Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Cell ; 174(3): 505-520, 2018 07 26.
Article in English | MEDLINE | ID: mdl-30053424

ABSTRACT

Although gene discovery in neuropsychiatric disorders, including autism spectrum disorder, intellectual disability, epilepsy, schizophrenia, and Tourette disorder, has accelerated, resulting in a large number of molecular clues, it has proven difficult to generate specific hypotheses without the corresponding datasets at the protein complex and functional pathway level. Here, we describe one path forward-an initiative aimed at mapping the physical and genetic interaction networks of these conditions and then using these maps to connect the genomic data to neurobiology and, ultimately, the clinic. These efforts will include a team of geneticists, structural biologists, neurobiologists, systems biologists, and clinicians, leveraging a wide array of experimental approaches and creating a collaborative infrastructure necessary for long-term investigation. This initiative will ultimately intersect with parallel studies that focus on other diseases, as there is a significant overlap with genes implicated in cancer, infectious disease, and congenital heart defects.


Subject(s)
Chromosome Mapping/methods , Neurodevelopmental Disorders/genetics , Systems Biology/methods , Gene Regulatory Networks/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Genomics/methods , Humans , Neurobiology/methods , Neuropsychiatry
2.
Nat Immunol ; 19(11): 1212-1223, 2018 11.
Article in English | MEDLINE | ID: mdl-30323343

ABSTRACT

Activation of innate immunity and deposition of blood-derived fibrin in the central nervous system (CNS) occur in autoimmune and neurodegenerative diseases, including multiple sclerosis (MS) and Alzheimer's disease (AD). However, the mechanisms that link disruption of the blood-brain barrier (BBB) to neurodegeneration are poorly understood, and exploration of fibrin as a therapeutic target has been limited by its beneficial clotting functions. Here we report the generation of monoclonal antibody 5B8, targeted against the cryptic fibrin epitope γ377-395, to selectively inhibit fibrin-induced inflammation and oxidative stress without interfering with clotting. 5B8 suppressed fibrin-induced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activation and the expression of proinflammatory genes. In animal models of MS and AD, 5B8 entered the CNS and bound to parenchymal fibrin, and its therapeutic administration reduced the activation of innate immunity and neurodegeneration. Thus, fibrin-targeting immunotherapy inhibited autoimmunity- and amyloid-driven neurotoxicity and might have clinical benefit without globally suppressing innate immunity or interfering with coagulation in diverse neurological diseases.


Subject(s)
Antibodies, Monoclonal/immunology , Fibrinogen/antagonists & inhibitors , Neurodegenerative Diseases/immunology , Animals , Epitopes , Humans , Inflammation/immunology , Mice , Rats
3.
Cell ; 148(6): 1204-22, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22424230

ABSTRACT

There are still no effective treatments to prevent, halt, or reverse Alzheimer's disease, but research advances over the past three decades could change this gloomy picture. Genetic studies demonstrate that the disease has multiple causes. Interdisciplinary approaches combining biochemistry, molecular and cell biology, and transgenic modeling have revealed some of its molecular mechanisms. Progress in chemistry, radiology, and systems biology is beginning to provide useful biomarkers, and the emergence of personalized medicine is poised to transform pharmaceutical development and clinical trials. However, investigative and drug development efforts should be diversified to fully address the multifactoriality of the disease.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Animals , Cognition , Humans , gamma-Aminobutyric Acid/metabolism
4.
Cell ; 149(3): 708-21, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22541439

ABSTRACT

Alzheimer's disease (AD) results in cognitive decline and altered network activity, but the mechanisms are unknown. We studied human amyloid precursor protein (hAPP) transgenic mice, which simulate key aspects of AD. Electroencephalographic recordings in hAPP mice revealed spontaneous epileptiform discharges, indicating network hypersynchrony, primarily during reduced gamma oscillatory activity. Because this oscillatory rhythm is generated by inhibitory parvalbumin (PV) cells, network dysfunction in hAPP mice might arise from impaired PV cells. Supporting this hypothesis, hAPP mice and AD patients had decreased levels of the interneuron-specific and PV cell-predominant voltage-gated sodium channel subunit Nav1.1. Restoring Nav1.1 levels in hAPP mice by Nav1.1-BAC expression increased inhibitory synaptic activity and gamma oscillations and reduced hypersynchrony, memory deficits, and premature mortality. We conclude that reduced Nav1.1 levels and PV cell dysfunction critically contribute to abnormalities in oscillatory rhythms, network synchrony, and memory in hAPP mice and possibly in AD.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Hippocampus/metabolism , Humans , In Vitro Techniques , Interneurons/metabolism , Learning , Memory , Mice , Mice, Inbred C57BL , Mice, Transgenic , NAV1.1 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Sodium Channels/metabolism , Synapses
5.
Neurobiol Dis ; 186: 106263, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37591465

ABSTRACT

The R47H variant of triggering receptor expressed on myeloid cells 2 (TREM2) increases the risk of Alzheimer's disease (AD). To investigate potential mechanisms, we analyzed knockin mice expressing human TREM2-R47H from one mutant mouse Trem2 allele. TREM2-R47H mice showed increased seizure activity in response to an acute excitotoxin challenge, compared to wildtype controls or knockin mice expressing the common variant of human TREM2. TREM2-R47H also increased spontaneous thalamocortical epileptiform activity in App knockin mice expressing amyloid precursor proteins bearing autosomal dominant AD mutations and a humanized amyloid-ß sequence. In mice with or without such App modifications, TREM2-R47H increased the density of putative synapses in cortical regions without amyloid plaques. TREM2-R47H did not affect synaptic density in hippocampal regions with or without plaques. We conclude that TREM2-R47H increases AD-related network hyperexcitability and that it may do so, at least in part, by causing an imbalance in synaptic densities across brain regions.


Subject(s)
Alzheimer Disease , Humans , Animals , Mice , Alzheimer Disease/genetics , Alleles , Seizures , Amyloid beta-Peptides , Disease Models, Animal , Plaque, Amyloid , Synapses , Membrane Glycoproteins/genetics , Receptors, Immunologic/genetics
6.
Nat Rev Neurosci ; 17(12): 777-792, 2016 12.
Article in English | MEDLINE | ID: mdl-27829687

ABSTRACT

The function of neural circuits and networks can be controlled, in part, by modulating the synchrony of their components' activities. Network hypersynchrony and altered oscillatory rhythmic activity may contribute to cognitive abnormalities in Alzheimer disease (AD). In this condition, network activities that support cognition are altered decades before clinical disease onset, and these alterations predict future pathology and brain atrophy. Although the precise causes and pathophysiological consequences of these network alterations remain to be defined, interneuron dysfunction and network abnormalities have emerged as potential mechanisms of cognitive dysfunction in AD and related disorders. Here, we explore the concept that modulating these mechanisms may help to improve brain function in these conditions.


Subject(s)
Alzheimer Disease/physiopathology , Brain/physiopathology , Interneurons/physiology , Nerve Net/physiopathology , Alzheimer Disease/pathology , Animals , Brain/pathology , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Humans , Interneurons/pathology , Nerve Net/pathology , Neural Pathways/pathology , Neural Pathways/physiopathology
8.
Proc Natl Acad Sci U S A ; 115(48): E11388-E11396, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30413620

ABSTRACT

Located within the brain's ventricles, the choroid plexus produces cerebrospinal fluid and forms an important barrier between the central nervous system and the blood. For unknown reasons, the choroid plexus produces high levels of the protein klotho. Here, we show that these levels naturally decline with aging. Depleting klotho selectively from the choroid plexus via targeted viral vector-induced knockout in Klothoflox/flox mice increased the expression of multiple proinflammatory factors and triggered macrophage infiltration of this structure in young mice, simulating changes in unmanipulated old mice. Wild-type mice infected with the same Cre recombinase-expressing virus did not show such alterations. Experimental depletion of klotho from the choroid plexus enhanced microglial activation in the hippocampus after peripheral injection of mice with lipopolysaccharide. In primary cultures, klotho suppressed thioredoxin-interacting protein-dependent activation of the NLRP3 inflammasome in macrophages by enhancing fibroblast growth factor 23 signaling. We conclude that klotho functions as a gatekeeper at the interface between the brain and immune system in the choroid plexus. Klotho depletion in aging or disease may weaken this barrier and promote immune-mediated neuropathogenesis.


Subject(s)
Aging/immunology , Brain/immunology , Choroid Plexus/immunology , Glucuronidase/immunology , Aging/genetics , Animals , Female , Glucuronidase/genetics , Hippocampus/immunology , Humans , Klotho Proteins , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology
9.
Neurobiol Dis ; 117: 181-188, 2018 09.
Article in English | MEDLINE | ID: mdl-29859869

ABSTRACT

Neural network dysfunction may contribute to functional decline and disease progression in neurodegenerative disorders. Diverse lines of evidence suggest that neuronal accumulation of tau promotes network dysfunction and cognitive decline. The A152T-variant of human tau (hTau-A152T) increases the risk of Alzheimer's disease (AD) and several other tauopathies. When overexpressed in neurons of transgenic mice, it causes age-dependent neuronal loss and cognitive decline, as well as non-convulsive epileptic activity, which is also seen in patients with AD. Using intracranial EEG recordings with electrodes implanted over the parietal cortex, we demonstrate that hTau-A152T increases the power of brain oscillations in the 0.5-6 Hz range more than wildtype human tau in transgenic lines with comparable levels of human tau protein in brain, and that genetic ablation of endogenous tau in Mapt-/- mice decreases the power of these oscillations as compared to wildtype controls. Suppression of hTau-A152T production in doxycycline-regulatable transgenic mice reversed their abnormal network activity. Treatment of hTau-A152T mice with the antiepileptic drug levetiracetam also rapidly and persistently reversed their brain dysrhythmia and network hypersynchrony. These findings suggest that both the level and the sequence of tau modulate the power of specific brain oscillations. The potential of EEG spectral changes as a biomarker deserves to be explored in clinical trials of tau-lowering therapeutics. Our results also suggest that levetiracetam treatment is able to counteract tau-dependent neural network dysfunction. Tau reduction and levetiracetam treatment may be of benefit in AD and other conditions associated with brain dysrhythmias and network hypersynchrony.


Subject(s)
Brain/metabolism , Delta Rhythm/physiology , Neurons/metabolism , Theta Rhythm/physiology , tau Proteins/metabolism , Animals , Brain/pathology , Brain Waves/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/pathology
10.
Neurobiol Dis ; 110: 29-36, 2018 02.
Article in English | MEDLINE | ID: mdl-29100987

ABSTRACT

Adenosine A2A receptors are putative therapeutic targets for neurological disorders. The adenosine A2A receptor antagonist istradefylline is approved in Japan for Parkinson's disease and is being tested in clinical trials for this condition elsewhere. A2A receptors on neurons and astrocytes may contribute to Alzheimer's disease (AD) by impairing memory. However, it is not known whether istradefylline enhances cognitive function in aging animals with AD-like amyloid plaque pathology. Here, we show that elevated levels of Aß, C-terminal fragments of the amyloid precursor protein (APP), or amyloid plaques, but not overexpression of APP per se, increase astrocytic A2A receptor levels in the hippocampus and neocortex of aging mice. Moreover, in amyloid plaque-bearing mice, low-dose istradefylline treatment enhanced spatial memory and habituation, supporting the conclusion that, within a well-defined dose range, A2A receptor blockers might help counteract memory problems in patients with Alzheimer's disease.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Astrocytes/metabolism , Brain/drug effects , Memory Disorders , Purines/pharmacology , Aging , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides , Animals , Brain/pathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/pathology , Receptor, Adenosine A2A
11.
EMBO Rep ; 17(4): 530-51, 2016 04.
Article in English | MEDLINE | ID: mdl-26931567

ABSTRACT

A152T-variant human tau (hTau-A152T) increases risk for tauopathies, including Alzheimer's disease. Comparing mice with regulatable expression of hTau-A152T or wild-type hTau (hTau-WT), we find age-dependent neuronal loss, cognitive impairments, and spontaneous nonconvulsive epileptiform activity primarily in hTau-A152T mice. However, overexpression of either hTau species enhances neuronal responses to electrical stimulation of synaptic inputs and to an epileptogenic chemical. hTau-A152T mice have higher hTau protein/mRNA ratios in brain, suggesting that A152T increases production or decreases clearance of hTau protein. Despite their functional abnormalities, aging hTau-A152T mice show no evidence for accumulation of insoluble tau aggregates, suggesting that their dysfunctions are caused by soluble tau. In human amyloid precursor protein (hAPP) transgenic mice, co-expression of hTau-A152T enhances risk of early death and epileptic activity, suggesting copathogenic interactions between hTau-A152T and amyloid-ß peptides or other hAPP metabolites. Thus, the A152T substitution may augment risk for neurodegenerative diseases by increasing hTau protein levels, promoting network hyperexcitability, and synergizing with the adverse effects of other pathogenic factors.


Subject(s)
Aging , Neurons/pathology , tau Proteins/genetics , tau Proteins/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Frontotemporal Dementia/metabolism , Humans , Mice , Mice, Transgenic , Tauopathies/genetics , Tauopathies/physiopathology , tau Proteins/chemistry
12.
J Biol Chem ; 291(4): 1719-1734, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26589795

ABSTRACT

Diverse lines of evidence suggest that amyloid-ß (Aß) peptides causally contribute to the pathogenesis of Alzheimer disease (AD), the most frequent neurodegenerative disorder. However, the mechanisms by which Aß impairs neuronal functions remain to be fully elucidated. Previous studies showed that soluble Aß oligomers interfere with synaptic functions by depleting NMDA-type glutamate receptors (NMDARs) from the neuronal surface and that overexpression of the receptor tyrosine kinase EphB2 can counteract this process. Through pharmacological treatments and biochemical analyses of primary neuronal cultures expressing wild-type or mutant forms of EphB2, we demonstrate that this protective effect of EphB2 depends on its PDZ-binding motif and the presence of neuronal activity but not on its kinase activity. We further present evidence that the protective effect of EphB2 may be mediated by the AMPA-type glutamate receptor subunit GluA2, which can become associated with the PDZ-binding motif of EphB2 through PDZ domain-containing proteins and can promote the retention of NMDARs in the membrane. In addition, we show that the Aß-induced depletion of surface NMDARs does not depend on several factors that have been implicated in the pathogenesis of Aß-induced neuronal dysfunction, including aberrant neuronal activity, tau, prion protein (PrP(C)), and EphB2 itself. Thus, although EphB2 does not appear to be directly involved in the Aß-induced depletion of NMDARs, increasing its expression may counteract this pathogenic process through a neuronal activity- and PDZ-dependent regulation of AMPA-type glutamate receptors.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptor, EphB2/chemistry , Receptor, EphB2/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Alzheimer Disease/genetics , Amino Acid Motifs , Animals , Cells, Cultured , Female , Hippocampus/cytology , Hippocampus/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Neurons/chemistry , PDZ Domains , Protein Binding , Receptor, EphB2/genetics , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/genetics
13.
Ann Neurol ; 80(6): 858-870, 2016 12.
Article in English | MEDLINE | ID: mdl-27696483

ABSTRACT

OBJECTIVE: Seizures are more frequent in patients with Alzheimer's disease (AD) and can hasten cognitive decline. However, the incidence of subclinical epileptiform activity in AD and its consequences are unknown. Motivated by results from animal studies, we hypothesized higher than expected rates of subclinical epileptiform activity in AD with deleterious effects on cognition. METHODS: We prospectively enrolled 33 patients (mean age, 62 years) who met criteria for AD, but had no history of seizures, and 19 age-matched, cognitively normal controls. Subclinical epileptiform activity was assessed, blinded to diagnosis, by overnight long-term video-electroencephalography (EEG) and a 1-hour resting magnetoencephalography exam with simultaneous EEG. Patients also had comprehensive clinical and cognitive evaluations, assessed longitudinally over an average period of 3.3 years. RESULTS: Subclinical epileptiform activity was detected in 42.4% of AD patients and 10.5% of controls (p = 0.02). At the time of monitoring, AD patients with epileptiform activity did not differ clinically from those without such activity. However, patients with subclinical epileptiform activity showed faster declines in global cognition, determined by the Mini-Mental State Examination (3.9 points/year in patients with epileptiform activity vs 1.6 points/year in patients without; p = 0.006), and in executive function (p = 0.01). INTERPRETATION: Extended monitoring detects subclinical epileptiform activity in a substantial proportion of patients with AD. Patients with this indicator of network hyperexcitability are at risk for accelerated cognitive decline and might benefit from antiepileptic therapies. These data call for more sensitive and comprehensive neurophysiological assessments in AD patient evaluations and impending clinical trials. Ann Neurol 2016;80:858-870.


Subject(s)
Alzheimer Disease/epidemiology , Seizures/epidemiology , California/epidemiology , Case-Control Studies , Comorbidity , Electroencephalography , Female , Humans , Incidence , Magnetic Resonance Imaging , Magnetoencephalography , Male , Middle Aged , Neuroimaging , Neuropsychological Tests , Prodromal Symptoms , Prospective Studies
14.
Nature ; 469(7328): 47-52, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-21113149

ABSTRACT

Amyloid-ß oligomers may cause cognitive deficits in Alzheimer's disease by impairing neuronal NMDA-type glutamate receptors, whose function is regulated by the receptor tyrosine kinase EphB2. Here we show that amyloid-ß oligomers bind to the fibronectin repeats domain of EphB2 and trigger EphB2 degradation in the proteasome. To determine the pathogenic importance of EphB2 depletions in Alzheimer's disease and related models, we used lentiviral constructs to reduce or increase neuronal expression of EphB2 in memory centres of the mouse brain. In nontransgenic mice, knockdown of EphB2 mediated by short hairpin RNA reduced NMDA receptor currents and impaired long-term potentiation in the dentate gyrus, which are important for memory formation. Increasing EphB2 expression in the dentate gyrus of human amyloid precursor protein transgenic mice reversed deficits in NMDA receptor-dependent long-term potentiation and memory impairments. Thus, depletion of EphB2 is critical in amyloid-ß-induced neuronal dysfunction. Increasing EphB2 levels or function could be beneficial in Alzheimer's disease.


Subject(s)
Alzheimer Disease/physiopathology , Alzheimer Disease/therapy , Cognition/physiology , Receptor, EphB2/deficiency , Receptor, EphB2/metabolism , Amyloid beta-Peptides/metabolism , Animals , Cell Line , Cells, Cultured , Dentate Gyrus/metabolism , Disease Models, Animal , Humans , Long-Term Potentiation , Memory/physiology , Mice , Mice, Transgenic , Neuronal Plasticity , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Structure, Tertiary , Rats , Receptor, EphB2/chemistry , Receptor, EphB2/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism
15.
J Neurosci ; 35(6): 2358-71, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25673831

ABSTRACT

Aging is the principal demographic risk factor for Alzheimer disease (AD), the most common neurodegenerative disorder. Klotho is a key modulator of the aging process and, when overexpressed, extends mammalian lifespan, increases synaptic plasticity, and enhances cognition. Whether klotho can counteract deficits related to neurodegenerative diseases, such as AD, is unknown. Here we show that elevating klotho expression decreases premature mortality and network dysfunction in human amyloid precursor protein (hAPP) transgenic mice, which simulate key aspects of AD. Increasing klotho levels prevented depletion of NMDA receptor (NMDAR) subunits in the hippocampus and enhanced spatial learning and memory in hAPP mice. Klotho elevation in hAPP mice increased the abundance of the GluN2B subunit of NMDAR in postsynaptic densities and NMDAR-dependent long-term potentiation, which is critical for learning and memory. Thus, increasing wild-type klotho levels or activities improves synaptic and cognitive functions, and may be of therapeutic benefit in AD and other cognitive disorders.


Subject(s)
Amyloid beta-Protein Precursor/physiology , Cognition/physiology , Glucuronidase/physiology , Longevity/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal/physiology , Cognition Disorders/genetics , Cognition Disorders/psychology , Humans , Klotho Proteins , Longevity/physiology , Maze Learning/physiology , Mice , Mice, Transgenic , Nerve Net/pathology , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/pathology , tau Proteins/metabolism
16.
Ann Neurol ; 76(3): 443-56, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25042160

ABSTRACT

OBJECTIVE: Reducing levels of the microtubule-associated protein tau has shown promise as a potential treatment strategy for diseases with secondary epileptic features such as Alzheimer disease. We wanted to determine whether tau reduction may also be of benefit in intractable genetic epilepsies. METHODS: We studied a mouse model of Dravet syndrome, a severe childhood epilepsy caused by mutations in the human SCN1A gene encoding the voltage-gated sodium channel subunit Nav 1.1. We genetically deleted 1 or 2 Tau alleles in mice carrying an Nav 1.1 truncation mutation (R1407X) that causes Dravet syndrome in humans, and examined their survival, epileptic activity, related hippocampal alterations, and behavioral abnormalities using observation, electroencephalographic recordings, acute slice electrophysiology, immunohistochemistry, and behavioral assays. RESULTS: Tau ablation prevented the high mortality of Dravet mice and reduced the frequency of spontaneous and febrile seizures. It reduced interictal epileptic spikes in vivo and drug-induced epileptic activity in brain slices ex vivo. Tau ablation also prevented biochemical changes in the hippocampus indicative of epileptic activity and ameliorated abnormalities in learning and memory, nest building, and open field behaviors in Dravet mice. Deletion of only 1 Tau allele was sufficient to suppress epileptic activity and improve survival and nesting performance. INTERPRETATION: Tau reduction may be of therapeutic benefit in Dravet syndrome and other intractable genetic epilepsies.


Subject(s)
Epilepsies, Myoclonic/metabolism , Hippocampus/metabolism , Seizures/metabolism , tau Proteins/metabolism , Alleles , Animals , Behavior, Animal/physiology , Disease Models, Animal , Electroencephalography , Epilepsies, Myoclonic/physiopathology , Epilepsies, Myoclonic/therapy , Female , Hippocampus/pathology , Hippocampus/physiopathology , Learning/physiology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Seizures/etiology , Seizures/physiopathology , tau Proteins/genetics
17.
Proc Natl Acad Sci U S A ; 109(42): E2895-903, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-22869752

ABSTRACT

In light of the rising prevalence of Alzheimer's disease (AD), new strategies to prevent, halt, and reverse this condition are needed urgently. Perturbations of brain network activity are observed in AD patients and in conditions that increase the risk of developing AD, suggesting that aberrant network activity might contribute to AD-related cognitive decline. Human amyloid precursor protein (hAPP) transgenic mice simulate key aspects of AD, including pathologically elevated levels of amyloid-ß peptides in brain, aberrant neural network activity, remodeling of hippocampal circuits, synaptic deficits, and behavioral abnormalities. Whether these alterations are linked in a causal chain remains unknown. To explore whether hAPP/amyloid-ß-induced aberrant network activity contributes to synaptic and cognitive deficits, we treated hAPP mice with different antiepileptic drugs. Among the drugs tested, only levetiracetam (LEV) effectively reduced abnormal spike activity detected by electroencephalography. Chronic treatment with LEV also reversed hippocampal remodeling, behavioral abnormalities, synaptic dysfunction, and deficits in learning and memory in hAPP mice. Our findings support the hypothesis that aberrant network activity contributes causally to synaptic and cognitive deficits in hAPP mice. LEV might also help ameliorate related abnormalities in people who have or are at risk for AD.


Subject(s)
Alzheimer Disease/drug therapy , Anticonvulsants/pharmacology , Cognition Disorders/drug therapy , Cognition/drug effects , Nerve Net/drug effects , Piracetam/analogs & derivatives , Synapses/drug effects , Alzheimer Disease/complications , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Analysis of Variance , Animals , Anticonvulsants/blood , Anticonvulsants/therapeutic use , Blotting, Western , Cognition Disorders/etiology , Electroencephalography , Humans , Immunohistochemistry , Levetiracetam , Maze Learning/drug effects , Mice , Mice, Transgenic , Nerve Net/physiopathology , Piracetam/blood , Piracetam/pharmacology , Piracetam/therapeutic use
18.
bioRxiv ; 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38328105

ABSTRACT

Clustering is a critical step in the analysis of single-cell data, as it enables the discovery and characterization of putative cell types and states. However, most popular clustering tools do not subject clustering results to statistical inference testing, leading to risks of overclustering or underclustering data and often resulting in ineffective identification of cell types with widely differing prevalence. To address these challenges, we present CHOIR (clustering hierarchy optimization by iterative random forests), which applies a framework of random forest classifiers and permutation tests across a hierarchical clustering tree to statistically determine which clusters represent distinct populations. We demonstrate the enhanced performance of CHOIR through extensive benchmarking against 14 existing clustering methods across 100 simulated and 4 real single-cell RNA-seq, ATAC-seq, spatial transcriptomic, and multi-omic datasets. CHOIR can be applied to any single-cell data type and provides a flexible, scalable, and robust solution to the important challenge of identifying biologically relevant cell groupings within heterogeneous single-cell data.

19.
J Neurosci ; 31(29): 10427-31, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21775587

ABSTRACT

Previous studies suggested that the cellular prion protein (PrP(c)) plays a critical role in the pathogenesis of Alzheimer's disease (AD). Specifically, amyloid-ß (Aß) oligomers were proposed to cause synaptic and cognitive dysfunction by binding to PrP(c). To test this hypothesis, we crossed human amyloid precursor protein (hAPP) transgenic mice from line J20 onto a PrP(c)-deficient background. Ablation of PrP(c) did not prevent the premature mortality and abnormal neural network activity typically seen in hAPPJ20 mice. Furthermore, hAPPJ20 mice with or without PrP(c) expression showed comparably robust abnormalities in learning and memory and in other behavioral domains at 6-8 months of age. Notably, these abnormalities are not refractory to therapeutic manipulations in general: they can be effectively prevented by interventions that prevent Aß-dependent neuronal dysfunction also in other lines of hAPP transgenic mice. Thus, at least in this model, PrP(c) is not an important mediator of Aß-induced neurological impairments.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Cognition Disorders/genetics , Cognition Disorders/prevention & control , PrPSc Proteins/metabolism , Age Factors , Amyloid beta-Peptides/metabolism , Analysis of Variance , Animals , Animals, Genetically Modified , Chi-Square Distribution , Cognition Disorders/mortality , Cognition Disorders/physiopathology , Disease Models, Animal , Electroencephalography/methods , Enzyme-Linked Immunosorbent Assay/methods , Exploratory Behavior/physiology , Humans , Kaplan-Meier Estimate , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Peptide Fragments/metabolism , PrPSc Proteins/genetics
20.
J Neurosci ; 31(2): 700-11, 2011 Jan 12.
Article in English | MEDLINE | ID: mdl-21228179

ABSTRACT

Alzheimer's disease (AD), the most common neurodegenerative disorder, is a growing public health problem and still lacks effective treatments. Recent evidence suggests that microtubule-associated protein tau may mediate amyloid-ß peptide (Aß) toxicity by modulating the tyrosine kinase Fyn. We showed previously that tau reduction prevents, and Fyn overexpression exacerbates, cognitive deficits in human amyloid precursor protein (hAPP) transgenic mice overexpressing Aß. However, the mechanisms by which Aß, tau, and Fyn cooperate in AD-related pathogenesis remain to be fully elucidated. Here we examined the synaptic and network effects of this pathogenic triad. Tau reduction prevented cognitive decline induced by synergistic effects of Aß and Fyn. Tau reduction also prevented synaptic transmission and plasticity deficits in hAPP mice. Using electroencephalography to examine network effects, we found that tau reduction prevented spontaneous epileptiform activity in multiple lines of hAPP mice. Tau reduction also reduced the severity of spontaneous and chemically induced seizures in mice overexpressing both Aß and Fyn. To better understand these protective effects, we recorded whole-cell currents in acute hippocampal slices from hAPP mice with and without tau. hAPP mice with tau had increased spontaneous and evoked excitatory currents, reduced inhibitory currents, and NMDA receptor dysfunction. Tau reduction increased inhibitory currents and normalized excitation/inhibition balance and NMDA receptor-mediated currents in hAPP mice. Our results indicate that Aß, tau, and Fyn jointly impair synaptic and network function and suggest that disrupting the copathogenic relationship between these factors could be of therapeutic benefit.


Subject(s)
Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/physiology , Cognition Disorders/physiopathology , Nerve Net/physiology , Proto-Oncogene Proteins c-fyn/physiology , Synapses/physiology , tau Proteins/metabolism , Alzheimer Disease/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/mortality , Animals , Cognition Disorders/metabolism , Cognition Disorders/psychology , Disease Models, Animal , Electroencephalography , Female , Hippocampus/physiopathology , In Vitro Techniques , Male , Mice , Mice, Mutant Strains , Neuronal Plasticity , Seizures/metabolism , Seizures/physiopathology , Species Specificity , Synaptic Transmission , tau Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL