Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Cell ; 186(20): 4438-4453.e23, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37774681

ABSTRACT

Cellular perturbations underlying Alzheimer's disease (AD) are primarily studied in human postmortem samples and model organisms. Here, we generated a single-nucleus atlas from a rare cohort of cortical biopsies from living individuals with varying degrees of AD pathology. We next performed a systematic cross-disease and cross-species integrative analysis to identify a set of cell states that are specific to early AD pathology. These changes-which we refer to as the early cortical amyloid response-were prominent in neurons, wherein we identified a transitional hyperactive state preceding the loss of excitatory neurons, which we confirmed by acute slice physiology on independent biopsy specimens. Microglia overexpressing neuroinflammatory-related processes also expanded as AD pathology increased. Finally, both oligodendrocytes and pyramidal neurons upregulated genes associated with ß-amyloid production and processing during this early hyperactive phase. Our integrative analysis provides an organizing framework for targeting circuit dysfunction, neuroinflammation, and amyloid production early in AD pathogenesis.


Subject(s)
Alzheimer Disease , Frontal Lobe , Microglia , Neurons , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid , Amyloid beta-Peptides/metabolism , Microglia/pathology , Neurons/pathology , Pyramidal Cells , Biopsy , Frontal Lobe/pathology , Single-Cell Gene Expression Analysis , Cell Nucleus/metabolism , Cell Nucleus/pathology
2.
Cell ; 177(7): 1873-1887.e17, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31178122

ABSTRACT

Defining cell types requires integrating diverse single-cell measurements from multiple experiments and biological contexts. To flexibly model single-cell datasets, we developed LIGER, an algorithm that delineates shared and dataset-specific features of cell identity. We applied it to four diverse and challenging analyses of human and mouse brain cells. First, we defined region-specific and sexually dimorphic gene expression in the mouse bed nucleus of the stria terminalis. Second, we analyzed expression in the human substantia nigra, comparing cell states in specific donors and relating cell types to those in the mouse. Third, we integrated in situ and single-cell expression data to spatially locate fine subtypes of cells present in the mouse frontal cortex. Finally, we jointly defined mouse cortical cell types using single-cell RNA-seq and DNA methylation profiles, revealing putative mechanisms of cell-type-specific epigenomic regulation. Integrative analyses using LIGER promise to accelerate investigations of cell-type definition, gene regulation, and disease states.


Subject(s)
DNA Methylation , Gene Expression Regulation , Septal Nuclei , Sequence Analysis, RNA , Single-Cell Analysis , Substantia Nigra , Adolescent , Adult , Aged , Animals , Female , Humans , Male , Mice , Middle Aged , Septal Nuclei/cytology , Septal Nuclei/metabolism , Substantia Nigra/cytology , Substantia Nigra/metabolism
3.
Nature ; 625(7993): 101-109, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38093010

ABSTRACT

Recent technological innovations have enabled the high-throughput quantification of gene expression and epigenetic regulation within individual cells, transforming our understanding of how complex tissues are constructed1-6. However, missing from these measurements is the ability to routinely and easily spatially localize these profiled cells. We developed a strategy, Slide-tags, in which single nuclei within an intact tissue section are tagged with spatial barcode oligonucleotides derived from DNA-barcoded beads with known positions. These tagged nuclei can then be used as an input into a wide variety of single-nucleus profiling assays. Application of Slide-tags to the mouse hippocampus positioned nuclei at less than 10 µm spatial resolution and delivered whole-transcriptome data that are indistinguishable in quality from ordinary single-nucleus RNA-sequencing data. To demonstrate that Slide-tags can be applied to a wide variety of human tissues, we performed the assay on brain, tonsil and melanoma. We revealed cell-type-specific spatially varying gene expression across cortical layers and spatially contextualized receptor-ligand interactions driving B cell maturation in lymphoid tissue. A major benefit of Slide-tags is that it is easily adaptable to almost any single-cell measurement technology. As a proof of principle, we performed multiomic measurements of open chromatin, RNA and T cell receptor (TCR) sequences in the same cells from metastatic melanoma, identifying transcription factor motifs driving cancer cell state transitions in spatially distinct microenvironments. Slide-tags offers a universal platform for importing the compendium of established single-cell measurements into the spatial genomics repertoire.


Subject(s)
DNA Barcoding, Taxonomic , Genomics , Animals , Humans , Mice , Brain/cytology , Brain/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA Barcoding, Taxonomic/methods , Epigenesis, Genetic , Gene Expression Profiling , Genomics/methods , Melanoma/genetics , Melanoma/pathology , Palatine Tonsil/cytology , Palatine Tonsil/metabolism , Receptors, Antigen, T-Cell/genetics , RNA/genetics , Single-Cell Analysis/methods , Transcriptome/genetics , Tumor Microenvironment , Hippocampus/cytology , Hippocampus/metabolism , Single-Cell Gene Expression Analysis , Organ Specificity , Ligands , Response Elements/genetics , Transcription Factors/metabolism
4.
Nature ; 624(7991): 333-342, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38092915

ABSTRACT

The function of the mammalian brain relies upon the specification and spatial positioning of diversely specialized cell types. Yet, the molecular identities of the cell types and their positions within individual anatomical structures remain incompletely known. To construct a comprehensive atlas of cell types in each brain structure, we paired high-throughput single-nucleus RNA sequencing with Slide-seq1,2-a recently developed spatial transcriptomics method with near-cellular resolution-across the entire mouse brain. Integration of these datasets revealed the cell type composition of each neuroanatomical structure. Cell type diversity was found to be remarkably high in the midbrain, hindbrain and hypothalamus, with most clusters requiring a combination of at least three discrete gene expression markers to uniquely define them. Using these data, we developed a framework for genetically accessing each cell type, comprehensively characterized neuropeptide and neurotransmitter signalling, elucidated region-specific specializations in activity-regulated gene expression and ascertained the heritability enrichment of neurological and psychiatric phenotypes. These data, available as an online resource ( www.BrainCellData.org ), should find diverse applications across neuroscience, including the construction of new genetic tools and the prioritization of specific cell types and circuits in the study of brain diseases.


Subject(s)
Brain , Gene Expression Profiling , Animals , Mice , Brain/anatomy & histology , Brain/cytology , Brain/metabolism , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing , Hypothalamus/cytology , Hypothalamus/metabolism , Mesencephalon/cytology , Mesencephalon/metabolism , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Phenotype , Rhombencephalon/cytology , Rhombencephalon/metabolism , Single-Cell Gene Expression Analysis , Transcriptome/genetics
5.
Nature ; 598(7879): 214-219, 2021 10.
Article in English | MEDLINE | ID: mdl-34616064

ABSTRACT

The cerebellar cortex is a well-studied brain structure with diverse roles in motor learning, coordination, cognition and autonomic regulation. However,  a complete inventory of cerebellar cell types is currently lacking. Here, using recent advances in high-throughput transcriptional profiling1-3, we molecularly define cell types across individual lobules of the adult mouse cerebellum. Purkinje neurons showed considerable regional specialization, with the greatest diversity occurring in the posterior lobules. For several types of cerebellar interneuron, the molecular variation within each type was more continuous, rather than discrete. In particular, for the unipolar brush cells-an interneuron population previously subdivided into discrete populations-the continuous variation in gene expression was associated with a graded continuum of electrophysiological properties. Notably, we found that molecular layer interneurons were composed of two molecularly and functionally distinct types. Both types show a continuum of morphological variation through the thickness of the molecular layer, but electrophysiological recordings revealed marked differences between the two types in spontaneous firing, excitability and electrical coupling. Together, these findings provide a comprehensive cellular atlas of the cerebellar cortex, and outline a methodological and conceptual framework for the integration of molecular, morphological and physiological ontologies for defining brain cell types.


Subject(s)
Cerebellar Cortex/cytology , Gene Expression Profiling , Transcriptome , Adult , Animals , Atlases as Topic , Electrophysiology , Female , Humans , Interneurons/classification , Interneurons/cytology , Interneurons/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroglia/classification , Neuroglia/cytology , Neuroglia/metabolism , Neurons/classification , Neurons/cytology , Neurons/metabolism
7.
Nucleic Acids Res ; 51(14): 7109-7124, 2023 08 11.
Article in English | MEDLINE | ID: mdl-37188501

ABSTRACT

Antisense oligonucleotides (ASOs) dosed into cerebrospinal fluid (CSF) distribute broadly throughout the central nervous system (CNS). By modulating RNA, they hold the promise of targeting root molecular causes of disease and hold potential to treat myriad CNS disorders. Realization of this potential requires that ASOs must be active in the disease-relevant cells, and ideally, that monitorable biomarkers also reflect ASO activity in these cells. The biodistribution and activity of such centrally delivered ASOs have been deeply characterized in rodent and non-human primate (NHP) models, but usually only in bulk tissue, limiting our understanding of the distribution of ASO activity across individual cells and across diverse CNS cell types. Moreover, in human clinical trials, target engagement is usually monitorable only in a single compartment, CSF. We sought a deeper understanding of how individual cells and cell types contribute to bulk tissue signal in the CNS, and how these are linked to CSF biomarker outcomes. We employed single nucleus transcriptomics on tissue from mice treated with RNase H1 ASOs against Prnp and Malat1 and NHPs treated with an ASO against PRNP. Pharmacologic activity was observed in every cell type, though sometimes with substantial differences in magnitude. Single cell RNA count distributions implied target RNA suppression in every single sequenced cell, rather than intense knockdown in only some cells. Duration of action up to 12 weeks post-dose differed across cell types, being shorter in microglia than in neurons. Suppression in neurons was generally similar to, or more robust than, the bulk tissue. In macaques, PrP in CSF was lowered 40% in conjunction with PRNP knockdown across all cell types including neurons, arguing that a CSF biomarker readout is likely to reflect ASO pharmacodynamic effect in disease-relevant cells in a neuronal disorder. Our results provide a reference dataset for ASO activity distribution in the CNS and establish single nucleus sequencing as a method for evaluating cell type specificity of oligonucleotide therapeutics and other modalities.


Antisense oligonucleotide (ASO) drugs are a type of chemically modified DNA that can be injected into cerebrospinal fluid in order to enter brain cells and reduce the amount of RNA from a specific gene. The brain is a complex mixture of hundreds of billions of cells. When an ASO lowers a target gene's RNA by 50%, is that a 50% reduction in 100% of cells, or a 100% reduction in 50% of cells? Are the many different cell types of the brain affected equally? This new study uses single cell RNA sequencing to answer these questions, finding that ASOs are broadly active across cell types and individual cells, and linking reduction of target protein in cerebrospinal fluid to disease-relevant cells.


Subject(s)
Brain , Oligonucleotides, Antisense , Animals , Mice , Brain/drug effects , Brain/metabolism , Oligonucleotides/metabolism , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/analysis , RNA/metabolism , Tissue Distribution , Transcription Factors/metabolism , Cerebrospinal Fluid/chemistry , Central Nervous System Diseases/therapy
8.
Nat Methods ; 18(11): 1352-1362, 2021 11.
Article in English | MEDLINE | ID: mdl-34711971

ABSTRACT

Charting an organs' biological atlas requires us to spatially resolve the entire single-cell transcriptome, and to relate such cellular features to the anatomical scale. Single-cell and single-nucleus RNA-seq (sc/snRNA-seq) can profile cells comprehensively, but lose spatial information. Spatial transcriptomics allows for spatial measurements, but at lower resolution and with limited sensitivity. Targeted in situ technologies solve both issues, but are limited in gene throughput. To overcome these limitations we present Tangram, a method that aligns sc/snRNA-seq data to various forms of spatial data collected from the same region, including MERFISH, STARmap, smFISH, Spatial Transcriptomics (Visium) and histological images. Tangram can map any type of sc/snRNA-seq data, including multimodal data such as those from SHARE-seq, which we used to reveal spatial patterns of chromatin accessibility. We demonstrate Tangram on healthy mouse brain tissue, by reconstructing a genome-wide anatomically integrated spatial map at single-cell resolution of the visual and somatomotor areas.


Subject(s)
Brain/metabolism , Chromatin/genetics , Deep Learning , Gene Expression Regulation , Single-Cell Analysis/methods , Software , Transcriptome , Animals , Chromatin/chemistry , Chromatin/metabolism , Female , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , RNA-Seq , Regulatory Sequences, Nucleic Acid
9.
Am J Pathol ; 193(5): 532-547, 2023 05.
Article in English | MEDLINE | ID: mdl-36804377

ABSTRACT

Chordoma is a rare malignant tumor demonstrating notochordal differentiation. It is dependent on brachyury (TBXT), a hallmark notochordal gene and transcription factor, and shares histologic features and the same anatomic location as the notochord. This study involved a molecular comparison of chordoma and notochord to identify dysregulated cellular pathways. The lack of a molecular reference from appropriate control tissue limits our understanding of chordoma and its relationship to notochord. Therefore, an unbiased comparison of chordoma, human notochord, and an atlas of normal and cancerous tissue was conducted using gene expression profiling to clarify the chordoma/notochord relationship and potentially identify novel drug targets. The study found striking consistency in gene expression profiles between chordoma and notochord, supporting the hypothesis that chordoma develops from notochordal remnants. A 12-gene diagnostic chordoma signature was identified and the TBXT/transforming growth factor beta (TGF-ß)/SOX6/SOX9 pathway was hyperactivated in the tumor, suggesting that pathways associated with chondrogenesis were a central driver of chordoma development. Experimental validation in chordoma cells confirmed these findings and emphasized the dependence of chordoma proliferation and survival on TGF-ß. The computational and experimental evidence provided the first molecular connection between notochord and chordoma and identified core members of a chordoma regulatory pathway involving TBXT. This pathway provides new therapeutic targets for this unique malignant neoplasm and highlights TGF-ß as a prime druggable candidate.


Subject(s)
Chordoma , Humans , Chordoma/genetics , Chordoma/pathology , Notochord/metabolism , Notochord/pathology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
11.
EMBO J ; 37(7)2018 04 03.
Article in English | MEDLINE | ID: mdl-29472250

ABSTRACT

The transition between soluble intrinsically disordered tau protein and aggregated tau in neurofibrillary tangles in Alzheimer's disease is unknown. Here, we propose that soluble tau species can undergo liquid-liquid phase separation (LLPS) under cellular conditions and that phase-separated tau droplets can serve as an intermediate toward tau aggregate formation. We demonstrate that phosphorylated or mutant aggregation prone recombinant tau undergoes LLPS, as does high molecular weight soluble phospho-tau isolated from human Alzheimer brain. Droplet-like tau can also be observed in neurons and other cells. We found that tau droplets become gel-like in minutes, and over days start to spontaneously form thioflavin-S-positive tau aggregates that are competent of seeding cellular tau aggregation. Since analogous LLPS observations have been made for FUS, hnRNPA1, and TDP43, which aggregate in the context of amyotrophic lateral sclerosis, we suggest that LLPS represents a biophysical process with a role in multiple different neurodegenerative diseases.


Subject(s)
Alzheimer Disease/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Brain/metabolism , Neurons/metabolism , Protein Aggregation, Pathological/metabolism , tau Proteins/chemistry , tau Proteins/isolation & purification , tau Proteins/metabolism , Aged, 80 and over , Amino Acid Sequence , Animals , Benzothiazoles/metabolism , Biophysical Phenomena , Cloning, Molecular , DNA-Binding Proteins/metabolism , Escherichia coli/genetics , Female , HEK293 Cells , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Humans , Liquid-Liquid Extraction , Mice , Mice, Transgenic , Molecular Weight , Neuroblastoma/metabolism , Neurodegenerative Diseases/metabolism , Neurofibrillary Tangles/metabolism , Phosphorylation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Analysis, Protein , Sf9 Cells
12.
J Neural Transm (Vienna) ; 128(4): 575-587, 2021 04.
Article in English | MEDLINE | ID: mdl-33439365

ABSTRACT

X-Linked Dystonia-Parkinsonism (XDP) is a neurodegenerative disease affecting individuals with ancestry to the island of Panay in the Philippines. In recent years there has been considerable progress at elucidating the genetic basis of XDP and candidate disease mechanisms in patient-derived cellular models, but the neural substrates that give rise to XDP in vivo are still poorly understood. Previous studies of limited XDP postmortem brain samples have reported a selective dropout of medium spiny neurons within the striatum, although neuroimaging of XDP patients has detected additional abnormalities in multiple brain regions beyond the basal ganglia. Given the need to fully define the CNS structures that are affected in this disease, we created a brain bank in Panay to serve as a tissue resource for detailed studies of XDP-related neuropathology. Here we describe this platform, from donor recruitment and consent to tissue collection, processing, and storage, that was assembled within a predominantly rural region of the Philippines with limited access to medical and laboratory facilities. Thirty-six brains from XDP individuals have been collected over an initial 4 years period. Tissue quality was assessed based on histologic staining of cortex, RNA integrity scores, detection of neuronal transcripts in situ by fluorescent hybridization chain reaction, and western blotting of neuronal and glial proteins. The results indicate that this pipeline preserves tissue integrity to an extent compatible with a range of morphologic, molecular, and biochemical analyses. Thus the algorithms that we developed for working in rural communities may serve as a guide for establishing similar brain banks for other rare diseases in indigenous populations.


Subject(s)
Dystonia , Dystonic Disorders , Neurodegenerative Diseases , Brain/diagnostic imaging , Dystonic Disorders/genetics , Genetic Diseases, X-Linked , Humans
13.
J Neurochem ; 147(1): 24-39, 2018 10.
Article in English | MEDLINE | ID: mdl-29806693

ABSTRACT

Synaptic dysfunction and loss are core pathological features in Alzheimer disease (AD). In the vicinity of amyloid-ß plaques in animal models, synaptic toxicity occurs and is associated with chronic activation of the phosphatase calcineurin (CN). Indeed, pharmacological inhibition of CN blocks amyloid-ß synaptotoxicity. We therefore hypothesized that CN-mediated transcriptional changes may contribute to AD neuropathology and tested this by examining the impact of CN over-expression on neuronal gene expression in vivo. We found dramatic transcriptional down-regulation, especially of synaptic mRNAs, in neurons chronically exposed to CN activation. Importantly, the transcriptional profile parallels the changes in human AD tissue. Bioinformatics analyses suggest that both nuclear factor of activated T cells and numerous microRNAs may all be impacted by CN, and parallel findings are observed in AD. These data and analyses support the hypothesis that at least part of the synaptic failure characterizing AD may result from aberrant CN activation leading to down-regulation of synaptic genes, potentially via activation of specific transcription factors and expression of repressive microRNAs. OPEN PRACTICES: Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/ Read the Editorial Highlight for this article on page 8.


Subject(s)
Alzheimer Disease/genetics , Calcineurin/genetics , Neurons/metabolism , Alzheimer Disease/pathology , Animals , Computational Biology , Gene Expression Regulation , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Hippocampus , Male , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Neurons/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Synapses/metabolism , Transcriptional Activation
14.
Ann Neurol ; 81(3): 395-406, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28019657

ABSTRACT

OBJECTIVE: Individuals with Parkinson disease are more likely to develop melanoma, and melanoma patients are reciprocally at higher risk of developing Parkinson disease. Melanoma is strongly tied to red hair/fair skin, a phenotype of loss-of-function polymorphisms in the MC1R (melanocortin 1 receptor) gene. Loss-of-function variants of MC1R have also been linked to increased risk of Parkinson disease. The present study is to investigate the role of MC1R in dopaminergic neurons in vivo. METHODS: Genetic and pharmacological approaches were employed to manipulate MC1R, and nigrostriatal dopaminergic integrity was determined by comprehensive behavioral, neurochemical, and neuropathological measures. RESULTS: MC1Re/e mice, which carry an inactivating mutation of MC1R and mimic the human redhead phenotype, have compromised nigrostriatal dopaminergic neuronal integrity, and they are more susceptible to dopaminergic neuron toxins 6-hydroxydopamine and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Furthermore, a selective MC1R agonist protects against MPTP-induced dopaminergic neurotoxicity. INTERPRETATION: Our findings reveal a protective role of MC1R in the nigrostriatal dopaminergic system, and they provide a rationale for MC1R as a potential therapeutic target for Parkinson disease. Together with its established role in melanoma, MC1R may represent a common pathogenic pathway for melanoma and Parkinson disease. Ann Neurol 2017;81:395-406.


Subject(s)
Dopaminergic Neurons/metabolism , Neostriatum/metabolism , Pigmentation/genetics , Receptor, Melanocortin, Type 1/physiology , Substantia Nigra/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Behavior, Animal , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Humans , Male , Melanoma/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neostriatum/drug effects , Neurotoxins/pharmacology , Parkinson Disease/genetics , Substantia Nigra/drug effects
15.
Ann Neurol ; 81(1): 117-128, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27997036

ABSTRACT

OBJECTIVE: Recent studies have shown that positron emission tomography (PET) tracer AV-1451 exhibits high binding affinity for paired helical filament (PHF)-tau pathology in Alzheimer's brains. However, the ability of this ligand to bind to tau lesions in other tauopathies remains controversial. Our goal was to examine the correlation of in vivo and postmortem AV-1451 binding patterns in three autopsy-confirmed non-Alzheimer tauopathy cases. METHODS: We quantified in vivo retention of [F-18]-AV-1451 and performed autoradiography, [H-3]-AV-1451 binding assays, and quantitative tau measurements in postmortem brain samples from two progressive supranuclear palsy (PSP) cases and a MAPT P301L mutation carrier. They all underwent [F-18]-AV-1451 PET imaging before death. RESULTS: The three subjects exhibited [F-18]-AV-1451 in vivo retention predominantly in basal ganglia and midbrain. Neuropathological examination confirmed the PSP diagnosis in the first two subjects; the MAPT P301L mutation carrier had an atypical tauopathy characterized by grain-like tau-containing neurites in gray and white matter with heaviest burden in basal ganglia. In all three cases, autoradiography failed to show detectable [F-18]-AV-1451 binding in multiple brain regions examined, with the exception of entorhinal cortex (reflecting incidental age-related neurofibrillary tangles) and neuromelanin-containing neurons in the substantia nigra (off-target binding). The lack of a consistent significant correlation between in vivo [F-18]-AV-1541 retention and postmortem in vitro binding and tau measures in these cases suggests that this ligand has low affinity for tau lesions primarily made of straight tau filaments. INTERPRETATION: AV-1451 may have limited utility for in vivo selective and reliable detection of tau aggregates in these non-Alzheimer tauopathies. ANN NEUROL 2017;81:117-128.


Subject(s)
Brain/pathology , Carbolines/metabolism , Tauopathies/pathology , tau Proteins/genetics , Aged , Autoradiography , Brain/diagnostic imaging , Brain/metabolism , Fluorine Radioisotopes/metabolism , Functional Neuroimaging , Humans , Male , Middle Aged , Mutation , Positron-Emission Tomography , Radioligand Assay , Supranuclear Palsy, Progressive/diagnostic imaging , Supranuclear Palsy, Progressive/pathology , Tauopathies/diagnostic imaging , Tauopathies/metabolism , Tritium/metabolism , tau Proteins/metabolism
16.
Vet Pathol ; 54(5): 828-831, 2017 09.
Article in English | MEDLINE | ID: mdl-28651457

ABSTRACT

Metallosis is the accumulation of metallic debris in soft tissues resulting from wear following total joint replacement. A dog was evaluated for lameness 4 years after total hip arthroplasty using a titanium alloy and cobalt chromium total hip system. Radiographs revealed severe acetabular component wear, implant-bone interface deterioration, and peri-acetabular osteolysis. During surgical revision, black periarticular tissue surrounded the implants. Histologically, there was fibrosis and granulomatous inflammation with abundant, intra- and extracellular, black, granular material and smaller amounts of clear punctate to acicular material. Laser capture microdissection followed by x-ray fluorescence microscopy indicated the material contained large amounts of titanium with smaller amounts of vanadium, cobalt, and chromium, confirming the diagnosis of metallosis. The clear material was birefringent under cross-polarized light, stained positive with Oil-Red-O, and thus was consistent with polyethylene. Metallosis exhibits characteristic gross and histologic lesions and is a differential diagnosis for aseptic loosening of hip implants.


Subject(s)
Arthroplasty, Replacement, Hip/veterinary , Hip Prosthesis/adverse effects , Metals/adverse effects , Osteolysis/veterinary , Postoperative Complications/veterinary , Animals , Arthroplasty, Replacement, Hip/adverse effects , Arthroplasty, Replacement, Hip/instrumentation , Chromium Alloys/adverse effects , Diagnosis, Differential , Dogs , Laser Capture Microdissection/veterinary , Male , Osteolysis/etiology , Polyethylene , Prosthesis Failure , Reoperation/veterinary , Titanium/adverse effects
17.
Am J Med Genet B Neuropsychiatr Genet ; 174(3): 315-323, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28190298

ABSTRACT

Bipolar disorder (BD) is a common, recurring psychiatric illness with unknown pathogenesis. Recent studies suggest that microRNA (miRNA) levels in brains of BD patients are significantly altered, and these changes may offer insight into BD pathology or etiology. Previously, we observed significant alterations of miR-29c levels in extracellular vesicles (EVs) extracted from prefrontal cortex (Brodmann area 9, BA9) of BD patients. In this study, we show that EVs extracted from the anterior cingulate cortex (BA24), a crucial area for modulating emotional expression and affect, have increased levels of miR-149 in BD patients compared to controls. Because miR-149 has been shown to inhibit glial proliferation, increased miR-149 expression in BA24-derived EVs is consistent with the previously reported reduced glial cell numbers in BA24 of patients diagnosed with either familial BD or familial major depressive disorder. qPCR analysis of laser-microdissected neuronal and glial cells from BA24 cortical samples of BD patients verified that the glial, but not neuronal, population exhibits significantly increased miR-149 expression. Finally, we report altered expression of both miR-149 and miR-29c in EVs extracted from brains of Flinders Sensitive Line rats, a well-validated animal model exhibiting depressive-like behaviors and glial (astrocytic) dysfunction. These findings warrant future investigations into the potential of using EV miRNA signatures as biomarkers to further enhance the biological definition of BD. © 2017 Wiley Periodicals, Inc.


Subject(s)
Bipolar Disorder/diagnosis , Bipolar Disorder/genetics , MicroRNAs/genetics , Animals , Biomarkers/blood , Brain/pathology , Depressive Disorder, Major/pathology , Disease Models, Animal , Extracellular Vesicles/genetics , Female , Gyrus Cinguli/metabolism , Humans , Male , MicroRNAs/blood , Rats
18.
Ann Neurol ; 78(5): 787-800, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26344059

ABSTRACT

OBJECTIVE: To examine region- and substrate-specific autoradiographic and in vitro binding patterns of positron emission tomography tracer [F-18]-AV-1451 (previously known as T807), tailored to allow in vivo detection of paired helical filament-tau-containing lesions, and to determine whether there is off-target binding to other amyloid/non-amyloid proteins. METHODS: We applied [F-18]-AV-1451 phosphor screen autoradiography, [F-18]-AV-1451 nuclear emulsion autoradiography, and [H-3]-AV-1451 in vitro binding assays to the study of postmortem samples from patients with a definite pathological diagnosis of Alzheimer disease, frontotemporal lobar degeneration-tau, frontotemporal lobar degeneration-transactive response DNA binding protein 43 (TDP-43), progressive supranuclear palsy, corticobasal degeneration, dementia with Lewy bodies, multiple system atrophy, cerebral amyloid angiopathy and elderly controls free of pathology. RESULTS: Our data suggest that [F-18]-AV-1451 strongly binds to tau lesions primarily made of paired helical filaments in Alzheimer brains (eg, intraneuronal and extraneuronal tangles and dystrophic neurites), but does not seem to bind to a significant extent to neuronal and glial inclusions mainly composed of straight tau filaments in non-Alzheimer tauopathy brains or to lesions containing ß-amyloid, α-synuclein, or TDP-43. [F-18]-AV-1451 off-target binding to neuromelanin- and melanin-containing cells and, to a lesser extent, to brain hemorrhagic lesions was identified. INTERPRETATION: Our data suggest that [F-18]-AV-1451 holds promise as a surrogate marker for the detection of brain tau pathology in the form of tangles and paired helical filament-tau-containing neurites in Alzheimer brains but also point to its relatively lower affinity for lesions primarily made of straight tau filaments in non-Alzheimer tauopathy cases and to the existence of some [F-18]-AV-1451 off-target binding. These findings provide important insights for interpreting in vivo patterns of [F-18]-AV-1451 retention.


Subject(s)
Brain/diagnostic imaging , Carbolines , Radiopharmaceuticals , Tauopathies/diagnostic imaging , tau Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/diagnostic imaging , Amyloid/metabolism , Autoradiography , Cadaver , Dementia/diagnostic imaging , Female , Frontotemporal Lobar Degeneration/diagnostic imaging , Humans , Inclusion Bodies/diagnostic imaging , Intracranial Hemorrhages/diagnostic imaging , Male , Middle Aged , Positron-Emission Tomography , TDP-43 Proteinopathies/diagnostic imaging
19.
Hum Mol Genet ; 22(15): 3077-92, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23585551

ABSTRACT

Alzheimer's disease (AD) is a multifactorial and fatal neurodegenerative disorder for which the mechanisms leading to profound neuronal loss are incompletely recognized. MicroRNAs (miRNAs) are recently discovered small regulatory RNA molecules that repress gene expression and are increasingly acknowledged as prime regulators involved in human brain pathologies. Here we identified two homologous miRNAs, miR-132 and miR-212, downregulated in temporal cortical areas and CA1 hippocampal neurons of human AD brains. Sequence-specific inhibition of miR-132 and miR-212 induces apoptosis in cultured primary neurons, whereas their overexpression is neuroprotective against oxidative stress. Using primary neurons and PC12 cells, we demonstrate that miR-132/212 controls cell survival by direct regulation of PTEN, FOXO3a and P300, which are all key elements of AKT signaling pathway. Silencing of these three target genes by RNAi abrogates apoptosis caused by the miR-132/212 inhibition. We further demonstrate that mRNA and protein levels of PTEN, FOXO3a, P300 and most of the direct pro-apoptotic transcriptional targets of FOXO3a are significantly elevated in human AD brains. These results indicate that the miR-132/miR-212/PTEN/FOXO3a signaling pathway contributes to AD neurodegeneration.


Subject(s)
Alzheimer Disease/genetics , Apoptosis/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation , MicroRNAs/genetics , Neurons/metabolism , Alzheimer Disease/metabolism , Animals , Apoptosis/drug effects , Brain/metabolism , Brain/pathology , Down-Regulation , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Humans , Hydrogen Peroxide/pharmacology , Mice , MicroRNAs/metabolism , Models, Biological , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , RNA Interference , Rats , Signal Transduction , p300-CBP Transcription Factors/metabolism
20.
Proc Natl Acad Sci U S A ; 108(21): E128-35, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21555590

ABSTRACT

The most common monogenic cause of small-vessel disease leading to ischemic stroke and vascular dementia is the neurodegenerative syndrome cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), which is associated with mutations in the Notch 3 receptor. CADASIL pathology is characterized by vascular smooth muscle cell degeneration and accumulation of diagnostic granular osmiophilic material (GOM) in vessels. The functional nature of the Notch 3 mutations causing CADASIL and their mechanistic connection to small-vessel disease and GOM accumulation remain enigmatic. To gain insight into how Notch 3 function is linked to CADASIL pathophysiology, we studied two phenotypically distinct mutations, C455R and R1031C, respectively associated with early and late onset of stroke, by using hemodynamic analyses in transgenic mouse models, receptor activity assays in cell culture, and proteomic examination of postmortem human tissue. We demonstrate that the C455R and R1031C mutations define different hypomorphic activity states of Notch 3, a property linked to ischemic stroke susceptibility in mouse models we generated. Importantly, these mice develop osmiophilic deposits and other age-dependent phenotypes that parallel remarkably the human condition. Proteomic analysis of human brain vessels, carrying the same CADASIL mutations, identified clusterin and collagen 18 α1/endostatin as GOM components. Our findings link loss of Notch signaling with ischemic cerebral small-vessel disease, a prevalent human condition. We determine that CADASIL pathophysiology is associated with hypomorphic Notch 3 function in vascular smooth muscle cells and implicate the accumulation of clusterin and collagen 18 α1/endostatin in brain vessel pathology.


Subject(s)
Alleles , Arterioles/pathology , Cerebrovascular Disorders/etiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Brain/blood supply , Disease Models, Animal , Humans , Ischemia , Mice , Mutation, Missense , Receptor, Notch3 , Receptors, Notch/genetics , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL