-synuclein in Parkinson's disease, and low tau in progressive supranuclear palsy) and supported by the confluence of biologic, thermodynamic, and evolutionary principles with proteins having evolved to perform a function, not to become toxic, and where protein depletion is consequential. Such shift to a Proteinopenia paradigm is necessary to examining the safety and efficacy of protein replacement strategies instead of perpetuating a therapeutic paradigm with further antiprotein permutations.


Subject(s)
Alzheimer Disease , Parkinson Disease , Proteostasis Deficiencies , Supranuclear Palsy, Progressive , Animals , Humans , Parkinson Disease/pathology , Alzheimer Disease/pathology , Supranuclear Palsy, Progressive/metabolism
20.
J Neurol ; 270(5): 2451-2467, 2023 May.
Article in English | MEDLINE | ID: mdl-36633672

ABSTRACT

OBJECTIVES: To evaluate the difference of tau burden between patients with progressive supranuclear palsy (PSP) and healthy controls (HCs) or other neurodegenerative diseases using tau-positron emission tomography (PET) imaging. METHODS: A systematic search on PubMed, Embase, and Web of Science databases was performed for tau-PET studies in PSP patients, up to April 1, 2022. Standardized mean differences (SMDs) of tau tracer uptake were calculated using random-effects models. Subgroup analysis based on the type of tau tracers, meta-regression, and sensitivity analysis were conducted. RESULTS: Twenty-seven studies comprising 553 PSP, 626 HCs, and 406 other neurodegenerative diseases were included. Compared with HCs, PSP patients showed elevated tau binding in basal ganglia, midbrain, dentate nucleus, cerebellar white matter, and frontal lobe with decreasing SMD (SMD: 0.390-1.698). Compared with Parkinson's disease patients, increased tau binding was identified in the midbrain, basal ganglia, dentate nucleus, and frontal and parietal lobe in PSP patients with decreasing SMD (SMD: 0.503-1.853). PSP patients showed higher tau binding in the subthalamic nucleus (SMD = 1.351) and globus pallidus (SMD = 1.000), and lower binding in the cortex and parahippocampal gyrus than Alzheimer's disease patients (SMD: - 2.976 to - 1.018). PSP patients showed higher midbrain tau binding than multiple system atrophy patients (SMD = 1.269). CONCLUSION: Tau PET imaging indicates different topography of tau deposition between PSP patients and HCs or other neurodegenerative disorders. The affinity and selectivity of tracers for 4R-tau and the off-target binding of tracers should be considered when interpreting the results.


Subject(s)
Parkinson Disease , Supranuclear Palsy, Progressive , Humans , Supranuclear Palsy, Progressive/diagnostic imaging , Supranuclear Palsy, Progressive/metabolism , tau Proteins/metabolism , Basal Ganglia/diagnostic imaging , Parkinson Disease/metabolism , Positron-Emission Tomography/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...