Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 299(3): 102888, 2023 03.
Article in English | MEDLINE | ID: mdl-36634849

ABSTRACT

In several neurodegenerative disorders, the neuronal proteins tau and α-synuclein adopt aggregation-prone conformations capable of replicating within and between cells. To better understand how these conformational changes drive neuropathology, we compared the interactomes of tau and α-synuclein in the presence or the absence of recombinant fibril seeds. Human embryonic stem cells with an inducible neurogenin-2 transgene were differentiated into glutamatergic neurons expressing (1) WT 0N4R tau, (2) mutant (P301L) 0N4R tau, (3) WT α-synuclein, or (4) mutant (A53T) α-synuclein, each genetically fused to a promiscuous biotin ligase (BioID2). Neurons expressing unfused BioID2 served as controls. After treatment with fibrils or PBS, interacting proteins were labeled with biotin in situ and quantified using mass spectrometry via tandem mass tag labeling. By comparing interactions in mutant versus WT neurons and in fibril- versus PBS-treated neurons, we observed changes in protein interactions that are likely relevant to disease progression. We identified 45 shared interactors, suggesting that tau and α-synuclein function within some of the same pathways. Potential loci of shared interactions include microtubules, Wnt signaling complexes, and RNA granules. Following fibril treatment, physiological interactions decreased, whereas other interactions, including those between tau and 14-3-3 η, increased. We confirmed that 14-3-3 proteins, which are known to colocalize with protein aggregates during neurodegeneration, can promote or inhibit tau aggregation in vitro depending on the specific combination of 14-3-3 isoform and tau sequence.


Subject(s)
Biotin , Neurons , alpha-Synuclein , Humans , alpha-Synuclein/metabolism , Biotin/metabolism , Microtubules/metabolism , Neurons/metabolism , tau Proteins/metabolism
2.
J Virol ; 97(2): e0167222, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36651748

ABSTRACT

Phenotypic screening has yielded small-molecule inhibitors of prion replication that are effective in vivo against certain prion strains but not others. Here, we sought to test the small molecule anle138b in multiple mouse models of prion disease. In mice inoculated with the RML strain of prions, anle138b doubled survival and durably suppressed astrogliosis measured by live-animal bioluminescence imaging. In knock-in mouse models of the D178N and E200K mutations that cause genetic prion disease, however, we were unable to identify a clear, quantifiable disease endpoint against which to measure therapeutic efficacy. Among untreated animals, the mutations did not impact overall survival, and bioluminescence remained low out to >20 months of age. Vacuolization and PrP deposition were observed in some brain regions in a subset of mutant animals but appeared to be unable to carry the weight of a primary endpoint in a therapeutic study. We conclude that not all animal models of prion disease are suited to well-powered therapeutic efficacy studies, and care should be taken in choosing the models that will support drug development programs. IMPORTANCE There is an urgent need to develop drugs for prion disease, a currently untreatable neurodegenerative disease. In this effort, there is a debate over which animal models can best support a drug development program. While the study of prion disease benefits from excellent animal models because prions naturally afflict many different mammals, different models have different capabilities and limitations. Here, we conducted a therapeutic efficacy study of the drug candidate anle138b in mouse models with two of the most common mutations that cause genetic prion disease. In a more typical model where prions are injected directly into the brain, we found anle138b to be effective. In the genetic models, however, the animals never reached a clear, measurable point of disease onset. We conclude that not all prion disease animal models are ideally suited to drug efficacy studies, and well-defined, quantitative disease metrics should be a priority.


Subject(s)
Prion Diseases , Pyrazoles , Animals , Mice , Disease Models, Animal , Mice, Transgenic , Prion Diseases/drug therapy , Prion Diseases/genetics , Prions/genetics , Pyrazoles/therapeutic use
3.
Int J Mol Sci ; 22(9)2021 May 04.
Article in English | MEDLINE | ID: mdl-34064393

ABSTRACT

Although it is not yet universally accepted that all neurodegenerative diseases (NDs) are prion disorders, there is little disagreement that Alzheimer's disease (AD), Parkinson's disease, frontotemporal dementia (FTD), and other NDs are a consequence of protein misfolding, aggregation, and spread. This widely accepted perspective arose from the prion hypothesis, which resulted from investigations on scrapie, a common transmissible disease of sheep and goats. The prion hypothesis argued that the causative infectious agent of scrapie was a novel proteinaceous pathogen devoid of functional nucleic acids and distinct from viruses, viroids, and bacteria. At the time, it seemed impossible that an infectious agent like the one causing scrapie could replicate and exist as diverse microbiological strains without nucleic acids. However, aggregates of a misfolded host-encoded protein, designated the prion protein (PrP), were shown to be the cause of scrapie as well as Creutzfeldt-Jakob disease (CJD) and Gerstmann-Sträussler-Scheinker syndrome (GSS), which are similar NDs in humans. This review discusses historical research on diseases caused by PrP misfolding, emphasizing principles of pathogenesis that were later found to be core features of other NDs. For example, the discovery that familial prion diseases can be caused by mutations in PrP was important for understanding prion replication and disease susceptibility not only for rare PrP diseases but also for far more common NDs involving other proteins. We compare diseases caused by misfolding and aggregation of APP-derived Aß peptides, tau, and α-synuclein with PrP prion disorders and argue for the classification of NDs caused by misfolding of these proteins as prion diseases. Deciphering the molecular pathogenesis of NDs as prion-mediated has provided new approaches for finding therapies for these intractable, invariably fatal disorders and has revolutionized the field.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Prion Proteins/genetics , Scrapie/genetics , alpha-Synuclein/genetics , tau Proteins/genetics , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Animals , Creutzfeldt-Jakob Syndrome/etiology , Creutzfeldt-Jakob Syndrome/genetics , Creutzfeldt-Jakob Syndrome/metabolism , Creutzfeldt-Jakob Syndrome/pathology , Frontotemporal Dementia/etiology , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Gene Expression , Gerstmann-Straussler-Scheinker Disease/etiology , Gerstmann-Straussler-Scheinker Disease/genetics , Gerstmann-Straussler-Scheinker Disease/metabolism , Gerstmann-Straussler-Scheinker Disease/pathology , Humans , Mice , Mutation , Parkinson Disease/etiology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Prion Proteins/chemistry , Prion Proteins/metabolism , Prions , Protein Folding , Scrapie/etiology , Scrapie/metabolism , Scrapie/pathology , Sheep , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , tau Proteins/chemistry , tau Proteins/metabolism
4.
Neuron ; 109(10): 1675-1691.e9, 2021 05 19.
Article in English | MEDLINE | ID: mdl-33848474

ABSTRACT

Tau aggregates contribute to neurodegenerative diseases, including frontotemporal dementia and Alzheimer's disease (AD). Although RNA promotes tau aggregation in vitro, whether tau aggregates in cells contain RNA is unknown. We demonstrate, in cell culture and mouse brains, that cytosolic and nuclear tau aggregates contain RNA with enrichment for small nuclear RNAs (snRNAs) and small nucleolar RNAs (snoRNAs). Nuclear tau aggregates colocalize with and alter the composition, dynamics, and organization of nuclear speckles, membraneless organelles involved in pre-mRNA splicing. Moreover, several nuclear speckle components, including SRRM2, mislocalize to cytosolic tau aggregates in cells, mouse brains, and brains of individuals with AD, frontotemporal dementia (FTD), and corticobasal degeneration (CBD). Consistent with these alterations, we observe that the presence of tau aggregates is sufficient to alter pre-mRNA splicing. This work identifies tau alteration of nuclear speckles as a feature of tau aggregation that may contribute to the pathology of tau aggregates.


Subject(s)
Alzheimer Disease/metabolism , Cell Nucleus/metabolism , RNA, Small Nucleolar/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Animals , Cell Nucleus/ultrastructure , Cytosol/metabolism , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Protein Binding , Protein Transport , RNA Splicing , RNA-Binding Proteins/metabolism
5.
Nucleic Acids Res ; 48(19): 10615-10631, 2020 11 04.
Article in English | MEDLINE | ID: mdl-32776089

ABSTRACT

Lowering of prion protein (PrP) expression in the brain is a genetically validated therapeutic hypothesis in prion disease. We recently showed that antisense oligonucleotide (ASO)-mediated PrP suppression extends survival and delays disease onset in intracerebrally prion-infected mice in both prophylactic and delayed dosing paradigms. Here, we examine the efficacy of this therapeutic approach across diverse paradigms, varying the dose and dosing regimen, prion strain, treatment timepoint, and examining symptomatic, survival, and biomarker readouts. We recapitulate our previous findings with additional PrP-targeting ASOs, and demonstrate therapeutic benefit against four additional prion strains. We demonstrate that <25% PrP suppression is sufficient to extend survival and delay symptoms in a prophylactic paradigm. Rise in both neuroinflammation and neuronal injury markers can be reversed by a single dose of PrP-lowering ASO administered after the detection of pathological change. Chronic ASO-mediated suppression of PrP beginning at any time up to early signs of neuropathology confers benefit similar to constitutive heterozygous PrP knockout. Remarkably, even after emergence of frank symptoms including weight loss, a single treatment prolongs survival by months in a subset of animals. These results support ASO-mediated PrP lowering, and PrP-lowering therapeutics in general, as a promising path forward against prion disease.


Subject(s)
Oligonucleotides, Antisense/therapeutic use , Prion Diseases/therapy , Prion Proteins/genetics , RNAi Therapeutics/methods , Animals , Brain/metabolism , Brain/pathology , Cell Line , Mice , Mice, Inbred C57BL , Oligonucleotides, Antisense/chemistry , Prion Proteins/metabolism
6.
Acta Neuropathol Commun ; 7(1): 81, 2019 05 20.
Article in English | MEDLINE | ID: mdl-31109379

ABSTRACT

Glial cytoplasmic inclusions (GCIs) containing aggregated and hyperphosphorylated α-synuclein are the signature neuropathological hallmark of multiple system atrophy (MSA). Native α-synuclein can adopt a prion conformation that self-propagates and spreads throughout the brain ultimately resulting in neurodegeneration. A growing body of evidence argues that, in addition to oligodendrocytes, astrocytes contain α-synuclein inclusions in MSA and other α-synucleinopathies at advanced stages of disease. To study the role of astrocytes in MSA, we added MSA brain homogenate to primary cultures of astrocytes from transgenic (Tg) mouse lines expressing human α-synuclein. Astrocytes from four Tg lines, expressing either wild-type or mutant (A53T or A30P) human α-synuclein, propagated and accumulated α-synuclein prions. Furthermore, we found that MSA-infected astrocytes formed two morphologically distinct α-synuclein inclusions: filamentous and granular. Both types of cytoplasmic inclusions shared several features characteristic of α-synuclein inclusions in synucleinopathies: hyperphosphorylation preceded by aggregation, ubiquitination, thioflavin S-positivity, and co-localization with p62. Our findings demonstrate that human α-synuclein forms distinct inclusion morphologies and propagates within cultured Tg astrocytes exposed to MSA prions, indicating that α-synuclein expression determines the tropism of inclusion formation in certain cells. Thus, our work may prove useful in elucidating the role of astrocytes in the pathogenic mechanisms that feature in neurodegeneration caused by MSA prions.


Subject(s)
Astrocytes/pathology , Multiple System Atrophy/pathology , Prions/metabolism , alpha-Synuclein/metabolism , Animals , Astrocytes/metabolism , Cells, Cultured , Dendritic Spines/metabolism , Dendritic Spines/pathology , Humans , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Mice, Transgenic , Multiple System Atrophy/metabolism , Recombinant Proteins/administration & dosage , alpha-Synuclein/administration & dosage
7.
Nat Neurosci ; 22(1): 57-64, 2019 01.
Article in English | MEDLINE | ID: mdl-30559471

ABSTRACT

The coexistence of amyloid-ß (Aß) plaques and tau neurofibrillary tangles in the neocortex is linked to neural system failure and cognitive decline in Alzheimer's disease. However, the underlying neuronal mechanisms are unknown. By employing in vivo two-photon Ca2+ imaging of layer 2/3 cortical neurons in mice expressing human Aß and tau, we reveal a dramatic tau-dependent suppression of activity and silencing of many neurons, which dominates over Aß-dependent neuronal hyperactivity. We show that neurofibrillary tangles are neither sufficient nor required for the silencing, which instead is dependent on soluble tau. Surprisingly, although rapidly effective in tau mice, suppression of tau gene expression was much less effective in rescuing neuronal impairments in mice containing both Aß and tau. Together, our results reveal how Aß and tau synergize to impair the functional integrity of neural circuits in vivo and suggest a possible cellular explanation contributing to disappointing results from anti-Aß therapeutic trials.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Nerve Net/metabolism , Neurons/metabolism , Plaque, Amyloid/metabolism , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Brain/pathology , Disease Models, Animal , Mice , Nerve Net/pathology , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neurons/pathology , Plaque, Amyloid/genetics , tau Proteins/genetics
8.
Brain ; 141(7): 2194-2212, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29733334

ABSTRACT

Several studies have now supported the use of a tau lowering agent as a possible therapy in the treatment of tauopathy disorders, including Alzheimer's disease. In human Alzheimer's disease, however, concurrent amyloid-ß deposition appears to synergize and accelerate tau pathological changes. Thus far, tau reduction strategies that have been tested in vivo have been examined in the setting of tau pathology without confounding amyloid-ß deposition. To determine whether reducing total human tau expression in a transgenic model where there is concurrent amyloid-ß plaque formation can still reduce tau pathology and protect against neuronal loss, we have taken advantage of the regulatable tau transgene in APP/PS1 × rTg4510 mice. These mice develop both neurofibrillary tangles as well as amyloid-ß plaques throughout the cortex and hippocampus. By suppressing human tau expression for 6 months in the APP/PS1 × rTg4510 mice using doxycycline, AT8 tau pathology, bioactivity, and astrogliosis were reduced, though importantly to a lesser extent than lowering tau in the rTg4510 alone mice. Based on non-denaturing gels and proteinase K digestions, the remaining tau aggregates in the presence of amyloid-ß exhibit a longer-lived aggregate conformation. Nonetheless, lowering the expression of the human tau transgene was sufficient to equally ameliorate thioflavin-S positive tangles and prevent neuronal loss equally well in both the APP/PS1 × rTg4510 mice and the rTg4510 cohort. Together, these results suggest that, although amyloid-ß stabilizes tau aggregates, lowering total tau levels is still an effective strategy for the treatment of tau pathology and neuronal loss even in the presence of amyloid-ß deposition.


Subject(s)
Plaque, Amyloid/pathology , Tauopathies/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Hippocampus/metabolism , Humans , Mice , Mice, Transgenic , Neurofibrillary Tangles/pathology , Neurons/metabolism , Phosphorylation , Plaque, Amyloid/metabolism , Presenilin-1/metabolism
9.
Proc Natl Acad Sci U S A ; 114(51): E11029-E11036, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29203673

ABSTRACT

Misfolding of tau proteins into prions and their propagation along neural circuits are thought to result in neurodegeneration causing Alzheimer's disease, progressive supranuclear palsy, chronic traumatic encephalopathy, and other tauopathies. Little is known about the molecular processes mediating tau prion replication and spreading in different brain regions. Using transgenic (Tg) mice with a neuronal promoter driving expression of human mutant (P301S) tau, we found that tau prion formation and histopathologic deposition is largely restricted to the hindbrain. Unexpectedly, tau mRNA and protein levels did not differ between the forebrain and hindbrain, suggesting that other factors modulating the conversion of tau into a prion exist and are region specific. Using a cell-based prion propagation assay, we discovered that tau prion replication is suppressed by forebrain-derived inhibitors, one of which is sortilin, a lysosomal sorting receptor. We also show that sortilin expression is higher in the forebrain than the hindbrain across the life span of the Tg mice, suggesting that sortilin, at least in part, inhibits forebrain tau prion replication in vivo. Our findings provide evidence for selective vulnerability in mice resulting in highly regulated levels of tau prion propagation, thus affording a model for identification of additional molecules that could mitigate the levels of tau prions in human tauopathies.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , tau Proteins/genetics , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Gene Expression , Humans , Mice , Mice, Transgenic , Neurons/metabolism , Phosphorylation , Protein Binding , Rhombencephalon/metabolism , Rhombencephalon/pathology
10.
Acta Neuropathol Commun ; 5(1): 84, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29157304

ABSTRACT

The larger brain of the rat enables a much greater repertoire of complex behaviors than mice, likely making rats preferential for investigating neurodegeneration. Because molecular tools for specific expression of transgenes in the rat brain are sparse, we chose Prnp encoding the prion protein (PrP) to develop a novel vector to drive transgene expression in the rat brain. We compared the rat Prnp sequence with mouse and Syrian hamster Prnp sequences, identifying conserved genetic elements and hypothesizing that these elements would be able to drive neuronal transgene expression. We investigated this by generating a vector termed RaPrnp that encompasses portions of the rat Prnp gene. Importantly, we replaced the rat Prnp open reading frame (ORF) with a cloning site for rapid and seamless In-Fusion cloning. To validate the in vivo neuronal specificity of the RaPrnp vector in rats, we generated stable RaPrnp-LacZ/enhanced green fluorescent protein (EGFP) transgenic (Tg) rat lines, which led to robust LacZ activity and high EGFP fluorescence in the central nervous system of embryos and adult animals. Next, we restored the rat Prnp ORF and generated multiple Tg(RaPrnp-PrP) lines, demonstrating that overexpression of Prnp accelerates the onset of scrapie. While the incubation time in wild-type (WT) rats was 175 ± 3 days post inoculation (dpi), one line, Tg2919, overexpressed RaPrPC at 4.4-fold and exhibited a reduced incubation time of 149 ± 2 dpi. The second line, Tg2922, overexpressed RaPrPC at 9.7-fold compared with WT animals and had an incubation time of 112 ± 0 dpi. Tg2922 rats inoculated with rat RML showed extensive vacuolation of the brainstem in contrast to WT and Tg2919 animals in which vacuolation was most prominent in the hippocampus and striatum as well as the motor and sensory cortices. It is possible that construction of Tg rats with modified phenotypes will prove more advantageous than mice for neurodegeneration studies.


Subject(s)
Central Nervous System/metabolism , Gene Transfer Techniques , Genetic Vectors , Prion Diseases/pathology , Prion Proteins/genetics , Animals , Animals, Genetically Modified , Animals, Newborn , Calcium-Binding Proteins/metabolism , Central Nervous System/pathology , Cricetinae , Disease Models, Animal , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mesocricetus , Mice , Microfilament Proteins/metabolism , Neurons/metabolism , Phosphopyruvate Hydratase/metabolism , Prion Diseases/genetics , Prion Proteins/metabolism , Rats
11.
Prog Mol Biol Transl Sci ; 150: 123-145, 2017.
Article in English | MEDLINE | ID: mdl-28838658

ABSTRACT

Early genetic studies on scrapie, an infectious neurodegenerative disease of sheep that was adapted to mice, provided evidence in support of the hypothesis that the agent was a slow virus with a nucleic acid genome independent of the host. Particularly compelling support for an independent genome came from the existence of strains of scrapie agent, some of which were true breeding, while others appeared to mutate under selective pressure. Kuru, a neurodegenerative disease in the remote highlands of Papua New Guinea, had pathological changes similar to those in scrapie and also proved to be transmissible. Genetic studies with the tools of molecular biology and transgenic mice forced a reevaluation of earlier work and supported the prion hypothesis of a novel pathogen devoid of nucleic acid. In this chapter, I discuss the contributions of classical and molecular genetics to understanding PrP prion diseases and to determining that heritable information is enciphered in protein conformation.


Subject(s)
Genetic Predisposition to Disease , Prion Diseases/genetics , Prion Proteins/chemistry , Prion Proteins/metabolism , Proteostasis Deficiencies/genetics , Animals , Humans , Prion Diseases/therapy , Protein Conformation , Proteostasis Deficiencies/therapy
12.
Am J Pathol ; 187(7): 1601-1612, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28500862

ABSTRACT

Amyloid plaques and neurofibrillary tangles co-occur in Alzheimer disease, but with different topological and temporal patterns. Whether these two lesions are independent or pathobiologically related is uncertain. For example, amyloid deposition in the neocortex precedes the spread of tau neurofibrillary tangles from the limbic areas to the cortex. We examined the aggregation properties of tau isolated from human cases with early tau pathology (Braak II) with and without plaques. Using a well-established HEK cell biosensor assay, we show that tau from cases with plaques has an enhanced ability to induce tau aggregates compared to tau from cases without plaques. To further explore this effect, we combined mice carrying the APP/PS1 transgene array that develop plaques with rTg4510 mice carrying the P301L mutant human tau transgene that develop extensive tau pathology with age. The resulting APP/PS1-rTg4510 mice had a threefold increase in tau seeding activity over the rTg4510 strain, without change in tau production or extracellular release. Surprisingly, this effect was observed before overt amyloid deposition. The enhancement of tau aggregation was also apparent by an increase in histological measures of tau pathology in young APP/PS1-rTg4510 mice and an increase in high-molecular-weight tau. Overall, these data provide evidence that amyloid ß acts to enhance tau pathology by increasing the formation of tau species capable of seeding new aggregates.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , tau Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Neocortex/metabolism , Neocortex/pathology , Neurofibrillary Tangles/metabolism , Phosphorylation , Plaque, Amyloid/metabolism , Protein Aggregation, Pathological , tau Proteins/genetics
13.
Am J Pathol ; 187(6): 1399-1412, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28408124

ABSTRACT

The clinical progression of Alzheimer disease (AD) is associated with the accumulation of tau neurofibrillary tangles, which may spread throughout the cortex by interneuronal tau transfer. If so, targeting extracellular tau species may slow the spreading of tau pathology and possibly cognitive decline. To identify suitable target epitopes, we tested the effects of a panel of tau antibodies on neuronal uptake and aggregation in vitro. Immunodepletion was performed on brain extract from tau-transgenic mice and postmortem AD brain and added to a sensitive fluorescence resonance energy transfer-based tau uptake assay to assess blocking efficacy. The antibodies reduced tau uptake in an epitope-dependent manner: N-terminal (Tau13) and middomain (6C5 and HT7) antibodies successfully prevented uptake of tau species, whereas the distal C-terminal-specific antibody (Tau46) had little effect. Phosphorylation-dependent (40E8 and p396) and C-terminal half (4E4) tau antibodies also reduced tau uptake despite removing less total tau by immunodepletion, suggesting specific interactions with species involved in uptake. Among the seven antibodies evaluated, 6C5 most efficiently blocked uptake and subsequent aggregation. More important, 6C5 also blocked neuron-to-neuron spreading of tau in a unique three-chamber microfluidic device. Furthermore, 6C5 slowed down the progression of tau aggregation even after uptake had begun. Our results imply that not all antibodies/epitopes are equally robust in terms of blocking tau uptake of human AD-derived tau species.


Subject(s)
Alzheimer Disease/metabolism , Neurons/metabolism , tau Proteins/metabolism , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Brain/metabolism , Brain/pathology , Cells, Cultured , Epitopes/immunology , Female , Humans , Interneurons/metabolism , Male , Mice, Transgenic , Microfluidic Analytical Techniques , Molecular Targeted Therapy/methods , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Phosphorylation , tau Proteins/antagonists & inhibitors , tau Proteins/immunology
14.
Article in English | MEDLINE | ID: mdl-28193724

ABSTRACT

Although the discovery of the prion protein (PrP) resulted from its co-purification with scrapie infectivity in Syrian hamsters, work with genetically defined and genetically modified mice proved crucial for understanding the fundamental processes involved not only in prion diseases caused by PrP misfolding, aggregation, and spread but also in other, much more common, neurodegenerative brain diseases. In this review, we focus on methodological and conceptual approaches used to study scrapie and related PrP misfolding diseases in mice and how these approaches have advanced our understanding of related disorders including Alzheimer's and Parkinson's disease.


Subject(s)
Neurodegenerative Diseases/etiology , Prion Diseases/transmission , Animals , Gene Transfer Techniques , Mice , Mice, Transgenic
15.
Dis Model Mech ; 10(5): 605-618, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28167615

ABSTRACT

Hereditary sensory and autonomic neuropathies (HSANs) are a genetically and clinically diverse group of disorders defined by peripheral nervous system (PNS) dysfunction. HSAN type III, known as familial dysautonomia (FD), results from a single base mutation in the gene IKBKAP that encodes a scaffolding unit (ELP1) for a multi-subunit complex known as Elongator. Since mutations in other Elongator subunits (ELP2 to ELP4) are associated with central nervous system (CNS) disorders, the goal of this study was to investigate a potential requirement for Ikbkap in the CNS of mice. The sensory and autonomic pathophysiology of FD is fatal, with the majority of patients dying by age 40. While signs and pathology of FD have been noted in the CNS, the clinical and research focus has been on the sensory and autonomic dysfunction, and no genetic model studies have investigated the requirement for Ikbkap in the CNS. Here, we report, using a novel mouse line in which Ikbkap is deleted solely in the nervous system, that not only is Ikbkap widely expressed in the embryonic and adult CNS, but its deletion perturbs both the development of cortical neurons and their survival in adulthood. Primary cilia in embryonic cortical apical progenitors and motile cilia in adult ependymal cells are reduced in number and disorganized. Furthermore, we report that, in the adult CNS, both autonomic and non-autonomic neuronal populations require Ikbkap for survival, including spinal motor and cortical neurons. In addition, the mice developed kyphoscoliosis, an FD hallmark, indicating its neuropathic etiology. Ultimately, these perturbations manifest in a developmental and progressive neurodegenerative condition that includes impairments in learning and memory. Collectively, these data reveal an essential function for Ikbkap that extends beyond the peripheral nervous system to CNS development and function. With the identification of discrete CNS cell types and structures that depend on Ikbkap, novel strategies to thwart the progressive demise of CNS neurons in FD can be developed.


Subject(s)
Carrier Proteins/genetics , Central Nervous System/metabolism , Dysautonomia, Familial/genetics , Animals , Behavior, Animal , Cell Survival/genetics , Central Nervous System/growth & development , Central Nervous System/pathology , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Mutation , Neurons/pathology
16.
Eur J Neurosci ; 44(12): 3056-3066, 2016 12.
Article in English | MEDLINE | ID: mdl-27748574

ABSTRACT

Alzheimer's disease is characterized by the presence of aggregates of amyloid beta (Aß) in senile plaques and tau in neurofibrillary tangles, as well as marked neuron and synapse loss. Of these pathological changes, synapse loss correlates most strongly with cognitive decline. Synapse loss occurs prominently around plaques due to accumulations of oligomeric Aß. Recent evidence suggests that tau may also play a role in synapse loss but the interactions of Aß and tau in synapse loss remain to be determined. In this study, we generated a novel transgenic mouse line, the APP/PS1/rTg21221 line, by crossing APP/PS1 mice, which develop Aß-plaques and synapse loss, with rTg21221 mice, which overexpress wild-type human tau. When compared to the APP/PS1 mice without human tau, the cross-sectional area of ThioS+ dense core plaques was increased by ~50%. Along with increased plaque size, we observed an increase in plaque-associated dystrophic neurites containing misfolded tau, but there was no exacerbation of neurite curvature or local neuron loss around plaques. Array tomography analysis similarly revealed no worsening of synapse loss around plaques, and no change in the accumulation of Aß at synapses. Together, these results indicate that adding human wild-type tau exacerbates plaque pathology and neurite deformation but does not exacerbate plaque-associated synapse loss.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Disease Models, Animal , Plaque, Amyloid/metabolism , Synapses/metabolism , Synapses/pathology , tau Proteins/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Astrocytes/metabolism , Humans , Mice , Mice, Transgenic , Neurons/metabolism , Phosphorylation , Presenilin-1/genetics , Presenilin-1/metabolism , tau Proteins/genetics
17.
Cell Chem Biol ; 23(8): 992-1001, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27499529

ABSTRACT

Heat shock protein 70 (Hsp70) is a chaperone that normally scans the proteome and initiates the turnover of some proteins (termed clients) by linking them to the degradation pathways. This activity is critical to normal protein homeostasis, yet it appears to fail in diseases associated with abnormal protein accumulation. It is not clear why Hsp70 promotes client degradation under some conditions, while sparing that protein under others. Here, we used a combination of chemical biology and genetic strategies to systematically perturb the affinity of Hsp70 for the model client, tau. This approach revealed that tight complexes between Hsp70 and tau were associated with enhanced turnover while transient interactions favored tau retention. These results suggest that client affinity is one important parameter governing Hsp70-mediated quality control.


Subject(s)
Benzothiazoles/pharmacology , HSP70 Heat-Shock Proteins/metabolism , Models, Biological , Tauopathies/drug therapy , Tauopathies/metabolism , Thiazolidines/pharmacology , tau Proteins/metabolism , Benzothiazoles/chemistry , Dose-Response Relationship, Drug , HSP70 Heat-Shock Proteins/chemistry , HeLa Cells , Humans , Molecular Structure , Protein Stability/drug effects , Structure-Activity Relationship , Thiazolidines/chemistry , Tumor Cells, Cultured , tau Proteins/chemistry
18.
PLoS One ; 11(7): e0159377, 2016.
Article in English | MEDLINE | ID: mdl-27441645

ABSTRACT

ENU mutagenesis is a powerful method for generating novel lines of mice that are informative with respect to both fundamental biological processes and human disease. Rapid developments in genomic technology have made the task of identifying causal mutations by positional cloning remarkably efficient. One limitation of this approach remains the mutation frequency achievable using standard treatment protocols, which currently generate approximately 1-2 sequence changes per megabase when optimized. In this study we used two strategies to attempt to increase the number of mutations induced by ENU treatment. One approach employed mice carrying a mutation in the DNA repair enzyme Msh6. The second strategy involved injection of ENU to successive generations of mice. To evaluate the number of ENU-induced mutations, single mice or pooled samples were analyzed using whole exome sequencing. The results showed that there is considerable variability in the induced mutation frequency using these approaches, but an overall increase in ENU-induced variants from one generation to another was observed. The analysis of the mice deficient for Msh6 also showed an increase in the ENU-induced variants compared to the wild-type ENU-treated mice. However, in both cases the increase in ENU-induced mutation frequency was modest.


Subject(s)
DNA Mismatch Repair/genetics , Ethylnitrosourea/chemistry , Mutagenesis/genetics , Animals , Base Pairing/genetics , Breeding , Female , Fertility , Mice, Inbred C57BL , Mice, Inbred ICR , Mutation/genetics , Survival Analysis
19.
Ann Neurol ; 80(3): 355-67, 2016 09.
Article in English | MEDLINE | ID: mdl-27351289

ABSTRACT

OBJECTIVE: Cerebrospinal fluid (CSF) tau is an excellent surrogate marker for assessing neuropathological changes that occur in Alzheimer's disease (AD) patients. However, whether the elevated tau in AD CSF is just a marker of neurodegeneration or, in fact, a part of the disease process is uncertain. Moreover, it is unknown how CSF tau relates to the recently described soluble high-molecular-weight (HMW) species that is found in the postmortem AD brain and can be taken up by neurons and seed aggregates. METHODS: We have examined seeding and uptake properties of brain extracellular tau from various sources, including interstitial fluid (ISF) and CSF from an AD transgenic mouse model and postmortem ventricular and antemortem lumbar CSF from AD patients. RESULTS: We found that brain ISF and CSF tau from the AD mouse model can be taken up by cells and induce intracellular aggregates. Ventricular CSF from AD patients contained a rare HMW tau species that exerted a higher seeding activity. Notably, the HMW tau species was also detected in lumbar CSF from AD patients, and its levels were significantly elevated compared to control subjects. HMW tau derived from CSF of AD patients was seed competent in vitro. INTERPRETATION: These findings suggest that CSF from an AD brain contains potentially bioactive HMW tau species, giving new insights into the role of CSF tau and biomarker development for AD. Ann Neurol 2016;80:355-367.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Brain/metabolism , tau Proteins/cerebrospinal fluid , Aged , Animals , Biomarkers/cerebrospinal fluid , Extracellular Fluid/metabolism , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged
20.
PLoS One ; 11(2): e0149410, 2016.
Article in English | MEDLINE | ID: mdl-26894278

ABSTRACT

Pituitary Prolactin (PRL) and Growth Hormone (GH) are separately controlled and sub-serve different purposes. Surprisingly, we demonstrate that extra-pituitary expression in the adult mammalian central nervous system (CNS) is coordinated at mRNA and protein levels. However this was not a uniform effect within populations, such that wide inter-individual variation was superimposed on coordinate PRL/GH expression. Up to 44% of individuals in healthy cohorts of mice and rats showed protein levels above the norm and coordinated expression of PRL and GH transcripts above baseline occurred in the amygdala, frontal lobe and hippocampus of 10% of human subjects. High levels of PRL and GH present in post mortem tissue were often presaged by altered responses in fear conditioning and stress induced hyperthermia behavioral tests. Our data define a common phenotype polymorphism in healthy mammalian brains, and, given the pleiotropic effects known for circulating PRL and GH, further consequences of coordinated CNS over-expression may await discovery.


Subject(s)
Brain/metabolism , Growth Hormone/biosynthesis , Phenotype , Prolactin/biosynthesis , Animals , Female , Gene Expression , Growth Hormone/blood , Growth Hormone/genetics , Humans , Male , Mice , Prolactin/blood , Prolactin/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...