Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nature ; 603(7900): 309-314, 2022 03.
Article in English | MEDLINE | ID: mdl-35236985

ABSTRACT

The ability to slow or reverse biological ageing would have major implications for mitigating disease risk and maintaining vitality1. Although an increasing number of interventions show promise for rejuvenation2, their effectiveness on disparate cell types across the body and the molecular pathways susceptible to rejuvenation remain largely unexplored. Here we performed single-cell RNA sequencing on 20 organs to reveal cell-type-specific responses to young and aged blood in heterochronic parabiosis. Adipose mesenchymal stromal cells, haematopoietic stem cells and hepatocytes are among those cell types that are especially responsive. On the pathway level, young blood invokes new gene sets in addition to reversing established ageing patterns, with the global rescue of genes encoding electron transport chain subunits pinpointing a prominent role of mitochondrial function in parabiosis-mediated rejuvenation. We observed an almost universal loss of gene expression with age that is largely mimicked by parabiosis: aged blood reduces global gene expression, and young blood restores it in select cell types. Together, these data lay the groundwork for a systemic understanding of the interplay between blood-borne factors and cellular integrity.


Subject(s)
Parabiosis , Single-Cell Analysis , Adipocytes , Aging/genetics , Electron Transport/genetics , Hematopoietic Stem Cells , Hepatocytes , Mesenchymal Stem Cells , Mitochondria , Organ Specificity/genetics , RNA-Seq , Rejuvenation
2.
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
3.
Nature ; 568(7751): 187-192, 2019 04.
Article in English | MEDLINE | ID: mdl-30944478

ABSTRACT

Microglia maintain homeostasis in the central nervous system through phagocytic clearance of protein aggregates and cellular debris. This function deteriorates during ageing and neurodegenerative disease, concomitant with cognitive decline. However, the mechanisms of impaired microglial homeostatic function and the cognitive effects of restoring this function remain unknown. We combined CRISPR-Cas9 knockout screens with RNA sequencing analysis to discover age-related genetic modifiers of microglial phagocytosis. These screens identified CD22, a canonical B cell receptor, as a negative regulator of phagocytosis that is upregulated on aged microglia. CD22 mediates the anti-phagocytic effect of α2,6-linked sialic acid, and inhibition of CD22 promotes the clearance of myelin debris, amyloid-ß oligomers and α-synuclein fibrils in vivo. Long-term central nervous system delivery of an antibody that blocks CD22 function reprograms microglia towards a homeostatic transcriptional state and improves cognitive function in aged mice. These findings elucidate a mechanism of age-related microglial impairment and a strategy to restore homeostasis in the ageing brain.


Subject(s)
Aging/physiology , Brain/cytology , Homeostasis/drug effects , Microglia/drug effects , N-Acetylneuraminic Acid/pharmacology , Phagocytosis/drug effects , Sialic Acid Binding Ig-like Lectin 2/antagonists & inhibitors , Aging/drug effects , Aging/genetics , Animals , Brain/drug effects , Brain/physiology , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Cognition/drug effects , Cognition/physiology , Female , Homeostasis/genetics , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , N-Acetylneuraminic Acid/chemistry , Phagocytosis/genetics , Sequence Analysis, RNA , Sialic Acid Binding Ig-like Lectin 2/genetics , Sialic Acid Binding Ig-like Lectin 2/metabolism
4.
Am J Respir Crit Care Med ; 206(7): 857-873, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35671465

ABSTRACT

Rationale: The leading cause of death in coronavirus disease 2019 (COVID-19) is severe pneumonia, with many patients developing acute respiratory distress syndrome (ARDS) and diffuse alveolar damage (DAD). Whether DAD in fatal COVID-19 is distinct from other causes of DAD remains unknown. Objective: To compare lung parenchymal and vascular alterations between patients with fatal COVID-19 pneumonia and other DAD-causing etiologies using a multidimensional approach. Methods: This autopsy cohort consisted of consecutive patients with COVID-19 pneumonia (n = 20) and with respiratory failure and histologic DAD (n = 21; non-COVID-19 viral and nonviral etiologies). Premortem chest computed tomography (CT) scans were evaluated for vascular changes. Postmortem lung tissues were compared using histopathological and computational analyses. Machine-learning-derived morphometric analysis of the microvasculature was performed, with a random forest classifier quantifying vascular congestion (CVasc) in different microscopic compartments. Respiratory mechanics and gas-exchange parameters were evaluated longitudinally in patients with ARDS. Measurements and Main Results: In premortem CT, patients with COVID-19 showed more dilated vasculature when all lung segments were evaluated (P = 0.001) compared with controls with DAD. Histopathology revealed vasculopathic changes, including hemangiomatosis-like changes (P = 0.043), thromboemboli (P = 0.0038), pulmonary infarcts (P = 0.047), and perivascular inflammation (P < 0.001). Generalized estimating equations revealed significant regional differences in the lung microarchitecture among all DAD-causing entities. COVID-19 showed a larger overall CVasc range (P = 0.002). Alveolar-septal congestion was associated with a significantly shorter time to death from symptom onset (P = 0.03), length of hospital stay (P = 0.02), and increased ventilatory ratio [an estimate for pulmonary dead space fraction (Vd); p = 0.043] in all cases of ARDS. Conclusions: Severe COVID-19 pneumonia is characterized by significant vasculopathy and aberrant alveolar-septal congestion. Our findings also highlight the role that vascular alterations may play in Vd and clinical outcomes in ARDS in general.


Subject(s)
COVID-19 , Pneumonia , Respiratory Distress Syndrome , Vascular Diseases , COVID-19/complications , Humans , Lung/diagnostic imaging , Lung/pathology , Pulmonary Alveoli/pathology , Respiratory Distress Syndrome/etiology
5.
Mod Pathol ; 34(7): 1345-1357, 2021 07.
Article in English | MEDLINE | ID: mdl-33727695

ABSTRACT

COVID-19 has been associated with cardiac injury and dysfunction. While both myocardial inflammatory cell infiltration and myocarditis with myocyte injury have been reported in patients with fatal COVID-19, clinical-pathologic correlations remain limited. The objective was to determine the relationships between cardiac pathological changes in patients dying from COVID-19 and cardiac infection by SARS-CoV-2, laboratory measurements, clinical features, and treatments. In a retrospective study, 41 consecutive autopsies of patients with fatal COVID-19 were analyzed for the associations between cardiac inflammation, myocarditis, cardiac infection by SARS-CoV-2, clinical features, laboratory measurements, and treatments. Cardiac infection was assessed by in situ hybridization and NanoString transcriptomic profiling. Cardiac infection by SARS-CoV-2 was present in 30/41 cases: virus+ with myocarditis (n = 4), virus+ without myocarditis (n = 26), and virus- without myocarditis (n = 11). In the cases with cardiac infection, SARS-CoV-2+ cells in the myocardium were rare, with a median density of 1 cell/cm2. Virus+ cases showed higher densities of myocardial CD68+ macrophages and CD3+ lymphocytes, as well as more electrocardiographic changes (23/27 vs 4/10; P = 0.01). Myocarditis was more prevalent with IL-6 blockade than with nonbiologic immunosuppression, primarily glucocorticoids (2/3 vs 0/14; P = 0.02). Overall, SARS-CoV-2 cardiac infection was less prevalent in patients treated with nonbiologic immunosuppression (7/14 vs 21/24; P = 0.02). Myocardial macrophage and lymphocyte densities overall were positively correlated with the duration of symptoms but not with underlying comorbidities. In summary, cardiac infection with SARS-CoV-2 is common among patients dying from COVID-19 but often with only rare infected cells. Cardiac infection by SARS-CoV-2 is associated with more cardiac inflammation and electrocardiographic changes. Nonbiologic immunosuppression is associated with lower incidences of myocarditis and cardiac infection by SARS-CoV-2.


Subject(s)
COVID-19/pathology , Aged , Anticoagulants/therapeutic use , Autopsy , COVID-19/blood , Echocardiography , Electrocardiography , Female , Humans , Immunosuppressive Agents/therapeutic use , Male , Myocarditis/pathology , Myocarditis/virology , Myocardium/pathology , Retrospective Studies , SARS-CoV-2/physiology , COVID-19 Drug Treatment
6.
Genes Dev ; 26(9): 974-87, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22499592

ABSTRACT

Neurotransmitter receptor recruitment at postsynaptic specializations is key in synaptogenesis, since this step confers functionality to the nascent synapse. The Drosophila neuromuscular junction (NMJ) is a glutamatergic synapse, similar in composition and function to mammalian central synapses. Various mechanisms regulating the extent of postsynaptic ionotropic glutamate receptor (iGluR) clustering have been described, but none are known to be essential for the initial localization and clustering of iGluRs at postsynaptic densities (PSDs). We identified and characterized the Drosophila neto (neuropilin and tolloid-like) as an essential gene required for clustering of iGluRs at the NMJ. Neto colocalizes with the iGluRs at the PSDs in puncta juxtaposing the active zones. neto loss-of-function phenotypes parallel the loss-of-function defects described for iGluRs. The defects in neto mutants are effectively rescued by muscle-specific expression of neto transgenes. Neto clustering at the Drosophila NMJ coincides with and is dependent on iGluRs. Our studies reveal that Drosophila Neto is a novel, essential component of the iGluR complexes and is required for iGluR clustering, organization of PSDs, and synapse functionality.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Membrane Proteins/metabolism , Muscle, Striated/metabolism , Neuromuscular Junction/metabolism , Post-Synaptic Density/metabolism , Receptors, Glutamate/metabolism , Animals , Drosophila Proteins/genetics , Membrane Proteins/genetics
7.
EMBO J ; 36(13): 1803-1805, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28623242

Subject(s)
Brain , Homeostasis , Humans
8.
Proc Natl Acad Sci U S A ; 110(13): 5058-63, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23479629

ABSTRACT

The acquisition of flight contributed to the success of insects and winged forms are present in most orders. Key to understanding the origin of wings will be knowledge of the earliest postembryonic events promoting wing outgrowth. The Drosophila melanogaster wing is intensely studied as a model appendage, and yet little is known about the beginning of wing outgrowth. Vein (Vn) is a neuregulin-like ligand for the EGF receptor (Egfr), which is necessary for global development of the early Drosophila wing disc. vn is not expressed in the embryonic wing primordium and thus has to be induced de novo in the nascent larval wing disc. We find that Decapentaplegic (Dpp), a Bone Morphogenetic Protein (BMP) family member, provides the instructive signal for initiating vn expression. The signaling involves paracrine communication between two epithelia in the early disc. Once initiated, vn expression is amplified and maintained by autocrine signaling mediated by the E-twenty six (ETS)-factor PointedP2 (PntP2). This interplay of paracrine and autocrine signaling underlies the spatial and temporal pattern of induction of Vn/Egfr target genes and explains both body wall development and wing outgrowth. It is possible this gene regulatory network governing expression of an EGF ligand is conserved and reflects a common origin of insect wings.


Subject(s)
Drosophila Proteins/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation/physiology , Receptors, Invertebrate Peptide/metabolism , Signal Transduction/physiology , Wings, Animal/growth & development , Animals , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster , ErbB Receptors/genetics , Neuregulins/biosynthesis , Neuregulins/genetics , Receptors, Invertebrate Peptide/genetics
9.
Nat Med ; 25(6): 988-1000, 2019 06.
Article in English | MEDLINE | ID: mdl-31086348

ABSTRACT

An aged circulatory environment can activate microglia, reduce neural precursor cell activity and impair cognition in mice. We hypothesized that brain endothelial cells (BECs) mediate at least some of these effects. We observe that BECs in the aged mouse hippocampus express an inflammatory transcriptional profile with focal upregulation of vascular cell adhesion molecule 1 (VCAM1), a protein that facilitates vascular-immune cell interactions. Concomitantly, levels of the shed, soluble form of VCAM1 are prominently increased in the plasma of aged humans and mice, and their plasma is sufficient to increase VCAM1 expression in cultured BECs and the hippocampi of young mice. Systemic administration of anti-VCAM1 antibody or genetic ablation of Vcam1 in BECs counteracts the detrimental effects of plasma from aged individuals on young brains and reverses aging aspects, including microglial reactivity and cognitive deficits, in the brains of aged mice. Together, these findings establish brain endothelial VCAM1 at the blood-brain barrier as a possible target to treat age-related neurodegeneration.


Subject(s)
Aging/blood , Brain/metabolism , Neural Stem Cells/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Adolescent , Adult , Aged , Aging/immunology , Aging/metabolism , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Brain/cytology , Cells, Cultured , Endothelial Cells/metabolism , Female , Gene Deletion , Hippocampus/cytology , Hippocampus/metabolism , Humans , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Microglia/metabolism , Neural Stem Cells/cytology , Vascular Cell Adhesion Molecule-1/blood , Vascular Cell Adhesion Molecule-1/genetics , Young Adult
10.
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
11.
Mol Neurodegener ; 10: 69, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26692002

ABSTRACT

BACKGROUND: Beclin 1 is a key regulator of multiple trafficking pathways, including autophagy and receptor recycling in yeast and microglia. Decreased beclin 1 levels in the CNS result in neurodegeneration, an effect attributed to impaired autophagy. However, neurons also rely heavily on trophic factors, and signaling through these pathways requires the proper trafficking of trophic factor receptors. RESULTS: We discovered that beclin 1 regulates signaling through the neuroprotective TGF-ß pathway. Beclin 1 is required for recycling of the type I TGF-ß receptor ALK5. We show that beclin 1 recruits the retromer to ALK5 and facilitates its localization to Rab11(+) endosomes. Decreased levels of beclin 1, or its binding partners VPS34 and UVRAG, impair TGF-ß signaling. CONCLUSIONS: These findings identify beclin 1 as a positive regulator of a trophic signaling pathway via receptor recycling, and suggest that neuronal death induced by decreased beclin 1 levels may also be due to impaired trophic factor signaling.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Microglia/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Animals , Autophagy/physiology , Beclin-1 , Endosomes/metabolism , Mice , Protein Transport/physiology , Receptor, Transforming Growth Factor-beta Type I
12.
Genetics ; 191(4): 1213-26, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22595244

ABSTRACT

The highly conserved epidermal growth factor receptor (Egfr) pathway is required in all animals for normal development and homeostasis; consequently, aberrant Egfr signaling is implicated in a number of diseases. Genetic analysis of Drosophila melanogaster Egfr has contributed significantly to understanding this conserved pathway and led to the discovery of new components and targets. Here we used microarray analysis of third instar wing discs, in which Egfr signaling was perturbed, to identify new Egfr-responsive genes. Upregulated transcripts included five known targets, suggesting the approach was valid. We investigated the function of 29 previously uncharacterized genes, which had pronounced responses. The Egfr pathway is important for wing-vein patterning and using reverse genetic analysis we identified five genes that showed venation defects. Three of these genes are expressed in vein primordia and all showed transcriptional changes in response to altered Egfr activity consistent with being targets of the pathway. Genetic interactions with Egfr further linked two of the genes, Sulfated (Sulf1), an endosulfatase gene, and CG4096, an A Disintegrin And Metalloproteinase with ThromboSpondin motifs (ADAMTS) gene, to the pathway. Sulf1 showed a strong genetic interaction with the neuregulin-like ligand vein (vn) and may influence binding of Vn to heparan-sulfated proteoglycans (HSPGs). How Drosophila Egfr activity is modulated by CG4096 is unknown, but interestingly vertebrate EGF ligands are regulated by a related ADAMTS protein. We suggest Sulf1 and CG4096 are negative feedback regulators of Egfr signaling that function in the extracellular space to influence ligand activity.


Subject(s)
Drosophila/metabolism , ErbB Receptors/metabolism , Feedback, Physiological , Signal Transduction , Animals , Body Patterning/genetics , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Epistasis, Genetic , ErbB Receptors/genetics , Female , Gene Expression Profiling , Gene Expression Regulation , Genotype , Ligands , Phenotype , Protein Binding , RNA Interference , Sulfatases/genetics , Sulfatases/metabolism , Transcriptome , Veins/metabolism , Wings, Animal/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL