Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
J Neurosci ; 41(17): 3917-3931, 2021 04 28.
Article in English | MEDLINE | ID: mdl-33731446

ABSTRACT

Tau deposition begins in the medial temporal lobe (MTL) in aging and Alzheimer's disease (AD), and MTL neural dysfunction is commonly observed in these groups. However, the association between tau and MTL neural activity has not been fully characterized. We investigated the effects of tau on repetition suppression, the reduction of activity for repeated stimulus presentations compared to novel stimuli. We used task-based functional MRI (fMRI) to assess MTL subregional activity in 21 young adults (YA) and 45 cognitively normal human older adults (OA; total sample: 37 females, 29 males). AD pathology was measured with position emission tomography (PET), using 18F-Flortaucipir for tau and 11C-Pittsburgh compound B (PiB) for amyloid-ß (Aß). The MTL was segmented into six subregions using high-resolution structural images. We compared the effects of low tau pathology, restricted to entorhinal cortex and hippocampus (Tau- OA), to high tau pathology, also occurring in temporal and limbic regions (Tau+ OA). Low levels of tau (Tau- OA vs YA) were associated with reduced repetition suppression activity specifically in anterolateral entorhinal cortex (alEC) and hippocampus, the first regions to accumulate tau. High tau pathology (Tau+ vs Tau- OA) was associated with widespread reductions in repetition suppression across MTL. Further analyses indicated that reduced repetition suppression was driven by hyperactivity to repeated stimuli, rather than decreased activity to novel stimuli. Increased activation was associated with entorhinal tau, but not Aß. These findings reveal a link between tau deposition and neural dysfunction in MTL, in which tau-related hyperactivity prevents deactivation to repeated stimuli, leading to reduced repetition suppression.SIGNIFICANCE STATEMENT Abnormal neural activity occurs in the medial temporal lobe (MTL) in aging and Alzheimer's disease (AD). Because tau pathology first deposits in the MTL in aging, this altered activity may be due to local tau pathology, and distinct MTL subregions may be differentially vulnerable. We demonstrate that in older adults (OAs) with low tau pathology, there are focal alterations in activity in MTL subregions that first develop tau pathology, while OAs with high tau pathology have aberrant activity throughout MTL. Tau was associated with hyperactivity to repeated stimulus presentations, leading to reduced repetition suppression, the discrimination between novel and repeated stimuli. Our data suggest that tau deposition is related to abnormal activity in MTL before the onset of cognitive decline.


Subject(s)
Aging/physiology , Temporal Lobe/physiology , tau Proteins/genetics , Aged , Aged, 80 and over , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Entorhinal Cortex/diagnostic imaging , Female , Hippocampus/diagnostic imaging , Humans , Magnetic Resonance Imaging , Male , Neuropsychological Tests , Positron-Emission Tomography , Repetition Priming , Tauopathies/diagnostic imaging , Tauopathies/psychology , Temporal Lobe/metabolism , Young Adult , tau Proteins/metabolism
2.
J Neurochem ; 158(6): 1394-1411, 2021 09.
Article in English | MEDLINE | ID: mdl-34272732

ABSTRACT

The basal forebrain cholinergic neurons (BFCN) provide the primary source of cholinergic innervation of the human cerebral cortex. They are involved in the cognitive processes of learning, memory, and attention. These neurons are differentially vulnerable in various neuropathologic entities that cause dementia. This review summarizes the relevance to BFCN of neuropathologic markers associated with dementias, including the plaques and tangles of Alzheimer's disease (AD), the Lewy bodies of diffuse Lewy body disease, the tauopathy of frontotemporal lobar degeneration (FTLD-TAU) and the TDP-43 proteinopathy of FTLD-TDP. Each of these proteinopathies has a different relationship to BFCN and their corticofugal axons. Available evidence points to early and substantial degeneration of the BFCN in AD and diffuse Lewy body disease. In AD, the major neurodegenerative correlate is accumulation of phosphotau in neurofibrillary tangles. However, these neurons are less vulnerable to the tauopathy of FTLD. An intriguing finding is that the intracellular tau of AD causes destruction of the BFCN, whereas that of FTLD does not. This observation has profound implications for exploring the impact of different species of tauopathy on neuronal survival. The proteinopathy of FTLD-TDP shows virtually no abnormal inclusions within the BFCN. Thus, the BFCN are highly vulnerable to the neurodegenerative effects of tauopathy in AD, resilient to the neurodegenerative effect of tauopathy in FTLD and apparently resistant to the emergence of proteinopathy in FTLD-TDP and perhaps also in Pick's disease. Investigations are beginning to shed light on the potential mechanisms of this differential vulnerability and their implications for therapeutic intervention.


Subject(s)
Basal Forebrain/metabolism , Choline O-Acetyltransferase/metabolism , Cholinergic Neurons/metabolism , Dementia/metabolism , Nerve Degeneration/metabolism , Receptors, Cholinergic/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Basal Forebrain/pathology , Cholinergic Neurons/pathology , Dementia/pathology , Dementia/psychology , Disease Susceptibility/metabolism , Disease Susceptibility/pathology , Disease Susceptibility/psychology , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Frontotemporal Lobar Degeneration/psychology , Humans , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Lewy Body Disease/psychology , Nerve Degeneration/pathology , Nerve Degeneration/psychology , Resilience, Psychological , Tauopathies/metabolism , Tauopathies/pathology , Tauopathies/psychology
3.
Hum Mol Genet ; 28(19): 3255-3269, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31261380

ABSTRACT

Understanding the biological functions of tau variants can illuminate differential etiologies of Alzheimer's disease (AD) and primary tauopathies. Though the end-stage neuropathological attributes of AD and primary tauopathies are similar, the etiology and behavioral outcomes of these diseases follow unique and divergent trajectories. To study the divergent physiological properties of tau variants on a uniform immunogenetic background, we created somatic transgenesis CNS models of tauopathy utilizing neonatal delivery of adeno-associated viruses expressing wild-type (WT) or mutant tau in non-transgenic mice. We selected four different tau variants-WT tau associated with AD, P301L mutant tau associated with frontotemporal dementia (FTD), S320F mutant tau associated with Pick's disease and a combinatorial approach using P301L/S320F mutant tau. CNS-targeted expression of WT and P301L mutant tau results in robust tau hyperphosphorylation without tangle pathology, gradually developing age-progressive memory deficits. In contrast, the S320F variant, especially in combination with P301L, produces an AD-type tangle pathology, focal neuroinflammation and memory impairment on an accelerated time scale. Using the doubly mutated P301L/S320F tau variant, we demonstrate that combining different mutations can have an additive effect on neuropathologies and associated co-morbidities, possibly hinting at involvement of unique functional pathways. Importantly, we also show that overexpression of wild-type tau as well as an FTD-associated tau variant can lead to cognitive deficits even in the absence of tangles. Together, our data highlights the synergistic neuropathologies and associated cognitive and synaptic alterations of the combinatorial tau variant leading to a robust model of tauopathy.


Subject(s)
Central Nervous System/metabolism , Mutation , Tauopathies/genetics , tau Proteins/genetics , tau Proteins/metabolism , Animals , Behavior, Animal , Disease Models, Animal , Female , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/psychology , Humans , Male , Mice , Mice, Transgenic , Phosphorylation , Pick Disease of the Brain/genetics , Pick Disease of the Brain/metabolism , Pick Disease of the Brain/psychology , Tauopathies/metabolism , Tauopathies/psychology
4.
Ann Neurol ; 88(5): 921-932, 2020 11.
Article in English | MEDLINE | ID: mdl-32799367

ABSTRACT

OBJECTIVE: The goal of this study was to examine sex differences in tau distribution across the brain of older adults, using positron emission tomography (PET), and investigate how these differences might associate with cognitive trajectories. METHODS: Participants were 343 clinically normal individuals (women, 58%; 73.8 [8.5] years) and 55 individuals with mild cognitive impairment (MCI; women, 38%; 76.9 [7.3] years) from the Harvard Aging Brain Study and the Alzheimer's Disease Neuroimaging Initiative. We examined 18 F-Flortaucipir (FTP)-positron emission tomography (PET) signal across 41 cortical and subcortical regions of interest (ROIs). Linear regression models estimated the effect of sex on FTP-signal for each ROI after adjusting for age and cohort. We also examined interactions between sex*Aß-PET positive / negative (+ / -) and sex*apolipoprotein ε4 (APOEε4) status. Linear mixed models estimated the moderating effect of sex on the relationship between a composite of sex-differentiated tau ROIs and cognitive decline. RESULTS: Women showed significantly higher FTP-signals than men across multiple regions of the cortical mantle (p < 0.007). ß-amyloid (Aß)-moderated sex differences in tau signal were localized to medial and inferio-lateral temporal regions (p < 0.007); Aß + women exhibited greater FTP-signal than other groups. APOEε4-moderated sex differences in FTP-signal were only found in the lateral occipital lobe. Women with higher FTP-signals in composite ROI exhibited faster cognitive decline than men (p = 0.04). INTERPRETATION: Tau vulnerability in women is not just limited to the medial temporal lobe and significantly contributed to greater risk of faster cognitive decline. Interactive effects of sex and Aß were predominantly localized in the temporal lobe, however, sex differences in extra-temporal tau highlights the possibility of accelerated tau proliferation in women with the onset of clinical symptomatology. ANN NEUROL 2020;88:921-932.


Subject(s)
Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/psychology , Tauopathies/diagnostic imaging , Tauopathies/psychology , Aged , Aged, 80 and over , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/psychology , Amyloid beta-Peptides/genetics , Apolipoprotein E4/genetics , Carbolines , Disease Progression , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neuroimaging , Occipital Lobe/diagnostic imaging , Positron-Emission Tomography , Radiopharmaceuticals , Sex Characteristics , Temporal Lobe/diagnostic imaging
5.
Acta Neuropathol ; 142(1): 139-158, 2021 07.
Article in English | MEDLINE | ID: mdl-33895869

ABSTRACT

ApoE4 enhances Tau neurotoxicity and promotes the early onset of AD. Pretangle Tau in the noradrenergic locus coeruleus (LC) is the earliest detectable AD-like pathology in the human brain. However, a direct relationship between ApoE4 and Tau in the LC has not been identified. Here we show that ApoE4 selectively binds to the vesicular monoamine transporter 2 (VMAT2) and inhibits neurotransmitter uptake. The exclusion of norepinephrine (NE) from synaptic vesicles leads to its oxidation into the toxic metabolite 3,4-dihydroxyphenyl glycolaldehyde (DOPEGAL), which subsequently activates cleavage of Tau at N368 by asparagine endopeptidase (AEP) and triggers LC neurodegeneration. Our data reveal that ApoE4 boosts Tau neurotoxicity via VMAT2 inhibition, reduces hippocampal volume, and induces cognitive dysfunction in an AEP- and Tau N368-dependent manner, while conversely ApoE3 binds Tau and protects it from cleavage. Thus, ApoE4 exacerbates Tau neurotoxicity by increasing VMAT2 vesicle leakage and facilitating AEP-mediated Tau proteolytic cleavage in the LC via DOPEGAL.


Subject(s)
Alzheimer Disease/pathology , Apolipoprotein E4/pharmacology , Locus Coeruleus/pathology , Tauopathies/pathology , Vesicular Monoamine Transport Proteins/antagonists & inhibitors , Acetaldehyde/analogs & derivatives , Acetaldehyde/metabolism , Aged , Alzheimer Disease/psychology , Animals , Cognition Disorders/psychology , Female , Hippocampus/pathology , Humans , Locus Coeruleus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Neurofibrillary Tangles/pathology , Norepinephrine/metabolism , Synaptic Vesicles/metabolism , Tauopathies/psychology
6.
Am J Geriatr Psychiatry ; 29(4): 375-383, 2021 04.
Article in English | MEDLINE | ID: mdl-32819825

ABSTRACT

Neuropsychiatric syndromes and symptoms play increasingly important roles in research diagnostic criteria for neurodegenerative disorders. Diagnostic criteria were reviewed including those for dementia, Alzheimer's disease, mild cognitive impairment, mild behavioral impairment, prodromal Alzheimer's disease, dementia with Lewy bodies, prodromal dementia with Lewy bodies, Parkinson's disease, multiple system atrophy, frontotemporal dementia, primary progressive aphasia, progressive supranuclear palsy, corticobasal degeneration, traumatic encephalopathy syndrome, Huntington' disease, amyotrophic lateral sclerorsis. All contemporary research diagnostic criteria for neurodegenerative disorders expect those for Parkinson's disease, primary progressive aphasia, multisystem atrophy and amyotrophic lateral sclerosis include neuropsychiatric phenomena as core diagnostic criteria. There are no disease-specific neuropsychiatric symptoms; apathy and disinhibition are common in tauopathies, and rapid-eye-movement sleep behavioral disorder occurs almost exclusively in synucleinopathies. Neuropsychiatric symptoms and syndromes are increasingly integrated into research diagnostic criteria for neurodegenerative disorders; they require clinician skills for recognition; their biology is better understood as their relationships to cognitive, motor, and autonomic symptoms of neurodegenerative disorders are studied.


Subject(s)
Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/psychology , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/psychology , Dementia/diagnosis , Dementia/physiopathology , Dementia/psychology , Humans , Neurodegenerative Diseases/physiopathology , Neuropsychological Tests , Parkinson Disease/diagnosis , Parkinson Disease/physiopathology , Parkinson Disease/psychology , Tauopathies/diagnosis , Tauopathies/physiopathology , Tauopathies/psychology
7.
Brain ; 143(12): 3748-3762, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33184651

ABSTRACT

In Alzheimer's disease, the tauopathy is known as a major mechanism responsible for the development of cognitive deficits. Early biomarkers of such affectations for diagnosis/stratification are crucial in Alzheimer's disease research, and brain connectome studies increasingly show their potential establishing pathology fingerprints at the network level. In this context, we conducted an in vivo multimodal MRI study on young Thy-Tau22 transgenic mice expressing tauopathy, performing resting state functional MRI and structural brain imaging to identify early connectome signatures of the pathology, relating with histological and behavioural investigations. In the prodromal phase of tauopathy, before the emergence of cognitive impairments, Thy-Tau22 mice displayed selective modifications of brain functional connectivity involving three main centres: hippocampus (HIP), amygdala (AMG) and the isocortical areas, notably the somatosensory (SS) cortex. Each of these regions showed differential histopathological profiles. Disrupted ventral HIP-AMG functional pathway and altered dynamic functional connectivity were consistent with high pathological tau deposition and astrogliosis in both hippocampus and amygdala, and significant microglial reactivity in amygdalar nuclei. These patterns were concurrent with widespread functional hyperconnectivity of memory-related circuits of dorsal hippocampus-encompassing dorsal HIP-SS communication-in the absence of significant cortical histopathological markers. These findings suggest the coexistence of two intermingled mechanisms of response at the functional connectome level in the early phases of pathology: a maladaptive and a likely compensatory response. Captured in the connectivity patterns, such first responses to pathology could further be used in translational investigations as a lead towards an early biomarker of tauopathy as well as new targets for future treatments.


Subject(s)
Memory Disorders/pathology , Memory Disorders/psychology , Nerve Net/pathology , Tauopathies/pathology , Tauopathies/psychology , Animals , Astrocytes/pathology , Brain/diagnostic imaging , Brain/pathology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/psychology , Connectome , Disease Progression , Gliosis/pathology , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Memory Disorders/etiology , Mice , Mice, Transgenic , Nerve Net/diagnostic imaging , Tauopathies/complications , Tauopathies/diagnostic imaging , tau Proteins/metabolism
8.
Brain ; 142(11): 3636-3654, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31599329

ABSTRACT

Accumulating data support the role of tau pathology in cognitive decline in ageing and Alzheimer's disease, but underlying mechanisms remain ill-defined. Interestingly, ageing and Alzheimer's disease have been associated with an abnormal upregulation of adenosine A2A receptor (A2AR), a fine tuner of synaptic plasticity. However, the link between A2AR signalling and tau pathology has remained largely unexplored. In the present study, we report for the first time a significant upregulation of A2AR in patients suffering from frontotemporal lobar degeneration with the MAPT P301L mutation. To model these alterations, we induced neuronal A2AR upregulation in a tauopathy mouse model (THY-Tau22) using a new conditional strain allowing forebrain overexpression of the receptor. We found that neuronal A2AR upregulation increases tau hyperphosphorylation, potentiating the onset of tau-induced memory deficits. This detrimental effect was linked to a singular microglial signature as revealed by RNA sequencing analysis. In particular, we found that A2AR overexpression in THY-Tau22 mice led to the hippocampal upregulation of C1q complement protein-also observed in patients with frontotemporal lobar degeneration-and correlated with the loss of glutamatergic synapses, likely underlying the observed memory deficits. These data reveal a key impact of overactive neuronal A2AR in the onset of synaptic loss in tauopathies, paving the way for new therapeutic approaches.


Subject(s)
Complement C1q/metabolism , Neurons/metabolism , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Synapses/pathology , Tauopathies/genetics , Tauopathies/pathology , Animals , Autopsy , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Humans , Memory Disorders/etiology , Memory Disorders/psychology , Mice , Mice, Transgenic , Mutation , Spatial Learning , Tauopathies/psychology , tau Proteins/genetics
9.
J Neurosci ; 38(1): 74-92, 2018 01 03.
Article in English | MEDLINE | ID: mdl-29133432

ABSTRACT

The brainstem locus coeruleus (LC) supplies norepinephrine to the forebrain and degenerates in Alzheimer's disease (AD). Loss of LC neurons is correlated with increased severity of other AD hallmarks, including ß-amyloid (Aß) plaques, tau neurofibrillary tangles, and cognitive deficits, suggesting that it contributes to the disease progression. Lesions of the LC in amyloid-based transgenic mouse models of AD exacerbate Aß pathology, neuroinflammation, and cognitive deficits, but it is unknown how the loss of LC neurons affects tau-mediated pathology or behavioral abnormalities. Here we investigate the impact of LC degeneration in a mouse model of tauopathy by lesioning the LC of male and female P301S tau transgenic mice with the neurotoxin N-(2-chloroethyl)-N-ethyl-bromobenzylamine (DSP-4) starting at 2 months of age. By 6 months, deficits in hippocampal-dependent spatial (Morris water maze) and associative (contextual fear conditioning) memory were observed in lesioned P301S mice while performance remained intact in all other genotype and treatment groups, indicating that tau and LC degeneration act synergistically to impair cognition. By 10 months, the hippocampal neuroinflammation and neurodegeneration typically observed in unlesioned P301S mice were exacerbated by DSP-4, and mortality was also accelerated. These DSP-4-induced changes were accompanied by only a mild aggravation of tau pathology, suggesting that increased tau burden cannot fully account for the effects of LC degeneration. Combined, these experiments demonstrate that loss of LC noradrenergic neurons exacerbates multiple phenotypes caused by pathogenic tau, and provides complementary data to highlight the dual role LC degeneration has on both tau and Aß pathologies in AD.SIGNIFICANCE STATEMENT Elucidating the mechanisms underlying AD is crucial to developing effective diagnostics and therapeutics. The degeneration of the LC and loss of noradrenergic transmission have been recognized as ubiquitous events in AD pathology, and previous studies demonstrated that LC lesions exacerbate pathology and cognitive deficits in amyloid-based mouse models. Here, we reveal a complementary role of LC degeneration on tau-mediated aspects of the disease by using selective lesions of the LC and the noradrenergic system to demonstrate an exacerbation of cognitive deficits, neuroinflammation, neurodegeneration in a transgenic mouse model of tauopathy. Our data support an integral role for the LC in modulating the severity of both canonical AD-associated pathologies, as well as the detrimental consequences of LC degeneration during disease progression.


Subject(s)
Cognition Disorders/pathology , Cognition Disorders/psychology , Genes, Lethal/genetics , Locus Coeruleus/pathology , Tauopathies/genetics , Tauopathies/pathology , tau Proteins/genetics , Animals , Benzylamines/toxicity , Conditioning, Psychological/drug effects , Fear/drug effects , Female , Hippocampus/pathology , Inflammation/pathology , Male , Maze Learning/drug effects , Memory Disorders/pathology , Memory Disorders/psychology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Norepinephrine/metabolism , Tauopathies/psychology
10.
Glia ; 66(4): 813-827, 2018 04.
Article in English | MEDLINE | ID: mdl-29315804

ABSTRACT

Early white matter (WM) changes are common in dementia and may contribute to functional decline. We here examine this phenomenon in an induced dementia model for the first time. We report a novel and selective form of myelin injury as the first manifestation of tauopathy in the adult central nervous system. Myelin pathology rapidly followed the induction of a P301 tau mutation associated with fronto-temporal dementia in humans (rTG4510 line). Damage involved focal disruption of the ad-axonal myelin lamella and internal oligodendrocyte tongue process, followed by myelin remodeling with features of re-myelination that included myelin thinning and internodal shortening. The evolution of the re-myelinated phenotype was complete in the molecular layer of the dentate gyrus after 1 month and in the optic nerve (ON) after 9 months of transgene induction and proceeded in the absence of actual demyelination, reactive glial changes or inflammatory response. The initial rapid myelin pathology was associated with loss of WM function and performance decline in a novel recognition test and both these effects largely reversed during the myelin re-modeling phase. The initial phase of myelin injury was accompanied by disruption of the vesicle population present in the axoplasm of hippocampal and ON axons. Axoplasmic vesicle release is significant for the regulation of myelin plasticity and disruption of this pathway may underlie the myelin damage and remodeling evoked by tauopathy. WM dysfunction early in tauopathy will disorder neural circuits, the current findings suggest this event may make a significant contribution to early clinical deficit in dementia.


Subject(s)
Myelin Sheath/pathology , Myelin Sheath/physiology , Tauopathies/pathology , Tauopathies/physiopathology , White Matter/pathology , White Matter/physiopathology , Animals , Astrocytes/pathology , Astrocytes/physiology , Axons/pathology , Axons/physiology , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/pathology , Disease Progression , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Male , Memory Disorders/pathology , Memory Disorders/physiopathology , Mice, Inbred C57BL , Mice, Transgenic , Optic Nerve/pathology , Optic Nerve/physiopathology , Recognition, Psychology/physiology , Tauopathies/psychology , tau Proteins/genetics , tau Proteins/metabolism
11.
Brain ; 140(4): 1128-1146, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28334843

ABSTRACT

Mutations in the gene encoding tau (MAPT) cause frontotemporal dementia spectrum disorders. A rare tau variant p.A152T was reported as a risk factor for frontotemporal dementia spectrum and Alzheimer's disease in an initial case-control study. Such findings need replication in an independent cohort. We analysed an independent multinational cohort comprising 3100 patients with neurodegenerative disease and 4351 healthy control subjects and found p.A152T associated with significantly higher risk for clinically defined frontotemporal dementia and progressive supranuclear palsy syndrome. To assess the functional and biochemical consequences of this variant, we generated transgenic zebrafish models expressing wild-type or A152T-tau, where A152T caused neurodegeneration and proteasome compromise. Impaired proteasome activity may also enhance accumulation of other proteins associated with this variant. We increased A152T clearance kinetics by both pharmacological and genetic upregulation of autophagy and ameliorated the disease pathology observed in A152T-tau fish. Thus, autophagy-upregulating therapies may be a strategy for the treatment for tauopathies.


Subject(s)
Autophagy , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/therapy , Supranuclear Palsy, Progressive/genetics , Supranuclear Palsy, Progressive/therapy , Tauopathies/genetics , Tauopathies/therapy , Zebrafish , tau Proteins/genetics , Alleles , Animals , Autophagy-Related Protein 5 , Behavior, Animal , Disease Models, Animal , Embryo, Nonmammalian , Frontotemporal Dementia/genetics , Humans , Kinetics , Polymorphism, Single Nucleotide , Proteasome Endopeptidase Complex/genetics , RNA/biosynthesis , RNA/genetics , Tauopathies/psychology , Zebrafish Proteins , tau Proteins/metabolism
12.
Rev Neurol (Paris) ; 172(11): 709-714, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27344208

ABSTRACT

While increasing life expectancy is a major achievement, the global aging of societies raises a number of medical issues, such as the development of age-related disorders, including neurodegenerative diseases. The three main disease groups constituting the majority of neurodegenerative diseases are tauopathies, alpha-synucleinopathies and diseases due to repetitions of glutamine (including Huntington's disease). In each neurodegenerative disease, the accumulation of one or more aggregated proteins has been identified as the molecular signature of the disease (as seen, for example, in Alzheimer's disease, Parkinson's disease, dementia with Lewy bodies, amyotrophic lateral sclerosis and frontotemporal dementia). The etiology of neurodegenerative diseases is often multifactorial, and the known risk factors include, in addition to genetic polymorphisms and age, some other possible causes, such as certain immune and metabolic conditions, endocrine pathologies, gender, socioeconomic or professional status, oxidative stress or inflammation, vitamin deficiencies and environmental factors (chemical exposure, metals). However, innovative strategies to elaborate suitable diagnostic and therapeutic approaches (aiming to at least delay or possibly even reverse disease progression) require further knowledge of the genetic and adaptive immunological characteristics of neurodegenerative diseases.


Subject(s)
Nerve Degeneration/etiology , Neurodegenerative Diseases/etiology , Parkinsonian Disorders/complications , Tauopathies/complications , Brain/anatomy & histology , Brain/pathology , Cytoskeleton/pathology , Humans , Inclusion Bodies/pathology , Lewy Body Disease/complications , Lewy Body Disease/psychology , Nerve Degeneration/pathology , Neurodegenerative Diseases/psychology , Parkinsonian Disorders/psychology , Tauopathies/psychology , alpha-Synuclein/physiology
13.
J Neurochem ; 135(2): 207-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26451974

ABSTRACT

This Editorial highlights a study by Hunsberger et al. (2015) in the current issue of Journal of Neurochemistry, in which the authors explore the effects of riluzole (R) treatment on tau-P301L transgenic mice. The authors employed a comprehensive analysis of possible restorative effects of the drug by examining glutamate levels in subregions of the hippocampus, expression of tau and its hyper-phosphorylated forms, and memory function using behavioral tests. The authors report a simultaneous reduction in glutamate reuptake and an increase in glutamate release in the tau-P301L model, both of which are ameliorated with riluzole treatment. The authors' findings have implications for our understanding of synaptic transmission mechanisms also associated with Alzheimer's disease pathology.


Subject(s)
Cognition Disorders/prevention & control , Cognition Disorders/psychology , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/metabolism , Neuroprotective Agents/pharmacology , Riluzole/pharmacology , Tauopathies/prevention & control , Tauopathies/psychology , tau Proteins/biosynthesis , Animals , Humans
14.
J Neurochem ; 135(2): 381-94, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26146790

ABSTRACT

Hyperexcitability of the hippocampus is a commonly observed phenomenon in the years preceding a diagnosis of Alzheimer's disease (AD). Our previous work suggests a dysregulation in glutamate neurotransmission may mediate this hyperexcitability, and glutamate dysregulation correlates with cognitive deficits in the rTg(TauP301L)4510 mouse model of AD. To determine whether improving glutamate regulation would attenuate cognitive deficits and AD-related pathology, TauP301L mice were treated with riluzole (~ 12.5 mg/kg/day p.o.), an FDA-approved drug for amyotrophic lateral sclerosis that lowers extracellular glutamate levels. Riluzole-treated TauP301L mice exhibited improved performance in the water radial arm maze and the Morris water maze, associated with a decrease in glutamate release and an increase in glutamate uptake in the dentate gyrus, cornu ammonis 3 (CA3), and cornu ammonis 1 (CA1) regions of the hippocampus. Riluzole also attenuated the TauP301L-mediated increase in hippocampal vesicular glutamate transporter 1, which packages glutamate into vesicles and influences glutamate release; and the TauP301L-mediated decrease in hippocampal glutamate transporter 1, the major transporter responsible for removing glutamate from the extracellular space. The TauP301L-mediated reduction in PSD-95 expression, a marker of excitatory synapses in the hippocampus, was also rescued by riluzole. Riluzole treatment reduced total levels of tau, as well as the pathological phosphorylation and conformational changes in tau associated with the P301L mutation. These findings open new opportunities for the development of clinically applicable therapeutic approaches to regulate glutamate in vulnerable circuits for those at risk for the development of AD.


Subject(s)
Cognition Disorders/prevention & control , Cognition Disorders/psychology , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/metabolism , Neuroprotective Agents/pharmacology , Riluzole/pharmacology , Tauopathies/prevention & control , Tauopathies/psychology , tau Proteins/biosynthesis , Alzheimer Disease/prevention & control , Alzheimer Disease/psychology , Animals , Brain Chemistry/drug effects , Humans , Maze Learning/drug effects , Mice , Mice, Transgenic , Synapses/drug effects , Synapses/pathology , Vesicular Glutamate Transport Protein 1/metabolism
15.
Psychiatr Danub ; 27 Suppl 1: S515-25, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26540742

ABSTRACT

Alzheimer's disease is increasing to epidemic levels with an estimated 36 million people affected worldwide (Wimo 2010). The aetiology of the disease is not known, which is hindering the progression of the treatment. This study is a longitudinal investigation into the performance of TgTauP301L mice as an animal model of Alzheimer's disease on the computer automated touchscreen 5-choice serial reaction time task (5-CSRTT). TgTauP301L mice have a single tau mutation in the P301L gene and develop the tau pathology that represents the observed tauopathy in patients with Alzheimer's disease. The aim of the investigation is to observe if tau pathology in the TgTauP301L mice causes a cognitive impairment in attention and executive function and at what stage this can be identified by the 5-CSRTT task. This will establish if the animals can be used as a therapeutic model for pre-clinical drug trials and help to identify an early indicator and intervention point in patients with Alzheimer's disease. The animals have previously been studied at 5-months and no differences between performances of the TgTauP301L mice and wild type mice were found (unpublished data). This study measured the performance of the animals at 7-months which is when the tauopathy begins to develop in TgTauP301L mice (Murakami 2005). The results of this study showed that there was no deficit in the performance of the TgTauP301L compared to the wild type mice and there had been no change in the animals' performance compared to at 5-months. The animals will be retested at 12-months once the pathology has extensively spread to see if the tauopathy causes a deficit in performance.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/psychology , Disease Models, Animal , Reaction Time/genetics , tau Proteins/genetics , Alzheimer Disease/drug therapy , Animals , Attention/physiology , Executive Function/physiology , Humans , Male , Mice , Mice, Transgenic , Mutation , Serial Learning/physiology , Tauopathies/genetics , Tauopathies/psychology
16.
J Neurol Neurosurg Psychiatry ; 85(9): 1016-23, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24521566

ABSTRACT

BACKGROUND: Mutations in C9ORF72 are an important cause of frontotemporal dementia (FTD) and motor neuron disease. Accumulating evidence suggests that FTD associated with C9ORF72 mutations (C9ORF72-FTD) is distinguished clinically by early prominent neuropsychiatric features that might collectively reflect deranged body schema processing. However, the pathophysiology of C9ORF72-FTD has not been elucidated. METHODS: We undertook a detailed neurophysiological investigation of five patients with C9ORF72-FTD, in relation to patients with FTD occurring sporadically and on the basis of mutations in the microtubule-associated protein tau gene and healthy older individuals. We designed or adapted behavioural tasks systematically to assess aspects of somatosensory body schema processing (tactile discrimination, proprioceptive and body part illusions and self/non-self differentiation). RESULTS: Patients with C9ORF72-FTD selectively exhibited deficits at these levels of body schema processing in relation to healthy individuals and other patients with FTD. CONCLUSIONS: Altered body schema processing is a novel, generic pathophysiological mechanism that may link the distributed cortico-subcortical network previously implicated in C9ORF72-FTD with a wide range of neuropsychiatric and behavioural symptoms, and constitute a physiological marker of this neurodegenerative proteinopathy.


Subject(s)
Body Image/psychology , Frontotemporal Dementia/genetics , Frontotemporal Dementia/psychology , Proteins/genetics , Aged , C9orf72 Protein , Case-Control Studies , Female , Humans , Illusions/psychology , Male , Middle Aged , Mutation , Self Concept , Tauopathies/genetics , Tauopathies/psychology , Touch Perception , tau Proteins/genetics
17.
J Neurosci ; 32(11): 3601-11, 2012 Mar 14.
Article in English | MEDLINE | ID: mdl-22423084

ABSTRACT

Neurodegenerative tauopathies, such as Alzheimer's disease (AD), are characterized by insoluble deposits of hyperphosphorylated tau protein within brain neurons. Increased phosphorylation and decreased solubility has been proposed to diminish normal tau stabilization of microtubules (MTs), thereby leading to neuronal dysfunction. Earlier studies have provided evidence that small molecule MT-stabilizing drugs that are used in the treatment of cancer may have utility in the treatment of tauopathies. However, it has not been established whether treatment with a small molecule MT-stabilizing compound will provide benefit in a transgenic model with pre-existing tau pathology, as would be seen in human patients with clinical symptoms. Accordingly, we describe here an interventional study of the brain-penetrant MT-stabilizing agent, epothilone D (EpoD), in aged PS19 mice with existing tau pathology and related behavioral deficits. EpoD treatment reduced axonal dystrophy and increased axonal MT density in the aged PS19 mice, which led to improved fast axonal transport and cognitive performance. Moreover, the EpoD-treated PS19 mice had less forebrain tau pathology and increased hippocampal neuronal integrity, with no dose-limiting side effects. These data reveal that brain-penetrant MT-stabilizing drugs hold promise for the treatment of AD and related tauopathies, and that EpoD could be a candidate for clinical testing.


Subject(s)
Aging/drug effects , Alzheimer Disease/drug therapy , Cognition Disorders/drug therapy , Epothilones/therapeutic use , Tauopathies/drug therapy , Tubulin Modulators/therapeutic use , Aging/pathology , Aging/psychology , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Axons/drug effects , Axons/pathology , Cognition Disorders/pathology , Cognition Disorders/psychology , Epothilones/pharmacology , Humans , Male , Mice , Mice, Transgenic , Microtubules/drug effects , Microtubules/pathology , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/psychology , Tauopathies/pathology , Tauopathies/psychology , Tubulin Modulators/pharmacology , tau Proteins/genetics
18.
J Neurosci ; 32(21): 7137-45, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22623658

ABSTRACT

Tau is a microtubule (MT)-stabilizing protein that is altered in Alzheimer's disease (AD) and other tauopathies. It is hypothesized that the hyperphosphorylated, conformationally altered, and multimeric forms of tau lead to a disruption of MT stability; however, direct evidence is lacking in vivo. In this study, an in vivo stable isotope-mass spectrometric technique was used to measure the turnover, or dynamicity, of MTs in brains of living animals. We demonstrated an age-dependent increase in MT dynamics in two different tau transgenic mouse models, 3xTg and rTg4510. MT hyperdynamicity was dependent on tau expression, since a reduction of transgene expression with doxycycline reversed the MT changes. Treatment of rTg4510 mice with the epothilone, BMS-241027, also restored MT dynamics to baseline levels. In addition, MT stabilization with BMS-241027 had beneficial effects on Morris water maze deficits, tau pathology, and neurodegeneration. Interestingly, pathological and functional benefits of BMS-241027 were observed at doses that only partially reversed MT hyperdynamicity. Together, these data suggest that tau-mediated loss of MT stability may contribute to disease progression and that very low doses of BMS-241027 may be useful in the treatment of AD and other tauopathies.


Subject(s)
Cognition Disorders/drug therapy , Epothilones/therapeutic use , Microtubules/pathology , Nerve Degeneration/drug therapy , Tauopathies/drug therapy , Tubulin Modulators/therapeutic use , tau Proteins/physiology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cognition Disorders/complications , Cognition Disorders/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Doxycycline/pharmacology , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/psychology , Epothilones/pharmacology , Female , Hippocampus/drug effects , Hippocampus/pathology , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubules/drug effects , Tauopathies/complications , Tauopathies/genetics , Tauopathies/pathology , Tauopathies/psychology , Tubulin Modulators/pharmacology , tau Proteins/antagonists & inhibitors , tau Proteins/biosynthesis , tau Proteins/genetics
19.
Neurocase ; 18(6): 478-88, 2012.
Article in English | MEDLINE | ID: mdl-22181323

ABSTRACT

BACKGROUND: Progressive supranuclear palsy (PSP) is a neurodegenerative disorder pathologically characterized by neuronal loss, gliosis and tau-positive neurofibrillary tangles in basal ganglia, brainstem and cerebellar nuclei. Five presenting clinical syndromes of PSP are well-described: (i) the classic Richardson's syndrome; (ii) asymmetric parkinsonism with tremor; (iii) freezing of gait; (iv) asymmetric limb apraxia, and (v) apraxia of speech. AIM: To determine whether autopsy-proven PSP cases may present with another clinical phenotype. METHODS: Medical records of 66 autopsy-proven PSP cases between 1973 and 2010 were reviewed to determine whether all could be classified into one of five well-defined syndromes listed above. Three cases presented with prominent behavioral and personality changes, meeting diagnosis of behavioral variant frontotemporal dementia. MRI midbrain and pons volumes and pons/midbrain ratios were compared to healthy controls and typical PSP cases. RESULTS: All three bvFTD cases developed at least one PSP symptom or sign that emerged up to 5 years after initial presentation. One case was re-diagnosed as PSP 6 years after presentation as bvFTD. Compared to controls, midbrain volume was significantly smaller in both bvFTD (p = .03) and PSP cases (p = .008), without significant difference between PSP and bvFTD cases (.44). However pontine volumes were similar across all three groups. CONCLUSIONS: While most autopsy-confirmed PSP cases present with one of the five well-described syndromes, there are cases that may present as bvFTD. In these, at least one cardinal symptom or sign of PSP later emerges, associated with smaller midbrain volume and increased pons/midbrain ratio. Thus underlying PSP pathology should be considered in these cases.


Subject(s)
Frontotemporal Dementia/pathology , Supranuclear Palsy, Progressive/pathology , Tauopathies/pathology , Aged , Aged, 80 and over , Autopsy , Behavioral Symptoms , Diagnosis, Differential , Female , Frontotemporal Dementia/classification , Frontotemporal Dementia/psychology , Humans , Male , Middle Aged , Retrospective Studies , Supranuclear Palsy, Progressive/classification , Supranuclear Palsy, Progressive/psychology , Tauopathies/classification , Tauopathies/psychology
20.
Eur Neurol ; 67(3): 142-50, 2012.
Article in English | MEDLINE | ID: mdl-22261560

ABSTRACT

Neurodegenerative tauopathies may be inherited as autosomal-dominant disorders with variable clinicopathological phenotypes, and causative mutations in the microtubule-associated protein tau (MAPT) gene are not regularly seen. Herein, we describe a patient with clinically typical and autopsy-proven corticobasal degeneration (CBD). Her mother was diagnosed to have Parkinson's disease, but autopsy showed CBD pathology as in the index patient. The sister of the index patient had the clinical symptoms of primary progressive aphasia (PPA), but no pathology was available to date. Molecular analysis did not reveal any mutation in the MAPT or progranulin (GRN) genes. Our findings illustrate that CBD, progressive supranuclear palsy and PPA may be overlapping diseases with a common pathological basis rather than distinct entities. Clinical presentation and course might be determined by additional, yet unknown, genetic modifying factors.


Subject(s)
Basal Ganglia Diseases/pathology , Brain/pathology , Nerve Degeneration/pathology , Tauopathies/pathology , Aphasia, Primary Progressive/genetics , Aphasia, Primary Progressive/pathology , Aphasia, Primary Progressive/psychology , Basal Ganglia Diseases/genetics , Basal Ganglia Diseases/psychology , Female , Humans , Middle Aged , Nerve Degeneration/genetics , Nerve Degeneration/psychology , Neurologic Examination , Neuropsychological Tests , Pedigree , Phenotype , Supranuclear Palsy, Progressive/genetics , Supranuclear Palsy, Progressive/pathology , Supranuclear Palsy, Progressive/psychology , Tauopathies/genetics , Tauopathies/psychology
SELECTION OF CITATIONS
SEARCH DETAIL