Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Article in English | MEDLINE | ID: mdl-38479808

ABSTRACT

OBJECTIVES: To stratify patients with mixed connective tissue disease (MCTD) based on their immunophenotype. METHODS: We analyzed the immunophenotype and transcriptome of 24 immune cell subsets from patients with MCTD, systemic lupus erythematosus (SLE), idiopathic inflammatory myopathy (IIM), and systemic sclerosis (SSc) from our functional genome database, ImmuNexUT (https://www.immunexut.org/). MCTD patients were stratified by employing machine learning models including Random Forest, trained by immunophenotyping data from SLE, IIM, and SSc patients. Transcriptomes were analyzed with gene set variation analysis (GSVA) and clinical features of MCTD subgroups were compared. RESULTS: This study included 215 patients, including 22 patients with MCTD. Machine learning models, constructed to classify SLE, IIM, and SSc patients based on immunophenotyping, were applied to MCTD patients, resulting in 16 classified as SLE-immunophenotype and 6 as non-SLE-immunophenotype. Among MCTD patients, patients with the SLE-immunophenotype had higher proportions of Th1 cells [2.85% (interquartile range (IQR) 1.54-3.91) vs 1.33% (IQR 0.99-1.74) p= 0.027] and plasmablasts [6.35% (IQR 4.17-17.49) vs 2.00% (IQR 1.20-2.80) p= 0.010]. Notably, the number of SLE-related symptoms was higher in patients with the SLE-immunophenotype [2.0 (IQR 1.0-2.0) vs 1.0 (IQR1.0-1.0) p= 0.038]. Moreover, GSVA scores of interferon-α and -γ responses were significantly higher in patients with the SLE-immunophenotype in central memory CD8+ T cells, while hedgehog signalling was higher in non-SLE-immunophenotype patients in 5 cell subsets. CONCLUSION: This study describes the stratification of MCTD patients based on immunophenotyping, suggesting the presence of distinct immunological processes behind the clinical subtypes of MCTD.

2.
Cell ; 184(11): 3006-3021.e17, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33930287

ABSTRACT

Genetic studies have revealed many variant loci that are associated with immune-mediated diseases. To elucidate the disease pathogenesis, it is essential to understand the function of these variants, especially under disease-associated conditions. Here, we performed a large-scale immune cell gene-expression analysis, together with whole-genome sequence analysis. Our dataset consists of 28 distinct immune cell subsets from 337 patients diagnosed with 10 categories of immune-mediated diseases and 79 healthy volunteers. Our dataset captured distinctive gene-expression profiles across immune cell types and diseases. Expression quantitative trait loci (eQTL) analysis revealed dynamic variations of eQTL effects in the context of immunological conditions, as well as cell types. These cell-type-specific and context-dependent eQTLs showed significant enrichment in immune disease-associated genetic variants, and they implicated the disease-relevant cell types, genes, and environment. This atlas deepens our understanding of the immunogenetic functions of disease-associated variants under in vivo disease conditions.


Subject(s)
Gene Expression Regulation/genetics , Gene Expression/immunology , Immune System Diseases/genetics , Adult , Female , Gene Expression/genetics , Gene Expression Regulation/immunology , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Humans , Immune System/cytology , Immune System/metabolism , Immune System Diseases/metabolism , Immune System Diseases/physiopathology , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , Quantitative Trait Loci/immunology , Transcriptome/genetics , Whole Genome Sequencing/methods
3.
Proc Natl Acad Sci U S A ; 116(47): 23653-23661, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31694883

ABSTRACT

The activation of innate immune receptors by pathogen-associated molecular patterns (PAMPs) is central to host defense against infections. On the other hand, these receptors are also activated by immunogenic damage-associated molecular patterns (DAMPs), typically released from dying cells, and the activation can evoke chronic inflammatory or autoimmune disorders. One of the best known receptors involved in the immune pathogenesis is Toll-like receptor 7 (TLR7), which recognizes RNA with single-stranded structure. However, the causative DAMP RNA(s) in the pathogenesis has yet to be identified. Here, we first developed a chemical compound, termed KN69, that suppresses autoimmunity in several established mouse models. A subsequent search for KN69-binding partners led to the identification of U11 small nuclear RNA (U11snRNA) as a candidate DAMP RNA involved in TLR7-induced autoimmunity. We then showed that U11snRNA robustly activated the TLR7 pathway in vitro and induced arthritis disease in vivo. We also found a correlation between high serum level of U11snRNA and autoimmune diseases in human subjects and established mouse models. Finally, by revealing the structural basis for U11snRNA's ability to activate TLR7, we developed more potent TLR7 agonists and TLR7 antagonists, which may offer new therapeutic approaches for autoimmunity or other immune-driven diseases. Thus, our study has revealed a hitherto unknown immune function of U11snRNA, providing insight into TLR7-mediated autoimmunity and its potential for further therapeutic applications.


Subject(s)
Membrane Glycoproteins/agonists , RNA, Small Nuclear/immunology , Toll-Like Receptor 7/agonists , Adult , Alarmins/chemistry , Animals , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/immunology , Autoimmune Diseases/blood , Autoimmune Diseases/immunology , Base Sequence , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immunosuppressive Agents/chemical synthesis , Immunosuppressive Agents/pharmacology , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology , Membrane Glycoproteins/deficiency , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Middle Aged , RNA/immunology , RNA/metabolism , Ribonucleoproteins, Small Nuclear/chemistry , Ribonucleoproteins, Small Nuclear/immunology , Sequence Analysis, RNA , Toll-Like Receptor 7/deficiency , Young Adult
4.
Environ Pollut ; 252(Pt A): 205-215, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31151059

ABSTRACT

This study aimed to evaluate and qualify field-based potential risks of seven neonicotinoid and phenylpyrazole (fipronil) insecticides on aquatic invertebrates, including estuary-resident marine crustaceans. One hundred and ninety-three estuarine water samples, with salinity ranging from 0.5 to 32.7, were collected from four estuarine sites in the Seto Inland Sea of Japan, in 2015-2018 and the insecticide levels were measured. Five neonicotinoid and fipronil insecticides were successfully identified, and their occurrence varied temporally. Marine crustaceans were simultaneously harvested every month from one of the estuarine water sampling sites in 2015-2017. Three predominant crustacean species, kuruma prawn (Penaeus japonicus), sand shrimp (Crangon uritai), and mysid (Neomysis awatschensis), were captured and their seasonal presence was species independent. A 96-h laboratory toxicity study with the insecticides using kuruma prawn, sand shrimp, and a surrogate mysid species (Americamysis bahia) indicated that fipronil exerted the highest toxicity to the three crustaceans. Using both toxicity data and insecticide occurrence in estuarine water (salinity ≥10, n = 169), the potential risks on the three marine crustaceans were quantified by calculating the proportion of mixture toxicity effects (Pmix). The Pmix of seven neonicotinoids on the crustaceans was less than 0.8%, which is likely to be too low to indicate adverse effects caused by the insecticides. However, short temporal detection of fipronil (exclusively in June and July) significantly affected the Pmix, which presented the maximal Pmix values of 21%, 3.4%, and 72% for kuruma prawn, sand shrimp, and mysid, respectively, indicating a significant effect on the organisms. As for estuarine water (salinity <10), some water samples contained imidacloprid and fipronil exceeding the freshwater benchmarks for aquatic invertebrates. The present study provides novel insights into the seasonally varying risks of insecticides to estuarine crustaceans and highlights the importance of considering whether ecological risk periods coincide with crustacean presence.


Subject(s)
Crangonidae/drug effects , Insecticides/analysis , Neonicotinoids/analysis , Nitro Compounds/analysis , Penaeidae/drug effects , Pyrazoles/analysis , Water Pollutants, Chemical/toxicity , Animals , Ecology , Estuaries , Fresh Water/chemistry , Insecticides/toxicity , Japan , Neonicotinoids/toxicity , Nitro Compounds/toxicity , Pyrazoles/toxicity , Water Pollutants, Chemical/analysis
5.
Proc Natl Acad Sci U S A ; 113(14): 3844-9, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27001836

ABSTRACT

Cellular components released into the external milieu as a result of cell death and sensed by the body are generally termed damage-associated molecular patterns (DAMPs). Although DAMPs are conventionally thought to be protective to the host by evoking inflammatory responses important for immunity and wound repair, there is the prevailing notion that dysregulated release of DAMPs can also underlie or exacerbate disease development. However, the critical issue for how resultant DAMP-mediated responses are regulated has heretofore not been fully addressed. In the present study, we identify prostaglandin E2 (PGE2) as a DAMP that negatively regulates immune responses. We show that the production of PGE2 is augmented under cell death-inducing conditions via the transcriptional induction of the cyclooxygenase 2 (COX2) gene and that cell-released PGE2 suppresses the expression of genes associated with inflammation, thereby limiting the cell's immunostimulatory activities. Consistent with this, inhibition of the PGE2 synthesis pathway potentiates the inflammation induced by dying cells. We also provide in vivo evidence for a protective role of PGE2 released upon acetaminophen-induced liver injury as well as a pathogenic role for PGE2 during tumor cell growth. Our study places this classically known lipid mediator in an unprecedented context-that is, an inhibitory DAMP vis-à-vis activating DAMPs, which may have translational implications for designing more effective therapeutic regimens for inflammation-associated diseases.


Subject(s)
Alarmins/metabolism , Cell Death/immunology , Cyclooxygenase 2/biosynthesis , Dinoprostone/metabolism , Inflammation/pathology , Acetaminophen/adverse effects , Animals , Cell Death/physiology , Cell Line, Tumor , Chemical and Drug Induced Liver Injury/immunology , HeLa Cells , Humans , Inflammation/immunology , Lipopolysaccharides , Mice , Mice, Inbred C57BL
6.
Proc Natl Acad Sci U S A ; 113(5): 1351-6, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26787880

ABSTRACT

Blunting immunopathology without abolishing host defense is the foundation for safe and effective modulation of infectious and autoimmune diseases. Sphingosine 1-phosphate receptor 1 (S1PR1) agonists are effective in treating infectious and multiple autoimmune pathologies; however, mechanisms underlying their clinical efficacy are yet to be fully elucidated. Here, we uncover an unexpected mechanism of convergence between S1PR1 and interferon alpha receptor 1 (IFNAR1) signaling pathways. Activation of S1PR1 signaling by pharmacological tools or endogenous ligand sphingosine-1 phosphate (S1P) inhibits type 1 IFN responses that exacerbate numerous pathogenic conditions. Mechanistically, S1PR1 selectively suppresses the type I IFN autoamplification loop in plasmacytoid dendritic cells (pDCs), a specialized DC subset, for robust type I IFN release. S1PR1 agonist suppression is pertussis toxin-resistant, but inhibited by an S1PR1 C-terminal-derived transactivating transcriptional activator (Tat)-fusion peptide that blocks receptor internalization. S1PR1 agonist treatment accelerates turnover of IFNAR1, suppresses signal transducer and activator of transcription 1 (STAT1) phosphorylation, and down-modulates total STAT1 levels, thereby inactivating the autoamplification loop. Inhibition of S1P-S1PR1 signaling in vivo using the selective antagonist Ex26 significantly elevates IFN-α production in response to CpG-A. Thus, multiple lines of evidence demonstrate that S1PR1 signaling sets the sensitivity of pDC amplification of IFN responses, thereby blunting pathogenic immune responses. These data illustrate a lipid G-protein coupled receptor (GPCR)-IFNAR1 regulatory loop that balances effective and detrimental immune responses and elevated endogenous S1PR1 signaling. This mechanism will likely be advantageous in individuals subject to a range of inflammatory conditions.


Subject(s)
Dendritic Cells/metabolism , Interferon-alpha/metabolism , Receptor, Interferon alpha-beta/metabolism , Receptors, Lysosphingolipid/physiology , Animals , Mice , Mice, Knockout , Proteolysis , Receptor, Interferon alpha-beta/genetics
7.
Proc Natl Acad Sci U S A ; 110(49): 19884-9, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24248350

ABSTRACT

A major function of innate immune receptors is to recognize pathogen-associated molecular patterns and then evoke immune responses appropriate to the nature of the invading pathogen(s). Because innate immune cells express various types of these receptors, distinct combinations of signaling pathways are activated in response to a given pathogen. Although the conventional wisdom is that these signaling pathways cooperate with one another to ensure an effective host response, a more nuanced view recognizes antagonism between the individual pathways, where the attenuation of a signaling pathway(s) by others may shape the immune response. In this study, we show that, on Listeria monocytogenes infection, Toll-like receptor-triggered MyD88 signaling pathways suppress type I IFN gene induction, which is detrimental to macrophage bactericidal activity. These pathways target and suppress the IFN regulatory factor 3 (IRF3) transcription factor that is activated by the stimulator of IFN genes-TANK-binding kinase-1 kinase pathway. We also provide evidence for the involvement of the MAPK phosphatase family members, which renders IRF3 hypophosphorylated on Toll-like receptor signaling by enhancing the formation of an MAPK phosphatase-IRF3-TANK-binding kinase-1 ternary complex. This study, therefore, reveals a hitherto unrecognized and important contribution of a beneficial innate signaling interference against bacterial infections.


Subject(s)
Immunity, Innate/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Multiprotein Complexes/immunology , Signal Transduction/immunology , Toll-Like Receptors/metabolism , Animals , Colony-Forming Units Assay , Dual Specificity Phosphatase 1/metabolism , Immunoblotting , Immunoprecipitation , Interferon Regulatory Factor-3/antagonists & inhibitors , Interferon Regulatory Factor-3/metabolism , Interferon Type I/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Protein Serine-Threonine Kinases/metabolism , Real-Time Polymerase Chain Reaction
8.
Article in English | MEDLINE | ID: mdl-24110539

ABSTRACT

To improve the throughput of high intensity focused ultrasound (HIFU) treatment, we have considered a focus switching method at two points. For this method, it is necessary to evaluate the thermal distribution under exposure to ultrasound. The thermal distribution was measured using a prototype thin-film thermocouple array, which has the advantage of minimizing the influence of the thermocouple on the acoustic and temperature fields. Focus switching was employed to enlarge the area of temperature increase and evaluate the proposed evaluation parameters with respect to safety and uniformity. The results indicate that focus switching can effectively expand the thermal lesion while maintaining a steep thermal boundary. In addition, the influence caused by the thin-film thermocouple array was estimated experimentally. This thermocouple was demonstrated to be an effective tool for the measurement of temperature distributions induced by HIFU.


Subject(s)
High-Intensity Focused Ultrasound Ablation/methods , Humans , Hyperthermia, Induced , Magnetic Resonance Imaging , Neoplasms/therapy
9.
Proc Natl Acad Sci U S A ; 109(51): 21016-21, 2012 Dec 18.
Article in English | MEDLINE | ID: mdl-23213237

ABSTRACT

The large intestinal epithelial cells and immune cells are exposed to a variety of molecules derived from commensal microbiota that can activate innate receptors, such as Toll-like receptors (TLRs) and retinoic acid-inducible gene-I-like receptors (RLRs). Although the activation of these receptors is known to be critical for homeostasis of the large intestine, the underlying gene regulatory mechanisms are not well understood. Here, we show that IFN regulatory factor (IRF)3 is critical for the suppression of dextran sulfate sodium-induced colitis. IRF3-deficient mice exhibited lethal defects in the inflammatory and recovery phases of the colitis, accompanied by marked defects in the gene induction for thymic stromal lymphopoietin (TSLP), a cytokine known to be essential for protection of the large intestine. We further provide evidence that DNA and RNA of the large intestinal contents are critical for Tslp gene induction via IRF3 activation by cytosolic nucleic acid receptors. We also demonstrate that IRF3 indeed activates the gene promoter of Tslp via IRF-binding sequences. This newly identified intestinal gene regulatory mechanism, wherein IRF3 activated by microbiota-derived nucleic acids plays a critical role in intestinal homeostasis, may have clinical implication in colonic inflammatory disorders.


Subject(s)
Cytokines/genetics , Gene Expression Regulation, Bacterial , Interferon Regulatory Factor-3/physiology , Intestines/microbiology , Metagenome , Animals , Colitis/microbiology , Cytokines/metabolism , Cytosol/metabolism , DNA/metabolism , Homeostasis , Interferon Regulatory Factor-3/genetics , Mice , Models, Biological , RNA/metabolism , Tretinoin/metabolism , Thymic Stromal Lymphopoietin
10.
Nat Immunol ; 13(7): 659-66, 2012 May 20.
Article in English | MEDLINE | ID: mdl-22610141

ABSTRACT

Although the mechanisms by which innate pathogen-recognition receptors enhance adaptive immune responses are increasingly well understood, whether signaling events from distinct classes of receptors affect each other in modulating adaptive immunity remains unclear. We found here that the activation of cytosolic RIG-I-like receptors (RLRs) resulted in the selective suppression of transcription of the gene encoding the p40 subunit of interleukin 12 (Il12b) that was effectively induced by the activation of Toll-like receptors (TLRs). The RLR-activated transcription factor IRF3 bound dominantly, relative to IRF5, to the Il12b promoter, where it interfered with the TLR-induced assembly of a productive transcription-factor complex. The activation of RLRs in mice attenuated TLR-induced responses of the T helper type 1 cell (T(H)1 cell) and interleukin 17-producing helper T cell (T(H)17 cell) subset types and, consequently, viral infection of mice caused death at sublethal doses of bacterial infection. The innate immune receptor cross-interference we describe may have implications for infection-associated clinical episodes.


Subject(s)
Signal Transduction/immunology , T-Lymphocytes/immunology , Toll-Like Receptors/immunology , Amino Acid Sequence , Animals , Bacterial Infections/immunology , Cells, Cultured , Gene Expression Regulation/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factors/metabolism , Interleukin-12 Subunit p40/metabolism , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Promoter Regions, Genetic , Th1 Cells/immunology , Th17 Cells/immunology , Transcription Factors/metabolism , Virus Diseases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL