Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Nature ; 603(7903): 885-892, 2022 03.
Article in English | MEDLINE | ID: mdl-35165441

ABSTRACT

The human brain vasculature is of great medical importance: its dysfunction causes disability and death1, and the specialized structure it forms-the blood-brain barrier-impedes the treatment of nearly all brain disorders2,3. Yet so far, we have no molecular map of the human brain vasculature. Here we develop vessel isolation and nuclei extraction for sequencing (VINE-seq) to profile the major vascular and perivascular cell types of the human brain through 143,793 single-nucleus transcriptomes from 25 hippocampus and cortex samples of 9 individuals with Alzheimer's disease and 8 individuals with no cognitive impairment. We identify brain-region- and species-enriched genes and pathways. We reveal molecular principles of human arteriovenous organization, recapitulating a gradual endothelial and punctuated mural cell continuum. We discover two subtypes of human pericytes, marked by solute transport and extracellular matrix (ECM) organization; and define perivascular versus meningeal fibroblast specialization. In Alzheimer's disease, we observe selective vulnerability of ECM-maintaining pericytes and gene expression patterns that implicate dysregulated blood flow. With an expanded survey of brain cell types, we find that 30 of the top 45 genes that have been linked to Alzheimer's disease risk by genome-wide association studies (GWASs) are expressed in the human brain vasculature, and we confirm this by immunostaining. Vascular GWAS genes map to endothelial protein transport, adaptive immune and ECM pathways. Many are microglia-specific in mice, suggesting a partial evolutionary transfer of Alzheimer's disease risk. Our work uncovers the molecular basis of the human brain vasculature, which will inform our understanding of overall brain health, disease and therapy.


Subject(s)
Alzheimer Disease , Brain , Disease Susceptibility , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Brain/blood supply , Brain/cytology , Brain/metabolism , Cerebral Cortex/blood supply , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Genome-Wide Association Study , Hippocampus/blood supply , Hippocampus/cytology , Hippocampus/metabolism , Humans , Mice , Microglia/metabolism , Pericytes/metabolism , Transcriptome
2.
Nature ; 595(7868): 565-571, 2021 07.
Article in English | MEDLINE | ID: mdl-34153974

ABSTRACT

Although SARS-CoV-2 primarily targets the respiratory system, patients with and survivors of COVID-19 can suffer neurological symptoms1-3. However, an unbiased understanding of the cellular and molecular processes that are affected in the brains of patients with COVID-19 is missing. Here we profile 65,309 single-nucleus transcriptomes from 30 frontal cortex and choroid plexus samples across 14 control individuals (including 1 patient with terminal influenza) and 8 patients with COVID-19. Although our systematic analysis yields no molecular traces of SARS-CoV-2 in the brain, we observe broad cellular perturbations indicating that barrier cells of the choroid plexus sense and relay peripheral inflammation into the brain and show that peripheral T cells infiltrate the parenchyma. We discover microglia and astrocyte subpopulations associated with COVID-19 that share features with pathological cell states that have previously been reported in human neurodegenerative disease4-6. Synaptic signalling of upper-layer excitatory neurons-which are evolutionarily expanded in humans7 and linked to cognitive function8-is preferentially affected in COVID-19. Across cell types, perturbations associated with COVID-19 overlap with those found in chronic brain disorders and reside in genetic variants associated with cognition, schizophrenia and depression. Our findings and public dataset provide a molecular framework to understand current observations of COVID-19-related neurological disease, and any such disease that may emerge at a later date.


Subject(s)
Astrocytes/pathology , Brain/pathology , COVID-19/diagnosis , COVID-19/pathology , Choroid Plexus/pathology , Microglia/pathology , Neurons/pathology , Aged , Aged, 80 and over , Brain/metabolism , Brain/physiopathology , Brain/virology , COVID-19/genetics , COVID-19/physiopathology , Cell Nucleus/genetics , Choroid Plexus/metabolism , Choroid Plexus/physiopathology , Choroid Plexus/virology , Female , Humans , Inflammation/virology , Male , Middle Aged , SARS-CoV-2/growth & development , SARS-CoV-2/pathogenicity , Single-Cell Analysis , Transcriptome , Virus Replication
3.
Nature ; 600(7889): 494-499, 2021 12.
Article in English | MEDLINE | ID: mdl-34880498

ABSTRACT

Physical exercise is generally beneficial to all aspects of human and animal health, slowing cognitive ageing and neurodegeneration1. The cognitive benefits of physical exercise are tied to an increased plasticity and reduced inflammation within the hippocampus2-4, yet little is known about the factors and mechanisms that mediate these effects. Here we show that 'runner plasma', collected from voluntarily running mice and infused into sedentary mice, reduces baseline neuroinflammatory gene expression and experimentally induced brain inflammation. Plasma proteomic analysis revealed a concerted increase in complement cascade inhibitors including clusterin (CLU). Intravenously injected CLU binds to brain endothelial cells and reduces neuroinflammatory gene expression in a mouse model of acute brain inflammation and a mouse model of Alzheimer's disease. Patients with cognitive impairment who participated in structured exercise for 6 months had higher plasma levels of CLU. These findings demonstrate the existence of anti-inflammatory exercise factors that are transferrable, target the cerebrovasculature and benefit the brain, and are present in humans who engage in exercise.


Subject(s)
Alzheimer Disease , Encephalitis , Alzheimer Disease/metabolism , Animals , Clusterin/genetics , Clusterin/metabolism , Endothelial Cells/metabolism , Humans , Mice , Proteomics
4.
Nature ; 583(7816): 425-430, 2020 07.
Article in English | MEDLINE | ID: mdl-32612231

ABSTRACT

The vascular interface of the brain, known as the blood-brain barrier (BBB), is understood to maintain brain function in part via its low transcellular permeability1-3. Yet, recent studies have demonstrated that brain ageing is sensitive to circulatory proteins4,5. Thus, it is unclear whether permeability to individually injected exogenous tracers-as is standard in BBB studies-fully represents blood-to-brain transport. Here we label hundreds of proteins constituting the mouse blood plasma proteome, and upon their systemic administration, study the BBB with its physiological ligand. We find that plasma proteins readily permeate the healthy brain parenchyma, with transport maintained by BBB-specific transcriptional programmes. Unlike IgG antibody, plasma protein uptake diminishes in the aged brain, driven by an age-related shift in transport from ligand-specific receptor-mediated to non-specific caveolar transcytosis. This age-related shift occurs alongside a specific loss of pericyte coverage. Pharmacological inhibition of the age-upregulated phosphatase ALPL, a predicted negative regulator of transport, enhances brain uptake of therapeutically relevant transferrin, transferrin receptor antibody and plasma. These findings reveal the extent of physiological protein transcytosis to the healthy brain, a mechanism of widespread BBB dysfunction with age and a strategy for enhanced drug delivery.


Subject(s)
Aging/metabolism , Aging/pathology , Blood-Brain Barrier/metabolism , Transcytosis , Alkaline Phosphatase/metabolism , Animals , Antibodies/metabolism , Biological Transport , Blood Proteins/administration & dosage , Blood Proteins/metabolism , Blood Proteins/pharmacokinetics , Brain/blood supply , Brain/metabolism , Drug Delivery Systems , Health , Humans , Male , Mice , Mice, Inbred C57BL , Plasma/metabolism , Proteome/administration & dosage , Proteome/metabolism , Proteome/pharmacokinetics , Receptors, Transferrin/immunology , Transcription, Genetic , Transferrin/metabolism
5.
Proc Natl Acad Sci U S A ; 119(11): e2121609119, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35259016

ABSTRACT

SignificanceNeurodegenerative diseases are poorly understood and difficult to treat. One common hallmark is lysosomal dysfunction leading to the accumulation of aggregates and other undegradable materials, which cause damage to brain resident cells. Lysosomes are acidic organelles responsible for breaking down biomolecules and recycling their constitutive parts. In this work, we find that the antiinflammatory and neuroprotective compound, discovered via a phenotypic screen, imparts its beneficial effects by targeting the lysosome and restoring its function. This is established using a genome-wide CRISPRi target identification screen and then confirmed using a variety of lysosome-targeted studies. The resulting small molecule from this study represents a potential treatment for neurodegenerative diseases as well as a research tool for the study of lysosomes in disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Lysosomes/drug effects , Neurodegenerative Diseases/metabolism , Animals , Anti-Inflammatory Agents/chemistry , Biomarkers , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Disease Susceptibility , Drug Development , Gene Expression Profiling , Humans , Mice , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/metabolism , Smad Proteins/agonists
6.
Nucleic Acids Res ; 49(2): e11, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33264392

ABSTRACT

Massively-parallel single-cell and single-nucleus RNA sequencing (scRNA-seq, snRNA-seq) requires extensive sequencing to achieve proper per-cell coverage, making sequencing resources and availability of sequencers critical factors for conducting deep transcriptional profiling. CoolMPS is a novel sequencing-by-synthesis approach that relies on nucleotide labeling by re-usable antibodies, but whether it is applicable to snRNA-seq has not been tested. Here, we use a low-cost and off-the-shelf protocol to chemically convert libraries generated with the widely-used Chromium 10X technology to be sequenceable with CoolMPS technology. To assess the quality and performance of converted libraries sequenced with CoolMPS, we generated a snRNA-seq dataset from the hippocampus of young and old mice. Native libraries were sequenced on an Illumina Novaseq and libraries that were converted to be compatible with CoolMPS were sequenced on a DNBSEQ-400RS. CoolMPS-derived data faithfully replicated key characteristics of the native library dataset, including correct estimation of ambient RNA-contamination, detection of captured cells, cell clustering results, spatial marker gene expression, inter- and intra-replicate differences and gene expression changes during aging. In conclusion, our results show that CoolMPS provides a viable alternative to standard sequencing of RNA from droplet-based libraries.


Subject(s)
Cell Encapsulation/methods , High-Throughput Nucleotide Sequencing/methods , RNA, Small Nuclear/chemistry , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Aging/genetics , Animals , Datasets as Topic , Fluorescent Antibody Technique, Direct , Gene Library , Gene Ontology , Hippocampus/chemistry , Hippocampus/growth & development , Male , Mice , Mice, Inbred C57BL , Microfluidics/methods , Nucleotides/immunology , Phosphorylation , RNA, Small Nuclear/isolation & purification , Specific Pathogen-Free Organisms
8.
Neuron ; 96(6): 1290-1302.e6, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29268096

ABSTRACT

Brain aging and neurodegeneration are associated with prominent microglial reactivity and activation of innate immune response pathways, commonly referred to as neuroinflammation. One such pathway, the type I interferon response, recognizes viral or mitochondrial DNA in the cytoplasm via activation of the recently discovered cyclic dinucleotide synthetase cGAS and the cyclic dinucleotide receptor STING. Here we show that the FDA-approved antiviral drug ganciclovir (GCV) induces a type I interferon response independent of its canonical thymidine kinase target. Inhibition of components of the STING pathway, including STING, IRF3, Tbk1, extracellular IFNß, and the Jak-Stat pathway resulted in reduced activity of GCV and its derivatives. Importantly, functional STING was necessary for GCV to inhibit inflammation in cultured myeloid cells and in a mouse model of multiple sclerosis. Collectively, our findings uncover an unexpected new activity of GCV and identify the STING pathway as a regulator of microglial reactivity and neuroinflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation/genetics , Interferon Type I/metabolism , Membrane Proteins/metabolism , Microglia/metabolism , Animals , Animals, Newborn , Antiviral Agents/therapeutic use , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Freund's Adjuvant/toxicity , Ganciclovir/therapeutic use , Gene Expression Regulation/drug effects , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Monocytes/drug effects , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/immunology , Pertussis Toxin/toxicity , Signal Transduction/drug effects , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL