Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3606, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38697975

ABSTRACT

Amyotrophic Lateral Sclerosis (ALS), like many other neurodegenerative diseases, is highly heritable, but with only a small fraction of cases explained by monogenic disease alleles. To better understand sporadic ALS, we report epigenomic profiles, as measured by ATAC-seq, of motor neuron cultures derived from a diverse group of 380 ALS patients and 80 healthy controls. We find that chromatin accessibility is heavily influenced by sex, the iPSC cell type of origin, ancestry, and the inherent variance arising from sequencing. Once these covariates are corrected for, we are able to identify ALS-specific signals in the data. Additionally, we find that the ATAC-seq data is able to predict ALS disease progression rates with similar accuracy to methods based on biomarkers and clinical status. These results suggest that iPSC-derived motor neurons recapitulate important disease-relevant epigenomic changes.


Subject(s)
Amyotrophic Lateral Sclerosis , Induced Pluripotent Stem Cells , Motor Neurons , Humans , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/metabolism , Induced Pluripotent Stem Cells/metabolism , Motor Neurons/metabolism , Motor Neurons/pathology , Male , Female , Middle Aged , Case-Control Studies , Chromatin/metabolism , Chromatin/genetics , Aged , Epigenomics/methods , Chromatin Immunoprecipitation Sequencing/methods , Disease Progression , Epigenesis, Genetic
2.
Cell Rep Med ; 5(5): 101546, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38703766

ABSTRACT

Mutations in SOD1 cause amyotrophic lateral sclerosis (ALS), a neurodegenerative disease characterized by motor neuron (MN) loss. We previously discovered that macrophage migration inhibitory factor (MIF), whose levels are extremely low in spinal MNs, inhibits mutant SOD1 misfolding and toxicity. In this study, we show that a single peripheral injection of adeno-associated virus (AAV) delivering MIF into adult SOD1G37R mice significantly improves their motor function, delays disease progression, and extends survival. Moreover, MIF treatment reduces neuroinflammation and misfolded SOD1 accumulation, rescues MNs, and corrects dysregulated pathways as observed by proteomics and transcriptomics. Furthermore, we reveal low MIF levels in human induced pluripotent stem cell-derived MNs from familial ALS patients with different genetic mutations, as well as in post mortem tissues of sporadic ALS patients. Our findings indicate that peripheral MIF administration may provide a potential therapeutic mechanism for modulating misfolded SOD1 in vivo and disease outcome in ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis , Macrophage Migration-Inhibitory Factors , Motor Neurons , Superoxide Dismutase-1 , Macrophage Migration-Inhibitory Factors/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/therapy , Amyotrophic Lateral Sclerosis/pathology , Animals , Humans , Motor Neurons/metabolism , Motor Neurons/pathology , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Mice , Induced Pluripotent Stem Cells/metabolism , Intramolecular Oxidoreductases/metabolism , Intramolecular Oxidoreductases/genetics , Mice, Transgenic , Dependovirus/genetics , Disease Models, Animal , Male , Mutation/genetics , Female , Protein Folding
3.
bioRxiv ; 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37502982

ABSTRACT

Several prior studies have proposed the involvement of various brain regions and cell types in Parkinson's disease (PD) pathology. Here, we performed snRNA-seq on the prefrontal cortex and anterior cingulate regions from post-mortem control and PD brain tissue. We found a significant association of oligodendrocytes (ODCs) and oligodendrocyte precursor cells (OPCs) with PD-linked risk loci and report several dysregulated genes and pathways, including regulation of tau-protein kinase activity, regulation of inclusion body assembly and protein processing involved in protein targeting to mitochondria. In an independent PD cohort with clinical measures (681 cases and 549 controls), polygenic risk scores derived from the dysregulated genes significantly predicted Montreal Cognitive Assessment (MoCA)-, and Beck Depression Inventory-II (BDI-II)-scores but not motor impairment (UPDRS-III). We extended our analysis of clinical outcome prediction by incorporating three separate datasets that were previously published by different laboratories. In the first dataset from the anterior cingulate cortex, we identified a correlation between ODCs and BDI-II. In the second dataset obtained from the substantia nigra (SN), OPCs displayed notable predictive ability for UPDRS-III. In the third dataset from the SN region, a distinct subtype of OPCs, labeled OPC_ADM, exhibited predictive ability for UPDRS-III. Intriguingly, the OPC_ADM cluster also demonstrated a significant increase in PD samples. These results suggest that by expanding our focus to glial cells, we can uncover region-specific molecular pathways associated with PD symptoms.

4.
Nat Cardiovasc Res ; 1(4): 389-400, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35571675

ABSTRACT

Endothelial cells have a crucial role in nervous system function, and mounting evidence points to endothelial impairment as a major contributor to a wide range of neurological diseases. However, tools to genetically interrogate these cells in vivo remain limited. Here, we describe AAV-BI30, a capsid that specifically and efficiently transduces endothelial cells throughout the central nervous system. At relatively low systemic doses, this vector transduces the majority of arterial, capillary, and venous endothelial cells in the brain, retina, and spinal cord vasculature of adult C57BL/6 mice. Furthermore, we show that AAV-BI30 robustly transduces endothelial cells in multiple mouse strains and rats in vivo and human brain microvascular endothelial cells in vitro. Finally, we demonstrate AAV-BI30's capacity to achieve efficient and endothelial-specific Cre-mediated gene manipulation in the central nervous system. This combination of attributes makes AAV-BI30 uniquely well-suited to address outstanding research questions in neurovascular biology and aid the development of therapeutics to remediate endothelial dysfunction in disease.

5.
Nat Neurosci ; 25(6): 702-713, 2022 06.
Article in English | MEDLINE | ID: mdl-35578131

ABSTRACT

To understand how the cerebellar cortex transforms mossy fiber (MF) inputs into Purkinje cell (PC) outputs, it is vital to delineate the elements of this circuit. Candelabrum cells (CCs) are enigmatic interneurons of the cerebellar cortex that have been identified based on their morphology, but their electrophysiological properties, synaptic connections and function remain unknown. Here, we clarify these properties using electrophysiology, single-nucleus RNA sequencing, in situ hybridization and serial electron microscopy in mice. We find that CCs are the most abundant PC layer interneuron. They are GABAergic, molecularly distinct and present in all cerebellar lobules. Their high resistance renders CC firing highly sensitive to synaptic inputs. CCs are excited by MFs and granule cells and are strongly inhibited by PCs. CCs in turn primarily inhibit molecular layer interneurons, which leads to PC disinhibition. Thus, inputs, outputs and local signals converge onto CCs to allow them to assume a unique role in controlling cerebellar output.


Subject(s)
Cerebellar Cortex , Interneurons , Animals , Cerebellar Cortex/physiology , Cerebellum/physiology , Interneurons/physiology , Mice , Neurons/physiology , Purkinje Cells/physiology
6.
Nat Neurosci ; 25(3): 306-316, 2022 03.
Article in English | MEDLINE | ID: mdl-35260865

ABSTRACT

A key aspect of nearly all single-cell sequencing experiments is dissociation of intact tissues into single-cell suspensions. While many protocols have been optimized for optimal cell yield, they have often overlooked the effects that dissociation can have on ex vivo gene expression. Here, we demonstrate that use of enzymatic dissociation on brain tissue induces an aberrant ex vivo gene expression signature, most prominently in microglia, which is prevalent in published literature and can substantially confound downstream analyses. To address this issue, we present a rigorously validated protocol that preserves both in vivo transcriptional profiles and cell-type diversity and yield across tissue types and species. We also identify a similar signature in postmortem human brain single-nucleus RNA-sequencing datasets, and show that this signature is induced in freshly isolated human tissue by exposure to elevated temperatures ex vivo. Together, our results provide a methodological solution for preventing artifactual gene expression changes during fresh tissue digestion and a reference for future deeper analysis of the potential confounding states present in postmortem human samples.


Subject(s)
Neuroglia , Transcriptome , Brain , Humans , Microglia/metabolism , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
8.
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
9.
Nat Protoc ; 15(11): 3632-3662, 2020 11.
Article in English | MEDLINE | ID: mdl-33046898

ABSTRACT

High-throughput single-cell sequencing technologies hold tremendous potential for defining cell types in an unbiased fashion using gene expression and epigenomic state. A key challenge in realizing this potential is integrating single-cell datasets from multiple protocols, biological contexts, and data modalities into a joint definition of cellular identity. We previously developed an approach, called linked inference of genomic experimental relationships (LIGER), that uses integrative nonnegative matrix factorization to address this challenge. Here, we provide a step-by-step protocol for using LIGER to jointly define cell types from multiple single-cell datasets. The main stages of the protocol are data preprocessing and normalization, joint factorization, quantile normalization and joint clustering, and visualization. We describe how to jointly define cell types from single-cell RNA-seq (scRNA-seq) and single-nucleus ATAC-seq (snATAC-seq) data, but similar steps apply across a wide range of other settings and data types, including cross-species analysis, single-nucleus DNA methylation, and spatial transcriptomics. Our protocol contains examples of expected results, describes common pitfalls, and relies only on our freely available, open-source R implementation of LIGER. We also provide R Markdown tutorials showing the outputs from each individual code segment. The analysis process can be performed in 1-4 h, depending on dataset size, and assumes no specialized bioinformatics training.


Subject(s)
Genomics/methods , Single-Cell Analysis/methods , Animals , Cell Nucleus/genetics , Cluster Analysis , DNA Methylation , Gene Expression Profiling/methods , Humans , Sequence Analysis, RNA/methods , Software , Workflow
10.
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
11.
BMC Med Genomics ; 11(1): 91, 2018 Oct 20.
Article in English | MEDLINE | ID: mdl-30342520

ABSTRACT

BACKGROUND: Detection of copy number variants (CNVs) is an important aspect of clinical testing for several disorders, including Duchenne muscular dystrophy, and is often performed using multiplex ligation-dependent probe amplification (MLPA). However, since many genetic carrier screens depend instead on next-generation sequencing (NGS) for wider discovery of small variants, they often do not include CNV analysis. Moreover, most computational techniques developed to detect CNVs from exome sequencing data are not suitable for carrier screening, as they require matched normals, very large cohorts, or extensive gene panels. METHODS: We present a computational software package, geneCNV ( http://github.com/vkozareva/geneCNV ), which can identify exon-level CNVs using exome sequencing data from only a few genes. The tool relies on a hierarchical parametric model trained on a small cohort of reference samples. RESULTS: Using geneCNV, we accurately inferred heterozygous CNVs in the DMD gene across a cohort of 15 test subjects. These results were validated against MLPA, the current standard for clinical CNV analysis in DMD. We also benchmarked the tool's performance against other computational techniques and found comparable or improved CNV detection in DMD using data from panels ranging from 4,000 genes to as few as 8 genes. CONCLUSIONS: geneCNV allows for the creation of cost-effective screening panels by allowing NGS sequencing approaches to generate results equivalent to bespoke genotyping assays like MLPA. By using a parametric model to detect CNVs, it also fulfills regulatory requirements to define a reference range for a genetic test. It is freely available and can be incorporated into any Illumina sequencing pipeline to create clinical assays for detection of exon duplications and deletions.


Subject(s)
DNA Copy Number Variations , Germ Cells/metabolism , Muscular Dystrophy, Duchenne/genetics , Bayes Theorem , Genetic Testing/methods , High-Throughput Nucleotide Sequencing , Humans , Muscular Dystrophy, Duchenne/pathology , Saliva/metabolism , Sequence Analysis, DNA , User-Computer Interface
SELECTION OF CITATIONS
SEARCH DETAIL
...