Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22.040
Filter
Add more filters

Publication year range
1.
Cell ; 186(7): 1337-1351.e20, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36870332

ABSTRACT

Leaf-feeding insects trigger high-amplitude, defense-inducing electrical signals called slow wave potentials (SWPs). These signals are thought to be triggered by the long-distance transport of low molecular mass elicitors termed Ricca's factors. We sought mediators of leaf-to-leaf electrical signaling in Arabidopsis thaliana and identified them as ß-THIOGLUCOSIDE GLUCOHYDROLASE 1 and 2 (TGG1 and TGG2). SWP propagation from insect feeding sites was strongly attenuated in tgg1 tgg2 mutants and wound-response cytosolic Ca2+ increases were reduced in these plants. Recombinant TGG1 fed into the xylem elicited wild-type-like membrane depolarization and Ca2+ transients. Moreover, TGGs catalyze the deglucosidation of glucosinolates. Metabolite profiling revealed rapid wound-induced breakdown of aliphatic glucosinolates in primary veins. Using in vivo chemical trapping, we found evidence for roles of short-lived aglycone intermediates generated by glucosinolate hydrolysis in SWP membrane depolarization. Our findings reveal a mechanism whereby organ-to-organ protein transport plays a major role in electrical signaling.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Animals , Glycoside Hydrolases/metabolism , Glucosinolates/metabolism , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Insecta
2.
Cell ; 185(8): 1325-1345.e22, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35366418

ABSTRACT

Protein aggregation is a hallmark of multiple human pathologies. Autophagy selectively degrades protein aggregates via aggrephagy. How selectivity is achieved has been elusive. Here, we identify the chaperonin subunit CCT2 as an autophagy receptor regulating the clearance of aggregation-prone proteins in the cell and the mouse brain. CCT2 associates with aggregation-prone proteins independent of cargo ubiquitination and interacts with autophagosome marker ATG8s through a non-classical VLIR motif. In addition, CCT2 regulates aggrephagy independently of the ubiquitin-binding receptors (P62, NBR1, and TAX1BP1) or chaperone-mediated autophagy. Unlike P62, NBR1, and TAX1BP1, which facilitate the clearance of protein condensates with liquidity, CCT2 specifically promotes the autophagic degradation of protein aggregates with little liquidity (solid aggregates). Furthermore, aggregation-prone protein accumulation induces the functional switch of CCT2 from a chaperone subunit to an autophagy receptor by promoting CCT2 monomer formation, which exposes the VLIR to ATG8s interaction and, therefore, enables the autophagic function.


Subject(s)
Chaperonin Containing TCP-1 , Macroautophagy , Protein Aggregates , Animals , Mice , Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Carrier Proteins/metabolism , Chaperonin Containing TCP-1/metabolism , Sequestosome-1 Protein/metabolism
3.
Cell ; 185(7): 1117-1129.e8, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35298912

ABSTRACT

Game animals are wildlife species traded and consumed as food and are potential reservoirs for SARS-CoV and SARS-CoV-2. We performed a meta-transcriptomic analysis of 1,941 game animals, representing 18 species and five mammalian orders, sampled across China. From this, we identified 102 mammalian-infecting viruses, with 65 described for the first time. Twenty-one viruses were considered as potentially high risk to humans and domestic animals. Civets (Paguma larvata) carried the highest number of potentially high-risk viruses. We inferred the transmission of bat-associated coronavirus from bats to civets, as well as cross-species jumps of coronaviruses from bats to hedgehogs, from birds to porcupines, and from dogs to raccoon dogs. Of note, we identified avian Influenza A virus H9N2 in civets and Asian badgers, with the latter displaying respiratory symptoms, as well as cases of likely human-to-wildlife virus transmission. These data highlight the importance of game animals as potential drivers of disease emergence.


Subject(s)
Animals, Wild/virology , Communicable Diseases, Emerging/virology , Disease Reservoirs , Mammals/virology , Virome , Animals , China , Phylogeny , Zoonoses
4.
Cell ; 185(8): 1346-1355.e15, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35247328

ABSTRACT

Misfolding and aggregation of disease-specific proteins, resulting in the formation of filamentous cellular inclusions, is a hallmark of neurodegenerative disease with characteristic filament structures, or conformers, defining each proteinopathy. Here we show that a previously unsolved amyloid fibril composed of a 135 amino acid C-terminal fragment of TMEM106B is a common finding in distinct human neurodegenerative diseases, including cases characterized by abnormal aggregation of TDP-43, tau, or α-synuclein protein. A combination of cryoelectron microscopy and mass spectrometry was used to solve the structures of TMEM106B fibrils at a resolution of 2.7 Å from postmortem human brain tissue afflicted with frontotemporal lobar degeneration with TDP-43 pathology (FTLD-TDP, n = 8), progressive supranuclear palsy (PSP, n = 2), or dementia with Lewy bodies (DLB, n = 1). The commonality of abundant amyloid fibrils composed of TMEM106B, a lysosomal/endosomal protein, to a broad range of debilitating human disorders indicates a shared fibrillization pathway that may initiate or accelerate neurodegeneration.


Subject(s)
Frontotemporal Dementia , Membrane Proteins , Nerve Tissue Proteins , Neurodegenerative Diseases , Amyloid , Cryoelectron Microscopy , DNA-Binding Proteins/metabolism , Frontotemporal Dementia/pathology , Humans , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism
5.
Cell ; 184(13): 3542-3558.e16, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34051138

ABSTRACT

Structural variations (SVs) and gene copy number variations (gCNVs) have contributed to crop evolution, domestication, and improvement. Here, we assembled 31 high-quality genomes of genetically diverse rice accessions. Coupling with two existing assemblies, we developed pan-genome-scale genomic resources including a graph-based genome, providing access to rice genomic variations. Specifically, we discovered 171,072 SVs and 25,549 gCNVs and used an Oryza glaberrima assembly to infer the derived states of SVs in the Oryza sativa population. Our analyses of SV formation mechanisms, impacts on gene expression, and distributions among subpopulations illustrate the utility of these resources for understanding how SVs and gCNVs shaped rice environmental adaptation and domestication. Our graph-based genome enabled genome-wide association study (GWAS)-based identification of phenotype-associated genetic variations undetectable when using only SNPs and a single reference assembly. Our work provides rich population-scale resources paired with easy-to-access tools to facilitate rice breeding as well as plant functional genomics and evolutionary biology research.


Subject(s)
Ecotype , Genetic Variation , Genome, Plant , Oryza/genetics , Adaptation, Physiological/genetics , Agriculture , Domestication , Gene Expression Profiling , Gene Expression Regulation, Plant , Genes, Plant , Genomic Structural Variation , Molecular Sequence Annotation , Phenotype
6.
Cell ; 182(1): 59-72.e15, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32492406

ABSTRACT

Early detection and effective treatment of severe COVID-19 patients remain major challenges. Here, we performed proteomic and metabolomic profiling of sera from 46 COVID-19 and 53 control individuals. We then trained a machine learning model using proteomic and metabolomic measurements from a training cohort of 18 non-severe and 13 severe patients. The model was validated using 10 independent patients, 7 of which were correctly classified. Targeted proteomics and metabolomics assays were employed to further validate this molecular classifier in a second test cohort of 19 COVID-19 patients, leading to 16 correct assignments. We identified molecular changes in the sera of COVID-19 patients compared to other groups implicating dysregulation of macrophage, platelet degranulation, complement system pathways, and massive metabolic suppression. This study revealed characteristic protein and metabolite changes in the sera of severe COVID-19 patients, which might be used in selection of potential blood biomarkers for severity evaluation.


Subject(s)
Coronavirus Infections/blood , Metabolomics , Pneumonia, Viral/blood , Proteomics , Adult , Amino Acids/metabolism , Biomarkers/blood , COVID-19 , Cluster Analysis , Coronavirus Infections/physiopathology , Female , Humans , Lipid Metabolism , Machine Learning , Macrophages/pathology , Male , Middle Aged , Pandemics , Pneumonia, Viral/physiopathology , Severity of Illness Index
7.
Cell ; 180(5): 878-894.e19, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32059783

ABSTRACT

Pathogenic autoantibodies arise in many autoimmune diseases, but it is not understood how the cells making them evade immune checkpoints. Here, single-cell multi-omics analysis demonstrates a shared mechanism with lymphoid malignancy in the formation of public rheumatoid factor autoantibodies responsible for mixed cryoglobulinemic vasculitis. By combining single-cell DNA and RNA sequencing with serum antibody peptide sequencing and antibody synthesis, rare circulating B lymphocytes making pathogenic autoantibodies were found to comprise clonal trees accumulating mutations. Lymphoma driver mutations in genes regulating B cell proliferation and V(D)J mutation (CARD11, TNFAIP3, CCND3, ID3, BTG2, and KLHL6) were present in rogue B cells producing the pathogenic autoantibody. Antibody V(D)J mutations conferred pathogenicity by causing the antigen-bound autoantibodies to undergo phase transition to insoluble aggregates at lower temperatures. These results reveal a pre-neoplastic stage in human lymphomagenesis and a cascade of somatic mutations leading to an iconic pathogenic autoantibody.


Subject(s)
Autoantibodies/genetics , Autoimmune Diseases/genetics , B-Lymphocytes/immunology , Lymphoma/genetics , Animals , Autoantibodies/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , B-Lymphocytes/pathology , CARD Signaling Adaptor Proteins/genetics , Carrier Proteins/genetics , Clonal Evolution/genetics , Clonal Evolution/immunology , Cyclin D3/genetics , Guanylate Cyclase/genetics , Humans , Immediate-Early Proteins/genetics , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Inhibitor of Differentiation Proteins/genetics , Lymphoma/immunology , Lymphoma/pathology , Mice , Mutation/genetics , Mutation/immunology , Neoplasm Proteins/genetics , Sequence Analysis, DNA/methods , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Suppressor Proteins/genetics , V(D)J Recombination/genetics
8.
Nat Immunol ; 22(2): 193-204, 2021 02.
Article in English | MEDLINE | ID: mdl-33398181

ABSTRACT

Metabolic reprograming toward aerobic glycolysis is a pivotal mechanism shaping immune responses. Here we show that deficiency in NF-κB-inducing kinase (NIK) impairs glycolysis induction, rendering CD8+ effector T cells hypofunctional in the tumor microenvironment. Conversely, ectopic expression of NIK promotes CD8+ T cell metabolism and effector function, thereby profoundly enhancing antitumor immunity and improving the efficacy of T cell adoptive therapy. NIK regulates T cell metabolism via a NF-κB-independent mechanism that involves stabilization of hexokinase 2 (HK2), a rate-limiting enzyme of the glycolytic pathway. NIK prevents autophagic degradation of HK2 through controlling cellular reactive oxygen species levels, which in turn involves modulation of glucose-6-phosphate dehydrogenase (G6PD), an enzyme that mediates production of the antioxidant NADPH. We show that the G6PD-NADPH redox system is important for HK2 stability and metabolism in activated T cells. These findings establish NIK as a pivotal regulator of T cell metabolism and highlight a post-translational mechanism of metabolic regulation.


Subject(s)
CD8-Positive T-Lymphocytes/enzymology , Colonic Neoplasms/enzymology , Energy Metabolism , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/enzymology , Melanoma, Experimental/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Cytotoxicity, Immunologic , Enzyme Stability , Female , Glucosephosphate Dehydrogenase/metabolism , Glycolysis , Hexokinase/genetics , Hexokinase/metabolism , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/transplantation , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Mice, Knockout , NADP/metabolism , Phenotype , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Microenvironment , NF-kappaB-Inducing Kinase
9.
Nat Immunol ; 22(3): 358-369, 2021 03.
Article in English | MEDLINE | ID: mdl-33432230

ABSTRACT

CD8+ T cell exhaustion dampens antitumor immunity. Although several transcription factors have been identified that regulate T cell exhaustion, the molecular mechanisms by which CD8+ T cells are triggered to enter an exhausted state remain unclear. Here, we show that interleukin-2 (IL-2) acts as an environmental cue to induce CD8+ T cell exhaustion within tumor microenvironments. We find that a continuously high level of IL-2 leads to the persistent activation of STAT5 in CD8+ T cells, which in turn induces strong expression of tryptophan hydroxylase 1, thus catalyzing the conversion to tryptophan to 5-hydroxytryptophan (5-HTP). 5-HTP subsequently activates AhR nuclear translocation, causing a coordinated upregulation of inhibitory receptors and downregulation of cytokine and effector-molecule production, thereby rendering T cells dysfunctional in the tumor microenvironment. This molecular pathway is not only present in mouse tumor models but is also observed in people with cancer, identifying IL-2 as a novel inducer of T cell exhaustion.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/drug effects , Interleukin-2/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Neoplasms/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Tumor Microenvironment , 5-Hydroxytryptophan/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , Humans , Interleukin-2/antagonists & inhibitors , Interleukin-2/genetics , Jurkat Cells , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , MCF-7 Cells , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , Tryptophan Hydroxylase/metabolism , Xenograft Model Antitumor Assays
10.
Immunity ; 57(1): 106-123.e7, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38159573

ABSTRACT

When the filtrate of the glomerulus flows through the renal tubular system, various microscopic sediment particles, including mineral crystals, are generated. Dislodging these particles is critical to ensuring the free flow of filtrate, whereas failure to remove them will result in kidney stone formation and obstruction. However, the underlying mechanism for the clearance is unclear. Here, using high-resolution microscopy, we found that the juxtatubular macrophages in the renal medulla constitutively formed transepithelial protrusions and "sampled" urine contents. They efficiently sequestered and phagocytosed intraluminal sediment particles and occasionally transmigrated to the tubule lumen to escort the excretion of urine particles. Mice with decreased renal macrophage numbers were prone to developing various intratubular sediments, including kidney stones. Mechanistically, the transepithelial behaviors of medulla macrophages required integrin ß1-mediated ligation to the tubular epithelium. These findings indicate that medulla macrophages sample urine content and remove intratubular particles to keep the tubular system unobstructed.


Subject(s)
Kidney Calculi , Kidney , Mice , Animals , Macrophages
11.
Immunity ; 56(5): 1013-1026.e6, 2023 05 09.
Article in English | MEDLINE | ID: mdl-36944334

ABSTRACT

Sepsis is a dysregulated inflammatory consequence of systemic infection. As a result, excessive platelet activation leads to thrombosis and coagulopathy, but we currently lack sufficient understanding of these processes. Here, using the cecal ligation and puncture (CLP) model of sepsis, we observed septic thrombosis and neutrophil extracellular trap formation (NETosis) within the mouse vasculature by intravital microscopy. STING activation in platelets was a critical driver of sepsis-induced pathology. Platelet-specific STING deficiency suppressed platelet activation and granule secretion, which alleviated sepsis-induced intravascular thrombosis and NETosis in mice. Mechanistically, sepsis-derived cGAMP promoted the binding of STING to STXBP2, the assembly of SNARE complex, granule secretion, and subsequent septic thrombosis, which probably depended on the palmitoylation of STING. We generated a peptide, C-ST5, to block STING binding to STXBP2. Septic mice treated with C-ST5 showed reduced thrombosis. Overall, platelet activation via STING reveals a potential strategy for limiting life-threatening sepsis-mediated coagulopathy.


Subject(s)
Extracellular Traps , Sepsis , Thrombosis , Animals , Mice , Blood Platelets/metabolism , Extracellular Traps/metabolism , Mice, Inbred C57BL , Munc18 Proteins/metabolism , Platelet Activation , Sepsis/metabolism , Thrombosis/metabolism
12.
Cell ; 170(6): 1120-1133.e17, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28803728

ABSTRACT

Immune-checkpoint blockade is able to achieve durable responses in a subset of patients; however, we lack a satisfying comprehension of the underlying mechanisms of anti-CTLA-4- and anti-PD-1-induced tumor rejection. To address these issues, we utilized mass cytometry to comprehensively profile the effects of checkpoint blockade on tumor immune infiltrates in human melanoma and murine tumor models. These analyses reveal a spectrum of tumor-infiltrating T cell populations that are highly similar between tumor models and indicate that checkpoint blockade targets only specific subsets of tumor-infiltrating T cell populations. Anti-PD-1 predominantly induces the expansion of specific tumor-infiltrating exhausted-like CD8 T cell subsets. In contrast, anti-CTLA-4 induces the expansion of an ICOS+ Th1-like CD4 effector population in addition to engaging specific subsets of exhausted-like CD8 T cells. Thus, our findings indicate that anti-CTLA-4 and anti-PD-1 checkpoint-blockade-induced immune responses are driven by distinct cellular mechanisms.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Melanoma/immunology , Melanoma/therapy , Neoplasm Metastasis/immunology , Neoplasm Metastasis/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocyte Subsets/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Flow Cytometry , Gene Expression Regulation , Humans , Immunotherapy , Melanoma/pathology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/pathology , Single-Cell Analysis , Transcription, Genetic
13.
Cell ; 170(1): 114-126.e15, 2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28666113

ABSTRACT

Rice feeds half the world's population, and rice blast is often a destructive disease that results in significant crop loss. Non-race-specific resistance has been more effective in controlling crop diseases than race-specific resistance because of its broad spectrum and durability. Through a genome-wide association study, we report the identification of a natural allele of a C2H2-type transcription factor in rice that confers non-race-specific resistance to blast. A survey of 3,000 sequenced rice genomes reveals that this allele exists in 10% of rice, suggesting that this favorable trait has been selected through breeding. This allele causes a single nucleotide change in the promoter of the bsr-d1 gene, which results in reduced expression of the gene through the binding of the repressive MYB transcription factor and, consequently, an inhibition of H2O2 degradation and enhanced disease resistance. Our discovery highlights this novel allele as a strategy for breeding durable resistance in rice.


Subject(s)
Oryza/genetics , Plant Proteins/genetics , Transcription Factors/genetics , Base Sequence , Breeding , Disease Resistance , Gene Knockout Techniques , Genome, Plant , Genome-Wide Association Study , Plant Diseases , Promoter Regions, Genetic
14.
Nat Immunol ; 20(7): 879-889, 2019 07.
Article in English | MEDLINE | ID: mdl-31182807

ABSTRACT

CD8+ T cells and natural killer (NK) cells are central cellular components of immune responses against pathogens and cancer, which rely on interleukin (IL)-15 for homeostasis. Here we show that IL-15 also mediates homeostatic priming of CD8+ T cells for antigen-stimulated activation, which is controlled by a deubiquitinase, Otub1. IL-15 mediates membrane recruitment of Otub1, which inhibits ubiquitin-dependent activation of AKT, a kinase that is pivotal for T cell activation and metabolism. Otub1 deficiency in mice causes aberrant responses of CD8+ T cells to IL-15, rendering naive CD8+ T cells hypersensitive to antigen stimulation characterized by enhanced metabolic reprograming and effector functions. Otub1 also controls the maturation and activation of NK cells. Deletion of Otub1 profoundly enhances anticancer immunity by unleashing the activity of CD8+ T cells and NK cells. These findings suggest that Otub1 controls the activation of CD8+ T cells and NK cells by functioning as a checkpoint of IL-15-mediated priming.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cysteine Endopeptidases/metabolism , Interleukin-15/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cysteine Endopeptidases/deficiency , Deubiquitinating Enzymes/metabolism , Disease Models, Animal , Energy Metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Interleukin-15/genetics , Melanoma, Experimental , Mice , Mice, Transgenic , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin-15/metabolism , Self Tolerance/genetics , Self Tolerance/immunology , Signal Transduction , T-Cell Antigen Receptor Specificity , Ubiquitination
15.
Nat Immunol ; 20(2): 183-194, 2019 02.
Article in English | MEDLINE | ID: mdl-30643264

ABSTRACT

Intestinal stem cells (ISCs) are maintained by stemness signaling for precise modulation of self-renewal and differentiation under homeostasis. However, the way in which intestinal immune cells regulate the self-renewal of ISCs remains elusive. Here we found that mouse and human Lgr5+ ISCs showed high expression of the immune cell-associated circular RNA circPan3 (originating from the Pan3 gene transcript). Deletion of circPan3 in Lgr5+ ISCs impaired their self-renewal capacity and the regeneration of gut epithelium in a manner dependent on immune cells. circPan3 bound mRNA encoding the cytokine IL-13 receptor subunit IL-13Rα1 (Il13ra1) in ISCs to increase its stability, which led to the expression of IL-13Rα1 in ISCs. IL-13 produced by group 2 innate lymphoid cells in the crypt niche engaged IL-13Rα1 on crypt ISCs and activated signaling mediated by IL-13‒IL-13R, which in turn initiated expression of the transcription factor Foxp1. Foxp1 is associated with ß-catenin in rendering its nuclear translocation, which caused activation of the ß-catenin pathway and the maintenance of Lgr5+ ISCs.


Subject(s)
Cell Self Renewal/immunology , Interleukin-13/metabolism , Intestinal Mucosa/immunology , RNA/metabolism , Stem Cells/physiology , Animals , Carrier Proteins/genetics , Cell Differentiation/immunology , Cell Self Renewal/genetics , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Humans , Interleukin-13/immunology , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-13 Receptor alpha1 Subunit/immunology , Interleukin-13 Receptor alpha1 Subunit/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , RNA/genetics , RNA/immunology , RNA, Circular , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/metabolism , Regeneration/genetics , Regeneration/immunology , Signal Transduction/genetics , Signal Transduction/immunology , beta Catenin/immunology , beta Catenin/metabolism
16.
Nat Immunol ; 20(8): 1023-1034, 2019 08.
Article in English | MEDLINE | ID: mdl-31263278

ABSTRACT

Stroke is a multiphasic process in which initial cerebral ischemia is followed by secondary injury from immune responses to ischemic brain components. Here we demonstrate that peripheral CD11b+CD45+ myeloid cells magnify stroke injury via activation of triggering receptor expressed on myeloid cells 1 (TREM1), an amplifier of proinflammatory innate immune responses. TREM1 was induced within hours after stroke peripherally in CD11b+CD45+ cells trafficking to ischemic brain. TREM1 inhibition genetically or pharmacologically improved outcome via protective antioxidant and anti-inflammatory mechanisms. Positron electron tomography imaging using radiolabeled antibody recognizing TREM1 revealed elevated TREM1 expression in spleen and, unexpectedly, in intestine. In the lamina propria, noradrenergic-dependent increases in gut permeability induced TREM1 on inflammatory Ly6C+MHCII+ macrophages, further increasing epithelial permeability and facilitating bacterial translocation across the gut barrier. Thus, following stroke, peripheral TREM1 induction amplifies proinflammatory responses to both brain-derived and intestinal-derived immunogenic components. Critically, targeting this specific innate immune pathway reduces cerebral injury.


Subject(s)
Brain/immunology , Intestinal Mucosa/immunology , Macrophages/immunology , Neutrophils/immunology , Stroke/pathology , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Animals , Brain/cytology , Cell Line , Immunity, Innate/immunology , Inflammation/pathology , Intestinal Mucosa/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , RAW 264.7 Cells
17.
Nat Immunol ; 20(12): 1644-1655, 2019 12.
Article in English | MEDLINE | ID: mdl-31636468

ABSTRACT

Invariant natural killer T (iNKT) cells recognize activating self and microbial lipids presented by CD1d. CD1d can also bind non-activating lipids, such as sphingomyelin. We hypothesized that these serve as endogenous regulators and investigated humans and mice deficient in acid sphingomyelinase (ASM), an enzyme that degrades sphingomyelin. We show that ASM absence in mice leads to diminished CD1d-restricted antigen presentation and iNKT cell selection in the thymus, resulting in decreased iNKT cell levels and resistance to iNKT cell-mediated inflammatory conditions. Defective antigen presentation and decreased iNKT cells are also observed in ASM-deficient humans with Niemann-Pick disease, and ASM activity in healthy humans correlates with iNKT cell phenotype. Pharmacological ASM administration facilitates antigen presentation and restores the levels of iNKT cells in ASM-deficient mice. Together, these results demonstrate that control of non-agonistic CD1d-associated lipids is critical for iNKT cell development and function in vivo and represents a tight link between cellular sphingolipid metabolism and immunity.


Subject(s)
Inflammation/immunology , Natural Killer T-Cells/immunology , Niemann-Pick Diseases/genetics , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelins/immunology , Thymus Gland/immunology , Animals , Antigen Presentation , Antigens, CD1d/metabolism , Cell Differentiation , Clonal Selection, Antigen-Mediated , Enzyme Replacement Therapy , Humans , Lymphocyte Activation , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelins/metabolism
18.
Cell ; 165(3): 668-78, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27062926

ABSTRACT

A key feature of inflammation is the timely recruitment of leukocytes, including monocytes, from blood into tissues, the latter maturing into macrophages over a period of 2-3 days. Using multi-channel spinning disk microscopy, we identified a rapid pathway of macrophage recruitment into an injured organ via a non-vascular route requiring no maturation from monocytes. In response to a sterile injury in liver, a reservoir of fully mature F4/80(hi)GATA6(+) peritoneal cavity macrophages rapidly invaded into afflicted tissue via direct recruitment across the mesothelium. The invasion was dependent on CD44 and DAMP molecule ATP and resulted in rapid replication and switching of macrophage toward an alternatively activated phenotype. These macrophages dismantled the nuclei of necrotic cells releasing DNA and forming a cover across the injury site. Rapid invasion of mature macrophages from body cavity with capacity for induction of reparative phenotype may impact altered tissues ranging from trauma to infections to cancer. VIDEO ABSTRACT.


Subject(s)
Cell Movement , Liver/physiology , Macrophages, Peritoneal/cytology , Wound Healing , Animals , Disease Models, Animal , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Inflammation , Leukocytes/immunology , Leukocytes/metabolism , Liver/injuries , Liver/pathology , Mice , Mice, Knockout , Specific Pathogen-Free Organisms
19.
Cell ; 166(3): 755-765, 2016 Jul 28.
Article in English | MEDLINE | ID: mdl-27372738

ABSTRACT

To provide a detailed analysis of the molecular components and underlying mechanisms associated with ovarian cancer, we performed a comprehensive mass-spectrometry-based proteomic characterization of 174 ovarian tumors previously analyzed by The Cancer Genome Atlas (TCGA), of which 169 were high-grade serous carcinomas (HGSCs). Integrating our proteomic measurements with the genomic data yielded a number of insights into disease, such as how different copy-number alternations influence the proteome, the proteins associated with chromosomal instability, the sets of signaling pathways that diverse genome rearrangements converge on, and the ones most associated with short overall survival. Specific protein acetylations associated with homologous recombination deficiency suggest a potential means for stratifying patients for therapy. In addition to providing a valuable resource, these findings provide a view of how the somatic genome drives the cancer proteome and associations between protein and post-translational modification levels and clinical outcomes in HGSC. VIDEO ABSTRACT.


Subject(s)
Neoplasm Proteins/genetics , Neoplasms, Cystic, Mucinous, and Serous/genetics , Ovarian Neoplasms/genetics , Proteome , Acetylation , Chromosomal Instability , DNA Repair , DNA, Neoplasm , Female , Gene Dosage , Humans , Mass Spectrometry , Phosphoproteins/genetics , Protein Processing, Post-Translational , Survival Analysis
20.
Mol Cell ; 83(4): 507-522.e6, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36630954

ABSTRACT

Genetic models suggested that SMARCA5 was required for DNA-templated events including transcription, DNA replication, and DNA repair. We engineered a degron tag into the endogenous alleles of SMARCA5, a catalytic component of the imitation switch complexes in three different human cell lines to define the effects of rapid degradation of this key regulator. Degradation of SMARCA5 was associated with a rapid increase in global nucleosome repeat length, which may allow greater chromatin compaction. However, there were few changes in nascent transcription within the first 6 h of degradation. Nevertheless, we demonstrated a requirement for SMARCA5 to control nucleosome repeat length at G1/S and during the S phase. SMARCA5 co-localized with CTCF and H2A.Z, and we found a rapid loss of CTCF DNA binding and disruption of nucleosomal phasing around CTCF binding sites. This spatiotemporal analysis indicates that SMARCA5 is continuously required for maintaining nucleosomal spacing.


Subject(s)
Chromatin , Chromosomal Proteins, Non-Histone , DNA Repair , Nucleosomes , Humans , Adenosine Triphosphatases/genetics , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Histones/genetics , Histones/metabolism , Nucleosomes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL