Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 118(50)2021 12 14.
Article in English | MEDLINE | ID: mdl-34893539

ABSTRACT

There are currently no treatments that can slow the progression of neurodegenerative diseases, such as Alzheimer's disease (AD). There is, however, a growing body of evidence that activation of the M1 muscarinic acetylcholine receptor (M1-receptor) can not only restore memory loss in AD patients but in preclinical animal models can also slow neurodegenerative disease progression. The generation of an effective medicine targeting the M1-receptor has however been severely hampered by associated cholinergic adverse responses. By using genetically engineered mouse models that express a G protein-biased M1-receptor, we recently established that M1-receptor mediated adverse responses can be minimized by ensuring activating ligands maintain receptor phosphorylation/arrestin-dependent signaling. Here, we use these same genetic models in concert with murine prion disease, a terminal neurodegenerative disease showing key hallmarks of AD, to establish that phosphorylation/arrestin-dependent signaling delivers neuroprotection that both extends normal animal behavior and prolongs the life span of prion-diseased mice. Our data point to an important neuroprotective property inherent to the M1-receptor and indicate that next generation M1-receptor ligands designed to drive receptor phosphorylation/arrestin-dependent signaling would potentially show low adverse responses while delivering neuroprotection that will slow disease progression.


Subject(s)
Prion Diseases/metabolism , Prion Diseases/pathology , Receptor, Muscarinic M1/metabolism , Animals , Cells, Cultured , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Expression Regulation/physiology , Mice , Mice, Knockout , Neurons/metabolism , Prion Diseases/genetics , Receptor, Muscarinic M1/genetics , Signal Transduction
2.
Brain ; 143(8): 2576-2593, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32705145

ABSTRACT

The glymphatic system, that is aquaporin 4 (AQP4) facilitated exchange of CSF with interstitial fluid (ISF), may provide a clearance pathway for protein species such as amyloid-ß and tau, which accumulate in the brain in Alzheimer's disease. Further, tau protein transference via the extracellular space, the compartment that is cleared by the glymphatic pathway, allows for its neuron-to-neuron propagation, and the regional progression of tauopathy in the disorder. The glymphatic system therefore represents an exciting new target for Alzheimer's disease. Here we aim to understand the involvement of glymphatic CSF-ISF exchange in tau pathology. First, we demonstrate impaired CSF-ISF exchange and AQP4 polarization in a mouse model of tauopathy, suggesting that this clearance pathway may have the potential to exacerbate or even induce pathogenic accumulation of tau. Subsequently, we establish the central role of AQP4 in the glymphatic clearance of tau from the brain; showing marked impaired glymphatic CSF-ISF exchange and tau protein clearance using the novel AQP4 inhibitor, TGN-020. As such, we show that this system presents as a novel druggable target for the treatment of Alzheimer's disease, and possibly other neurodegenerative diseases alike.


Subject(s)
Alzheimer Disease/metabolism , Aquaporin 4/metabolism , Brain/metabolism , Glymphatic System/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Animals , Brain/pathology , Cerebrospinal Fluid/metabolism , Disease Models, Animal , Extracellular Fluid/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
3.
J Neurosci ; 36(3): 762-72, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26791207

ABSTRACT

The interneuronal propagation of aggregated tau is believed to play an important role in the pathogenesis of human tauopathies. It requires the uptake of seed-competent tau into cells, seeding of soluble tau in recipient neurons and release of seeded tau into the extracellular space to complete the cycle. At present, it is not known which tau species are seed-competent. Here, we have dissected the molecular characteristics of seed-competent tau species from the TgP301S tau mouse model using various biochemical techniques and assessed their seeding ability in cell and animal models. We found that sucrose gradient fractions from brain lysates seeded cellular tau aggregation only when large (>10 mer) aggregated, hyperphosphorylated (AT8- and AT100-positive) and nitrated tau was present. In contrast, there was no detectable seeding by fractions containing small, oligomeric (<6 mer) tau. Immunodepletion of the large aggregated AT8-positive tau strongly reduced seeding; moreover, fractions containing these species initiated the formation and spreading of filamentous tau pathology in vivo, whereas fractions containing tau monomers and small oligomeric assemblies did not. By electron microscopy, seed-competent sucrose gradient fractions contained aggregated tau species ranging from ring-like structures to small filaments. Together, these findings indicate that a range of filamentous tau aggregates are the major species that underlie the spreading of tau pathology in the P301S transgenic model. Significance statement: The spread of tau pathology from neuron to neuron is postulated to account for, or at least to contribute to, the overall propagation of tau pathology during the development of human tauopathies including Alzheimer's disease. It is therefore important to characterize the native tau species responsible for this process of seeding and pathology spreading. Here, we use several biochemical techniques to dissect the molecular characteristics of native tau protein conformers from TgP301S tau mice and show that seed-competent tau species comprise small fibrils capable of seeding tau pathology in cell and animal models. Characterization of seed-competent tau gives insight into disease mechanisms and therapeutic interventions.


Subject(s)
Amyloid/genetics , Brain , Neurofibrillary Tangles/genetics , Tauopathies/genetics , tau Proteins/genetics , Animals , Brain/pathology , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurofibrillary Tangles/pathology , Tauopathies/pathology
4.
Mov Disord ; 30(7): 960-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25854893

ABSTRACT

BACKGROUND: Glial cytoplasmic inclusions containing α-synuclein are the pathological hallmark of multiple system atrophy (MSA). Minimal change (MC-MSA) is an unusual MSA subtype with neuronal loss largely restricted to the substantia nigra and locus coeruleus. METHODS: Immunohistochemistry on selected brain regions and semiquantitative assessment were performed on six MC-MSA and eight MSA control cases. RESULTS: More neuronal cytoplasmic inclusions were seen in the caudate and substantia nigra in MC-MSA than in MSA controls (P = 0.002), without any statistical difference in glial cytoplasmic inclusion load in any region. Severe glial cytoplasmic inclusion load was found in the ventrolateral medulla (P = 1.0) and nucleus raphe obscurus (P = 0.4) in both groups. When compared with MSA controls, the three MC-MSA cases who had died of sudden unexpected death had an earlier age of onset (mean: 38 vs. 57.6 y, P = 0.02), a numerically shorter disease duration (mean: 5.3 vs. 8 y, P = 0.2) and a more rapid clinical progression with most of the clinical milestones reached within 3 y of presentation, suggesting an aggressive variant of MSA. Another three MC-MSA cases, who had died of unrelated concurrent diseases, had an age of onset (mean: 57.7 y) and temporal course similar to controls, had less severe neuronal loss and gliosis in the medial and dorsolateral substantia nigra subregions (P < 0.05) than in MSA controls, and could be considered as a unique group with interrupted pathological progression. Significant respiratory dysfunction and early orthostatic hypotension were observed in all MC-MSA cases. CONCLUSIONS: Our findings could suggest that α-synuclein-associated oligodendroglial pathology may lead to neuronal dysfunction sufficient to cause clinical symptoms before overt neuronal loss in MSA. © 2015 International Parkinson and Movement Disorder Society.


Subject(s)
Brain/pathology , Inclusion Bodies/pathology , Multiple System Atrophy/classification , Multiple System Atrophy/pathology , Tissue Banks , alpha-Synuclein/metabolism , Adult , Age of Onset , Aged , Aged, 80 and over , Brain/metabolism , Humans , Inclusion Bodies/metabolism , Male , Middle Aged , Multiple System Atrophy/metabolism
5.
Acta Neuropathol ; 127(5): 667-83, 2014 May.
Article in English | MEDLINE | ID: mdl-24531916

ABSTRACT

Intracellular inclusions composed of hyperphosphorylated filamentous tau are a hallmark of Alzheimer's disease, progressive supranuclear palsy, Pick's disease and other sporadic neurodegenerative tauopathies. Recent in vitro and in vivo studies have shown that tau aggregates do not only seed further tau aggregation within neurons, but can also spread to neighbouring cells and functionally connected brain regions. This process is referred to as 'tau propagation' and may explain the stereotypic progression of tau pathology in the brains of Alzheimer's disease patients. Here, we describe a novel in vivo model of tau propagation using human P301S tau transgenic mice infused unilaterally with brain extract containing tau aggregates. Infusion-related neurofibrillary tangle pathology was first observed 2 weeks post-infusion and increased in a stereotypic, time-dependent manner. Contralateral and anterior/posterior spread of tau pathology was also evident in nuclei with strong synaptic connections (efferent and afferent) to the site of infusion, indicating that spread was dependent on synaptic connectivity rather than spatial proximity. This notion was further supported by infusion-related tau pathology in white matter tracts that interconnect these regions. The rapid and robust propagation of tau pathology in this model will be valuable for both basic research and the drug discovery process.


Subject(s)
Brain/pathology , Neurofibrillary Tangles/pathology , Tauopathies/pathology , tau Proteins/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Disease Progression , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immunohistochemistry , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/metabolism , Neural Pathways/pathology , Neurofibrillary Tangles/metabolism , Random Allocation , Synapses/metabolism , Synapses/pathology , Tauopathies/metabolism , Time Factors , White Matter/metabolism , White Matter/pathology , tau Proteins/genetics
6.
Alzheimers Res Ther ; 16(1): 71, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38576025

ABSTRACT

BACKGROUND: The aggregation and spread of misfolded amyloid structured proteins, such as tau and α-synuclein, are key pathological features associated with neurodegenerative disorders, including Alzheimer's and Parkinson's disease. These proteins possess a prion-like property, enabling their transmission from cell to cell leading to propagation throughout the central and peripheral nervous systems. While the mechanisms underlying their intracellular spread are still being elucidated, targeting the extracellular space has emerged as a potential therapeutic approach. The glymphatic system, a brain-wide pathway responsible for clearing extracellular metabolic waste from the central nervous system, has gained attention as a promising target for removing these toxic proteins. METHODS: In this study, we investigated the impact of long-term modulation of glymphatic function on tau aggregation and spread by chronically treating a mouse model of tau propagation with a pharmacological inhibitor of AQP4, TGN-020. Thy1-hTau.P301S mice were intracerebrally inoculated with tau into the hippocampus and overlying cortex, and subsequently treated with TGN-020 (3 doses/week, 50 mg/kg TGN-020, i.p.) for 10-weeks. During this time, animal memory was studied using cognitive behavioural tasks, and structural MR images were acquired of the brain in vivo prior to brain extraction for immunohistochemical characterisation. RESULTS: Our findings demonstrate increased tau aggregation in the brain and transhemispheric propagation in the hippocampus following the inhibition of glymphatic clearance. Moreover, disruption of the glymphatic system aggravated recognition memory in tau inoculated mice and exacerbated regional changes in brain volume detected in the model. When initiation of drug treatment was delayed for several weeks post-inoculation, the alterations were attenuated. CONCLUSIONS: These results indicate that by modulating AQP4 function and, consequently, glymphatic clearance, it is possible to modify the propagation and pathological impact of tau in the brain, particularly during the initial stages of the disease. These findings highlight the critical role of the glymphatic system in preserving healthy brain homeostasis and offer valuable insights into the therapeutic implications of targeting this system for managing neurodegenerative diseases characterized by protein aggregation and spread.


Subject(s)
Alzheimer Disease , Glymphatic System , Niacinamide/analogs & derivatives , Thiadiazoles , Mice , Animals , Alzheimer Disease/pathology , Brain/metabolism , Glymphatic System/metabolism , tau Proteins/metabolism
7.
Acta Neuropathol ; 125(5): 741-52, 2013 May.
Article in English | MEDLINE | ID: mdl-23371366

ABSTRACT

Corticobasal degeneration (CBD) is a disorder affecting cognition and movement due to a progressive neurodegeneration associated with distinctive neuropathologic features, including abnormal phosphorylated tau protein in neurons and glia in cortex, basal ganglia, diencephalon, and brainstem, as well as ballooned neurons and astrocytic plaques. We identified three cases of CBD with olivopontocerebellar atrophy (CBD-OPCA) that did not have α-synuclein-positive glial cytoplasmic inclusions of multiple system atrophy (MSA). Two patients had clinical features suggestive of progressive supranuclear palsy (PSP), and the third case had cerebellar ataxia thought to be due to idiopathic OPCA. Neuropathologic features of CBD-OPCA are compared to typical CBD, as well as MSA and PSP. CBD-OPCA and MSA had marked neuronal loss in pontine nuclei, inferior olivary nucleus, and Purkinje cell layer. Neuronal loss and grumose degeneration in the cerebellar dentate nucleus were comparable in CBD-OPCA and PSP. Image analysis of tau pathology showed greater infratentorial tau burden, especially in pontine base, in CBD-OPCA compared with typical CBD. In addition, CBD-OPCA had TDP-43 immunoreactive neuronal and glial cytoplasmic inclusions and threads throughout the basal ganglia and in olivopontocerebellar system. CBD-OPCA met neuropathologic research diagnostic criteria for CBD and shared tau biochemical characteristics with typical CBD. These results suggest that CBD-OPCA is a distinct clinicopathologic variant of CBD with olivopontocerebellar TDP-43 pathology.


Subject(s)
DNA-Binding Proteins/physiology , Olivopontocerebellar Atrophies/metabolism , Olivopontocerebellar Atrophies/pathology , Aged , Cerebellar Ataxia/etiology , Cerebellar Ataxia/metabolism , Cerebellar Ataxia/pathology , Female , Humans , Male , Nerve Degeneration/etiology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Olivopontocerebellar Atrophies/etiology , Supranuclear Palsy, Progressive/etiology , Supranuclear Palsy, Progressive/metabolism , Supranuclear Palsy, Progressive/pathology
8.
Acta Neuropathol ; 126(4): 537-544, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23995422

ABSTRACT

Recent studies have highlighted a group of 4-repeat (4R) tauopathies that are characterised neuropathologically by widespread, globular glial inclusions (GGIs). Tau immunohistochemistry reveals 4R immunoreactive globular oligodendroglial and astrocytic inclusions and the latter are predominantly negative for Gallyas silver staining. These cases are associated with a range of clinical presentations, which correlate with the severity and distribution of underlying tau pathology and neurodegeneration. Their heterogeneous clinicopathological features combined with their rarity and under-recognition have led to cases characterised by GGIs being described in the literature using various and redundant terminologies. In this report, a group of neuropathologists form a consensus on the terminology and classification of cases with GGIs. After studying microscopic images from previously reported cases with suspected GGIs (n = 22), this panel of neuropathologists with extensive experience in the diagnosis of neurodegenerative diseases and a documented record of previous experience with at least one case with GGIs, agreed that (1) GGIs were present in all the cases reviewed; (2) the morphology of globular astrocytic inclusions was different to tufted astrocytes and finally that (3) the cases represented a number of different neuropathological subtypes. They also agreed that the different morphological subtypes are likely to be part of a spectrum of a distinct disease entity, for which they recommend that the overarching term globular glial tauopathy (GGT) should be used. Type I cases typically present with frontotemporal dementia, which correlates with the fronto-temporal distribution of pathology. Type II cases are characterised by pyramidal features reflecting motor cortex involvement and corticospinal tract degeneration. Type III cases can present with a combination of frontotemporal dementia and motor neuron disease with fronto-temporal cortex, motor cortex and corticospinal tract being severely affected. Extrapyramidal features can be present in Type II and III cases and significant degeneration of the white matter is a feature of all GGT subtypes. Improved detection and classification will be necessary for the establishment of neuropathological and clinical diagnostic research criteria in the future.


Subject(s)
Neuroglia/pathology , Tauopathies/pathology , Astrocytes/pathology , Consensus , Humans , Immunohistochemistry , Inclusion Bodies/pathology , Neurodegenerative Diseases/pathology , Silver Staining , Tauopathies/classification , Terminology as Topic , tau Proteins/chemistry
9.
Mov Disord ; 27(9): 1186-90, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22806758

ABSTRACT

BACKGROUND: Multiple system atrophy (MSA) is a neurodegenerative disease leading to severe physical impairment, with a disease duration from onset to death of 6-9 years. METHODS: The clinical and neuropathological features of 4 MSA cases with disease duration of 15 years or more were analyzed. RESULTS: All patients presented with parkinsonism and had a mean latency of 11 years before the development of dysautonomia. Mean duration from onset of first symptoms to anterocollis, inspiratory stridor, and dysphagia was 9 years. Despite the limited levodopa response, all patients developed levodopa-induced dyskinesia. CONCLUSIONS: Late appearance of dysautonomia is a favorable prognostic factor in MSA-P. Greater awareness of this uncommon "benign" subgroup of MSA will improve diagnostic accuracy and help to more accurately inform treatment options.


Subject(s)
Multiple System Atrophy/pathology , Activities of Daily Living , Adult , Age of Onset , Aged , Autopsy , Brain/pathology , Cognition Disorders/complications , Cognition Disorders/psychology , Diagnostic Errors , Disease Progression , Female , Hallucinations/etiology , Hallucinations/psychology , Humans , Immunohistochemistry , Male , Middle Aged , Multiple System Atrophy/diagnosis , Neuropsychological Tests
10.
Brain ; 134(Pt 9): 2548-64, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21752791

ABSTRACT

Neuronal intermediate filament inclusion disease and atypical frontotemporal lobar degeneration are rare diseases characterized by ubiquitin-positive inclusions lacking transactive response DNA-binding protein-43 and tau. Recently, mutations in the fused in sarcoma gene have been shown to cause familial amyotrophic lateral sclerosis and fused in sarcoma-positive neuronal inclusions have subsequently been demonstrated in neuronal intermediate filament inclusion disease and atypical frontotemporal lobar degeneration with ubiquitinated inclusions. Here we provide clinical, imaging, morphological findings, as well as genetic and biochemical data in 14 fused in sarcoma proteinopathy cases. In this cohort, the age of onset was variable but included cases of young-onset disease. Patients with atypical frontotemporal lobar degeneration with ubiquitinated inclusions all presented with behavioural variant frontotemporal dementia, while the clinical presentation in neuronal intermediate filament inclusion disease was more heterogeneous, including cases with motor neuron disease and extrapyramidal syndromes. Neuroimaging revealed atrophy of the frontal and anterior temporal lobes as well as the caudate in the cases with atypical frontotemporal lobar degeneration with ubiquitinated inclusions, but was more heterogeneous in the cases with neuronal intermediate filament inclusion disease, often being normal to visual inspection early on in the disease. The distribution and severity of fused in sarcoma-positive neuronal cytoplasmic inclusions, neuronal intranuclear inclusions and neurites were recorded and fused in sarcoma was biochemically analysed in both subgroups. Fused in sarcoma-positive neuronal cytoplasmic and intranuclear inclusions were found in the hippocampal granule cell layer in variable numbers. Cortical fused in sarcoma-positive neuronal cytoplasmic inclusions were often 'Pick body-like' in neuronal intermediate filament inclusion disease, and annular and crescent-shaped inclusions were seen in both conditions. Motor neurons contained variable numbers of compact, granular or skein-like cytoplasmic inclusions in all fused in sarcoma-positive cases in which brainstem and spinal cord motor neurons were available for study (five and four cases, respectively). No fused in sarcoma mutations were found in any cases. Biochemically, two major fused in sarcoma species were found and shown to be more insoluble in the atypical frontotemporal lobar degeneration with ubiquitinated inclusions subgroup compared with neuronal intermediate filament inclusion disease. There is considerable overlap and also significant differences in fused in sarcoma-positive pathology between the two subgroups, suggesting they may represent a spectrum of the same disease. The co-existence of fused in sarcoma-positive inclusions in both motor neurons and extramotor cerebral structures is a characteristic finding in sporadic fused in sarcoma proteinopathies, indicating a multisystem disorder.


Subject(s)
Frontotemporal Lobar Degeneration , Inclusion Bodies/pathology , Intermediate Filaments/pathology , Neurons/pathology , RNA-Binding Protein FUS/metabolism , Adult , Age of Onset , Aged , Brain/pathology , Cohort Studies , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/pathology , Frontotemporal Lobar Degeneration/physiopathology , Humans , Intermediate Filaments/metabolism , Male , Middle Aged , Neurons/cytology , RNA-Binding Protein FUS/chemistry , RNA-Binding Protein FUS/genetics , Ubiquitin/metabolism , tau Proteins/genetics , tau Proteins/metabolism
11.
Am J Pathol ; 177(1): 311-24, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20522652

ABSTRACT

Progranulin (PGRN) is involved in wound repair, inflammation, and tumor formation, but its function in the central nervous system is unknown. Roles in development, sexual differentiation, and long-term neuronal survival have been suggested. Mutations in the GRN gene resulting in partial loss of the encoded PGRN protein cause frontotemporal lobar degeneration with ubiquitin immunoreactive inclusions. We sought to understand the neuropathological consequences of loss of PGRN function throughout the lifespan of GRN-deficient ((-/+) and (-/-)) mice. An aged series of GRN-deficient and wild-type mice were compared by histology, immunohistochemistry, and electron microscopy. Although GRN-deficient mice were viable, GRN(-/-) mice were produced at lower than predicted frequency. Neuropathologically, GRN(-/+) were indistinguishable from controls; however, GRN(-/-) mice developed age-associated, abnormal intraneuronal ubiquitin-positive autofluorescent lipofuscin. Lipofuscin was noted in aged GRN(+/+) mice at levels comparable with those of young GRN(-/-) mice. GRN(-/-) mice developed microgliosis, astrogliosis, and tissue vacuolation, with focal neuronal loss and severe gliosis apparent in the oldest GRN(-/-) mice. Although no overt frontotemporal lobar degeneration with ubiquitin immunoreactive inclusions type- or TAR DNA binding protein-43-positive lesions were observed, robust lipofuscinosis and ubiquitination in GRN(-/-) mice is strikingly similar to changes associated with aging and cellular decline in humans and animal models. Our data suggests that PGRN plays a key role in maintaining neuronal function during aging and supports the notion that PGRN is a trophic factor essential for long-term neuronal survival.


Subject(s)
Aging/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Mice, Knockout , Neuronal Ceroid-Lipofuscinoses/metabolism , Animals , Brain/cytology , Brain/metabolism , Brain/pathology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/pathology , Neurons/cytology , Neurons/metabolism , Progranulins , Ubiquitin/metabolism , Ubiquitination
12.
Acta Neuropathol ; 122(4): 415-28, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21773886

ABSTRACT

A number of recent studies have described cases with tau-positive globular oligodendroglial inclusions (GOIs) and such cases have overlapping pathological features with progressive supranuclear palsy (PSP), but present with clinical features of motor neuron disease (MND) and/or frontotemporal dementia (FTD). These two clinical phenotypes have been published independently and as a result, have come to be considered as distinct disease entities. We describe the clinicopathological and biochemical features of two cases with GOIs: one with clinical symptoms suggestive of MND and the other with FTD. Histological changes in our two cases were consistent with their clinical symptoms; the MND case had severe neurodegeneration in the primary motor cortex and corticospinal tract, whereas the FTD case had severe involvement of the frontotemporal cortices and associated white matter. Immunohistochemistry in both cases revealed significant 4-repeat (4R) tau pathology primarily in the form of GOIs, but also in astrocytes and neurons. Astrocytic tau pathology was morphologically similar to that seen in PSP, but in contrast was consistently negative for Gallyas silver staining. Tau-specific western blotting revealed 68, 64 and 35 kDa bands, showing further overlap with PSP. The underlying neuropathological features of these two cases were similar, with the major difference relating to the regional distribution of pathology and resulting clinical symptoms and signs. The globular nature of glial inclusions and the non-fibrillar properties of tau in astrocytes are characteristic features that allow them to be distinguished from PSP and other tauopathies. We, therefore, propose the term globular glial tauopathy as an encompassing term to classify this emerging class of 4R tauopathy.


Subject(s)
Frontotemporal Dementia/diagnosis , Frontotemporal Dementia/pathology , Motor Neuron Disease/diagnosis , Motor Neuron Disease/pathology , Oligodendroglia/pathology , Tauopathies/diagnosis , Tauopathies/pathology , Trinucleotide Repeat Expansion/genetics , Aged , Diagnosis, Differential , Fatal Outcome , Female , Frontotemporal Dementia/genetics , Humans , Male , Motor Neuron Disease/genetics , Tauopathies/genetics
13.
Sci Rep ; 11(1): 10309, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33986302

ABSTRACT

Intracellular tau inclusions are a pathological hallmark of Alzheimer's disease, progressive supranuclear palsy, corticobasal degeneration and other sporadic neurodegenerative tauopathies. Recent in vitro and in vivo studies have shown that tau aggregates may spread to neighbouring cells and functionally connected brain regions, where they can seed further tau aggregation. This process is referred to as tau propagation. Here we describe an ex vivo system using organotypic hippocampal slice cultures (OHCs) which recapitulates aspects of this phenomenon. OHCs are explants of hippocampal tissue which may be maintained in culture for months. They maintain their synaptic connections and multicellular 3D architecture whilst also permitting direct control of the environment and direct access for various analysis types. We inoculated OHCs prepared from P301S mouse pups with brain homogenate from terminally ill P301S mice and then examined the slices for viability and the production and localization of insoluble phosphorylated tau. We show that following seeding, phosphorylated insoluble tau accumulate in a time and concentration dependent manner within OHCs. Furthermore, we show the ability of the conformation dependent anti-tau antibody, MC1, to compromise tau accrual in OHCs, thus showcasing the potential of this therapeutic approach and the utility of OHCs as an ex vivo model system for assessing such therapeutics.


Subject(s)
Hippocampus/pathology , Tauopathies/pathology , Alzheimer Disease/pathology , Animals , Hippocampus/metabolism , In Vitro Techniques , Mice , Mice, Transgenic , Phosphorylation
14.
Cell Rep ; 37(7): 110022, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34788620

ABSTRACT

Alternative splicing is a post-transcriptional regulatory mechanism producing distinct mRNA molecules from a single pre-mRNA with a prominent role in the development and function of the central nervous system. We used long-read isoform sequencing to generate full-length transcript sequences in the human and mouse cortex. We identify novel transcripts not present in existing genome annotations, including transcripts mapping to putative novel (unannotated) genes and fusion transcripts incorporating exons from multiple genes. Global patterns of transcript diversity are similar between human and mouse cortex, although certain genes are characterized by striking differences between species. We also identify developmental changes in alternative splicing, with differential transcript usage between human fetal and adult cortex. Our data confirm the importance of alternative splicing in the cortex, dramatically increasing transcriptional diversity and representing an important mechanism underpinning gene regulation in the brain. We provide transcript-level data for human and mouse cortex as a resource to the scientific community.


Subject(s)
Cerebral Cortex/metabolism , Protein Isoforms/genetics , Transcriptome/genetics , Alternative Splicing/genetics , Animals , Brain/metabolism , Cerebral Cortex/physiology , Exons/genetics , Gene Expression/genetics , Gene Expression Profiling/methods , Genome , High-Throughput Nucleotide Sequencing/methods , Humans , Mice , Protein Isoforms/metabolism , RNA Precursors/genetics , RNA Splice Sites/genetics , RNA, Messenger/genetics , Sequence Analysis, RNA/methods
15.
Hum Mol Genet ; 17(23): 3631-42, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18723524

ABSTRACT

Loss-of-function mutations in progranulin (GRN) cause ubiquitin- and TAR DNA-binding protein 43 (TDP-43)-positive frontotemporal dementia (FTLD-U), a progressive neurodegenerative disease affecting approximately 10% of early-onset dementia patients. Here we expand the role of GRN in FTLD-U and demonstrate that a common genetic variant (rs5848), located in the 3'-untranslated region (UTR) of GRN in a binding-site for miR-659, is a major susceptibility factor for FTLD-U. In a series of pathologically confirmed FTLD-U patients without GRN mutations, we show that carriers homozygous for the T-allele of rs5848 have a 3.2-fold increased risk to develop FTLD-U compared with homozygous C-allele carriers (95% CI: 1.50-6.73). We further demonstrate that miR-659 can regulate GRN expression in vitro, with miR-659 binding more efficiently to the high risk T-allele of rs5848 resulting in augmented translational inhibition of GRN. A significant reduction in GRN protein was observed in homozygous T-allele carriers in vivo, through biochemical and immunohistochemical methods, mimicking the effect of heterozygous loss-of-function GRN mutations. In support of these findings, the neuropathology of homozygous rs5848 T-allele carriers frequently resembled the pathological FTLD-U subtype of GRN mutation carriers. We suggest that the expression of GRN is regulated by miRNAs and that common genetic variability in a miRNA binding-site can significantly increase the risk for FTLD-U. Translational regulation by miRNAs may represent a common mechanism underlying complex neurodegenerative disorders.


Subject(s)
DNA-Binding Proteins/metabolism , Dementia/genetics , Genetic Variation , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/genetics , MicroRNAs/metabolism , Aged , Base Sequence , Binding Sites , Brain/metabolism , Dementia/metabolism , Female , Gene Expression Regulation , Genotype , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , MicroRNAs/chemistry , MicroRNAs/genetics , Middle Aged , Molecular Sequence Data , Mutation , Progranulins , Protein Biosynthesis
16.
Curr Opin Neurol ; 23(4): 394-400, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20610990

ABSTRACT

PURPOSE OF REVIEW: Neurodegenerative tauopathies, of which progressive supranuclear palsy (PSP) is one of the most common, are clinically heterogeneous, reflecting differences in distribution and biochemical composition of tau pathology. This review highlights the range of clinical and pathologic presentations of PSP and its variants. RECENT FINDINGS: Progressive supranuclear palsy is a 4R tauopathy with neuronal and glial tau-immunoreactive lesions in neuroanatomically specific nuclei in the basal ganglia, diencephalon, brainstem and cerebellum, with restricted involvement of the neocortex. Hierarchical cluster analyses of clinical and pathologic features of PSP indicate that there are distinct clinicopathologic variants of PSP. In variants of PSP presenting with focal cortical syndromes, such as frontotemporal dementia, corticobasal syndrome and apraxia of speech, there is greater cortical pathology than in typical PSP. In variants of PSP presenting with levodopa-responsive Parkinsonism, as well as pure akinesia and gait failure, there is less cortical pathology and more severe degeneration in the cardinal nuclei - globus pallidus, subthalamic nucleus and substantia nigra - than in typical PSP. SUMMARY: Clinical variants in PSP reflect varying anatomical distribution of tau pathology, but they share histopathologic, biochemical and genetic features with typical PSP. The basis for anatomical selective vulnerability in PSP and its variants remains to be determined.


Subject(s)
Brain/pathology , Supranuclear Palsy, Progressive/pathology , Tauopathies/pathology , Brain/metabolism , Brain/physiopathology , Humans , Neurons/metabolism , Neurons/pathology , Phenotype , Supranuclear Palsy, Progressive/metabolism , Supranuclear Palsy, Progressive/physiopathology , Tauopathies/metabolism , Tauopathies/physiopathology , tau Proteins/physiology
17.
Mov Disord ; 25(5): 638-42, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20213825

ABSTRACT

There is growing awareness of impulsive-compulsive spectrum behaviors (ICBs) in patients with Parkinson's disease (PD) treated with dopamine replacement therapy (DRT). These include pathological gambling, hypersexuality, compulsive shopping, binge eating, punding and compulsive use of DRT, or dopamine dysregulation syndrome. In PD, difficulties exist in separating the effects of DRT from the underlying disease process and aberrant dopaminergic systems in determining the aetiology of ICBs. Recent reports of ICBs associated with dopamine agonist use for conditions other than PD may suggest a significant etiological role for these medications, but currently published cases thus far lack pathological confirmation of diagnoses. We present three cases of pathologically confirmed progressive supranuclear palsy who developed ICBs in association with dopamine agonist use. Pathological comparisons between these three cases and other case series of progressive supranuclear palsy are made.


Subject(s)
Disruptive, Impulse Control, and Conduct Disorders/etiology , Supranuclear Palsy, Progressive/complications , Supranuclear Palsy, Progressive/pathology , Aged , Dopamine/metabolism , Dopamine Agents/therapeutic use , Female , Humans , Male , Supranuclear Palsy, Progressive/drug therapy
18.
Cell Rep ; 30(6): 2040-2054.e5, 2020 02 11.
Article in English | MEDLINE | ID: mdl-32049030

ABSTRACT

Alzheimer's disease (AD) is associated with the intracellular aggregation of hyperphosphorylated tau and the accumulation of ß-amyloid in the neocortex. We use transgenic mice harboring human tau (rTg4510) and amyloid precursor protein (J20) mutations to investigate transcriptional changes associated with the progression of tau and amyloid pathology. rTg4510 mice are characterized by widespread transcriptional differences in the entorhinal cortex with changes paralleling neuropathological burden across multiple brain regions. Differentially expressed transcripts overlap with genes identified in genetic studies of familial and sporadic AD. Systems-level analyses identify discrete co-expression networks associated with the progressive accumulation of tau that are enriched for genes and pathways previously implicated in AD pathology and overlap with co-expression networks identified in human AD cortex. Our data provide further evidence for an immune-response component in the accumulation of tau and reveal molecular pathways associated with the progression of AD neuropathology.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/adverse effects , tau Proteins/adverse effects , Animals , Disease Models, Animal , Disease Progression , Humans , Mice , Mice, Transgenic
19.
Ann Neurol ; 63(2): 204-12, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17894374

ABSTRACT

OBJECTIVE: To test the hypothesis that beta-amyloid (Abeta) burden is associated with rates of brain atrophy. METHODS: Forty-five subjects who had been prospectively studied, died, and had an autopsy diagnosis of low, intermediate, or high probability of Alzheimer's disease who had two volumetric head magnetic resonance imaging scans were identified. Compact and total (compact + diffuse) Abeta burden was measured using a computerized image analyzer with software program to detect the proportion of gray matter occupied by Abeta. Visual ratings of Abeta burden were also performed. The boundary shift integral was used to calculate change over time in whole-brain and ventricular volume. All boundary shift integral results were annualized by adjusting for scan interval. Demographics, cognitive measures, clinical diagnoses, apolipoprotein E genotype, neurofibrillary tangle (NFT) pathology, and vascular lesion burden were determined. RESULTS: There was no correlation between compact or total Abeta burden, or visual Abeta ratings, and rates of brain loss or ventricular expansion in all subjects. However, significant correlations were observed between rates of brain loss and age, Braak NFT stage, and change over time in cognitive measures. These features also correlated with rates of ventricular expansion. The rates of brain loss and ventricular expansion were greater in demented compared with nondemented subjects. INTERPRETATION: These findings suggest that rate of brain volume loss is not determined by the amount of insoluble Abeta in the gray matter.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Atrophy/pathology , Brain/pathology , Plaque, Amyloid/pathology , Age of Onset , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Amyloid beta-Peptides/analysis , Apolipoproteins E/genetics , Atrophy/etiology , Atrophy/physiopathology , Autopsy , Brain/metabolism , Brain/physiopathology , Disease Progression , Female , Genotype , Humans , Lateral Ventricles/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Plaque, Amyloid/metabolism , Predictive Value of Tests , Prospective Studies , Severity of Illness Index , Time Factors
20.
Brain ; 131(Pt 2): 460-72, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18158316

ABSTRACT

Pallido-nigro-luysial atrophy (PNLA) is a rare disorder that in many cases has histopathological features similar to progressive supranuclear palsy (PSP). In a pathological series of over 400 cases of PSP, eight cases were noted to have features similar to those described in PNLA, including severe atrophy and neuronal loss in the globus pallidus, substantia nigra and subthalamic nucleus, in addition to many axonal spheroids in the globus pallidus and substantia nigra. These eight cases of PSP-PNLA were compared to 11 typical PSP cases with quantitative neuropathologic indices and assessment of demographics, clinical features and the timing of clinical features. PSP-PNLA cases were younger, had longer disease duration and more often were not initially diagnosed with PSP; in the end, they did not differ from PSP with respect to any major clinical feature. The clinical course of PSP-PNLA, however, was different, with earlier gait abnormalities and difficulty with handwriting, but later falls, rigidity and dysphagia than PSP. Pathologically, the same types of lesions were detected in both PSP and PSP-PNLA, but there were differences in the distribution and density of tau-pathology, with less tau-pathology in motor cortex, striatum, pontine nuclei and cerebellum in PSP-PNLA. These clinical and pathological findings suggest that PSP-PNLA should be considered a variant of PSP.


Subject(s)
Axons/pathology , Brain/pathology , Supranuclear Palsy, Progressive/diagnosis , Aged , Atrophy/complications , Atrophy/diagnosis , Atrophy/pathology , Cognition Disorders/etiology , Disease Progression , Female , Gait Disorders, Neurologic/etiology , Globus Pallidus/pathology , Humans , Male , Middle Aged , Severity of Illness Index , Substantia Nigra/pathology , Subthalamic Nucleus/pathology , Supranuclear Palsy, Progressive/complications , Supranuclear Palsy, Progressive/metabolism , Supranuclear Palsy, Progressive/pathology , Vision Disorders/etiology , tau Proteins/analysis
SELECTION OF CITATIONS
SEARCH DETAIL