Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 222
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 23(10): 1457-1469, 2022 10.
Article in English | MEDLINE | ID: mdl-36151396

ABSTRACT

In lupus, Toll-like receptor 7 (TLR7) and TLR9 mediate loss of tolerance to RNA and DNA, respectively. Yet, TLR7 promotes disease, while TLR9 protects from disease, implying differences in signaling. To dissect this 'TLR paradox', we generated two TLR9 point mutants (lacking either ligand (TLR9K51E) or MyD88 (TLR9P915H) binding) in lupus-prone MRL/lpr mice. Ameliorated disease of Tlr9K51E mice compared to Tlr9-/- controls revealed a TLR9 'scaffold' protective function that is ligand and MyD88 independent. Unexpectedly, Tlr9P915H mice were more protected than both Tlr9K51E and Tlr9WT mice, suggesting that TLR9 also possesses ligand-dependent, but MyD88-independent, regulatory signaling and MyD88-mediated proinflammatory signaling. Triple-mixed bone marrow chimeras showed that TLR9-MyD88-independent regulatory roles were B cell intrinsic and restrained differentiation into pathogenic age-associated B cells and plasmablasts. These studies reveal MyD88-independent regulatory roles of TLR9, shedding light on the biology of endosomal TLRs.


Subject(s)
Toll-Like Receptor 7 , Toll-Like Receptor 9 , Animals , DNA , Ligands , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , RNA , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism
2.
Nat Immunol ; 19(10): 1071-1082, 2018 10.
Article in English | MEDLINE | ID: mdl-30201994

ABSTRACT

TLR3 is a sensor of double-stranded RNA that is indispensable for defense against infection with herpes simplex virus type 1 (HSV-1) in the brain. We found here that TLR3 was required for innate immune responses to HSV-1 in neurons and astrocytes. During infection with HSV-1, TLR3 recruited the metabolic checkpoint kinase complex mTORC2, which led to the induction of chemokines and trafficking of TLR3 to the cell periphery. Such trafficking enabled the activation of molecules (including mTORC1) required for the induction of type I interferons. Intracranial infection of mice with HSV-1 was exacerbated by impairment of TLR3 responses with an inhibitor of mTOR and was significantly 'rescued' by potentiation of TLR3 responses with an agonistic antibody to TLR3. These results suggest that the TLR3-mTORC2 axis might be a therapeutic target through which to combat herpes simplex encephalitis.


Subject(s)
Encephalitis, Herpes Simplex/immunology , Mechanistic Target of Rapamycin Complex 2/immunology , Toll-Like Receptor 3/immunology , Animals , Herpesvirus 1, Human , Immunity, Innate/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NIH 3T3 Cells
3.
Nature ; 625(7993): 166-174, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38057662

ABSTRACT

Myeloid cells are known to suppress antitumour immunity1. However, the molecular drivers of immunosuppressive myeloid cell states are not well defined. Here we used single-cell RNA sequencing of human and mouse non-small cell lung cancer (NSCLC) lesions, and found that in both species the type 2 cytokine interleukin-4 (IL-4) was predicted to be the primary driver of the tumour-infiltrating monocyte-derived macrophage phenotype. Using a panel of conditional knockout mice, we found that only deletion of the IL-4 receptor IL-4Rα in early myeloid progenitors in bone marrow reduced tumour burden, whereas deletion of IL-4Rα in downstream mature myeloid cells had no effect. Mechanistically, IL-4 derived from bone marrow basophils and eosinophils acted on granulocyte-monocyte progenitors to transcriptionally programme the development of immunosuppressive tumour-promoting myeloid cells. Consequentially, depletion of basophils profoundly reduced tumour burden and normalized myelopoiesis. We subsequently initiated a clinical trial of the IL-4Rα blocking antibody dupilumab2-5 given in conjunction with PD-1/PD-L1 checkpoint blockade in patients with relapsed or refractory NSCLC who had progressed on PD-1/PD-L1 blockade alone (ClinicalTrials.gov identifier NCT05013450 ). Dupilumab supplementation reduced circulating monocytes, expanded tumour-infiltrating CD8 T cells, and in one out of six patients, drove a near-complete clinical response two months after treatment. Our study defines a central role for IL-4 in controlling immunosuppressive myelopoiesis in cancer, identifies a novel combination therapy for immune checkpoint blockade in humans, and highlights cancer as a systemic malady that requires therapeutic strategies beyond the primary disease site.


Subject(s)
Bone Marrow , Carcinogenesis , Interleukin-4 , Myelopoiesis , Signal Transduction , Animals , Humans , Mice , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Bone Marrow/drug effects , Bone Marrow/metabolism , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Immune Checkpoint Inhibitors/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Interleukin-4/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Monocytes/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Recurrence , Signal Transduction/drug effects
4.
Nature ; 622(7981): 188-194, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37704723

ABSTRACT

Inflammasome sensors detect pathogen- and danger-associated molecular patterns and promote inflammation and pyroptosis1. NLRP1 was the first inflammasome sensor to be described, and its hyperactivation is linked to autoinflammatory disease and cancer2-6. However, the mechanism underlying the activation and regulation of NLRP1 has not been clearly elucidated4,7,8. Here we identify ubiquitously expressed endogenous thioredoxin (TRX) as a binder of NLRP1 and a suppressor of the NLRP1 inflammasome. The cryo-electron microscopy structure of human NLRP1 shows NLRP1 bound to Spodoptera frugiperda TRX. Mutagenesis studies of NLRP1 and human TRX show that TRX in the oxidized form binds to the nucleotide-binding domain subdomain of NLRP1. This observation highlights the crucial role of redox-active cysteines of TRX in NLRP1 binding. Cellular assays reveal that TRX suppresses NLRP1 inflammasome activation and thus negatively regulates NLRP1. Our data identify the TRX system as an intrinsic checkpoint for innate immunity and provide opportunities for future therapeutic intervention in NLRP1 inflammasome activation targeting this system.


Subject(s)
Inflammasomes , NLR Proteins , Thioredoxins , Humans , Cryoelectron Microscopy , Inflammasomes/metabolism , NLR Proteins/antagonists & inhibitors , NLR Proteins/chemistry , NLR Proteins/metabolism , NLR Proteins/ultrastructure , Thioredoxins/chemistry , Thioredoxins/metabolism , Spodoptera , Insect Proteins , Oxidation-Reduction , Cysteine/metabolism , Immunity, Innate
5.
Immunity ; 48(4): 649-658.e4, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29625894

ABSTRACT

Toll-like receptor 9 (TLR9) recognizes DNA containing CpG motifs derived from bacteria and viruses and activates the innate immune response to eliminate them. TLR9 is known to bind to CpG DNA, and here, we identified another DNA binding site in TLR9 that binds DNA containing cytosine at the second position from the 5' end (5'-xCx DNA). 5'-xCx DNAs bound to TLR9 in the presence of CpG DNA and cooperatively promoted dimerization and activation of TLR9. Binding at both sites was important for efficient activation of TLR9. The 5'-xCx DNA bound the site corresponding to the nucleoside binding site in TLR7 and TLR8 as revealed by the structural analysis. This study revealed that TLR9 recognizes two types of DNA through its two binding sites for efficient activation. This information may contribute to the development of drugs that control the activity of TLR9.


Subject(s)
CpG Islands/genetics , DNA-Binding Proteins/metabolism , DNA/metabolism , Nucleotide Motifs/genetics , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism , Animals , Binding Sites/genetics , Cell Line , DNA/genetics , Dimerization , Drosophila , Enzyme Activation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/genetics , Toll-Like Receptor 8/metabolism
6.
Immunity ; 48(5): 911-922.e7, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768176

ABSTRACT

Unc-93 homolog B1 (UNC93B1) is a key regulator of nucleic acid (NA)-sensing Toll-like receptors (TLRs). Loss of NA-sensing TLR responses in UNC93B1-deficient patients facilitates Herpes simplex virus type 1 (HSV-1) encephalitis. UNC93B1 is thought to guide NA-sensing TLRs from the endoplasmic reticulum (ER) to their respective endosomal signaling compartments and to guide the flagellin receptor TLR5 to the cell surface, raising the question of how UNC93B1 mediates differential TLR trafficking. Here, we report that UNC93B1 regulates a step upstream of the differential TLR trafficking process. We discovered that UNC93B1 deficiency resulted in near-complete loss of TLR3 and TLR7 proteins in primary splenic mouse dendritic cells and macrophages, showing that UNC93B1 is critical for maintaining TLR expression. Notably, expression of an ER-retained UNC93B1 version was sufficient to stabilize TLRs and largely restore endosomal TLR trafficking and activity. These data are critical for an understanding of how UNC93B1 can regulate the function of a broad subset of TLRs.


Subject(s)
Endosomes/immunology , Membrane Transport Proteins/immunology , Molecular Chaperones/immunology , Toll-Like Receptors/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , HEK293 Cells , Humans , Macrophages/immunology , Macrophages/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Protein Stability , Protein Transport/immunology , Signal Transduction/genetics , Signal Transduction/immunology , THP-1 Cells , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
7.
Int Immunol ; 36(4): 183-196, 2024 Mar 09.
Article in English | MEDLINE | ID: mdl-38147536

ABSTRACT

In sarcoidosis, granulomas develop in multiple organs including the liver and lungs. Although mechanistic target of rapamycin complex 1 (mTORC1) activation in macrophages drives granuloma development in sarcoidosis by enhancing macrophage proliferation, little is known about the macrophage subsets that proliferate and mature into granuloma macrophages. Here, we show that aberrantly increased monocytopoiesis gives rise to granulomas in a sarcoidosis model, in which Tsc2, a negative regulator of mTORC1, is conditionally deleted in CSF1R-expressing macrophages (Tsc2csf1rΔ mice). In Tsc2csf1rΔ mice, common myeloid progenitors (CMPs), granulocyte-monocyte progenitors (GMPs), common monocyte progenitors / monocyte progenitors (cMoPs / MPs), inducible monocyte progenitors (iMoPs), and Ly6Cint CX3CR1low CD14- immature monocytes (iMOs), but not monocyte-dendritic cell progenitors (MDPs) and common dendritic cell progenitors (CDPs), accumulated and proliferated in the spleen. Consistent with this, monocytes, neutrophils, and neutrophil-like monocytes increased in the spleens of Tsc2csf1rΔ mice, whereas dendritic cells did not. The adoptive transfer of splenic iMOs into wild-type mice gave rise to granulomas in the liver and lungs. In these target organs, iMOs matured into Ly6Chi classical monocytes/macrophages (cMOs). Giant macrophages (gMAs) also accumulated in the liver and lungs, which were similar to granuloma macrophages in expression of cell surface markers such as MerTK and SLAMF7. Furthermore, the gMA-specific genes were expressed in human macrophages from sarcoidosis skin lesions. These results suggest that mTORC1 drives granuloma development by promoting the proliferation of monocyte/neutrophil progenitors and iMOs predominantly in the spleen, and that proliferating iMOs mature into cMOs and then gMAs to give rise to granuloma after migration into the liver and lungs in sarcoidosis.


Subject(s)
Macrophages , Sarcoidosis , Mice , Humans , Animals , Cell Differentiation , Macrophages/metabolism , Monocytes/metabolism , Granuloma/metabolism , Granuloma/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism
8.
Immunity ; 45(4): 737-748, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27742543

ABSTRACT

Toll-like receptor 7 (TLR7) is a single-stranded RNA (ssRNA) sensor in innate immunity and also responds to guanosine and chemical ligands, such as imidazoquinoline compounds. However, TLR7 activation mechanism by these ligands remain largely unknown. Here, we generated crystal structures of three TLR7 complexes, and found that all formed an activated m-shaped dimer with two ligand-binding sites. The first site conserved in TLR7 and TLR8 was used for small ligand-binding essential for its activation. The second site spatially distinct from that of TLR8 was used for a ssRNA-binding that enhanced the affinity of the first-site ligands. The first site preferentially recognized guanosine and the second site specifically bound to uridine moieties in ssRNA. Our structural, biochemical, and mutagenesis studies indicated that TLR7 is a dual receptor for guanosine and uridine-containing ssRNA. Our findings have important implications for understanding of TLR7 function, as well as for therapeutic manipulation of TLR7 activation.


Subject(s)
Guanosine/metabolism , RNA/metabolism , Toll-Like Receptor 7/chemistry , Toll-Like Receptor 7/metabolism , Animals , Binding Sites/immunology , Cell Line , Drosophila , Guanosine/immunology , HEK293 Cells , Humans , Immunity, Innate/immunology , Ligands , Macaca mulatta , RNA/immunology , Toll-Like Receptor 7/immunology
9.
J Allergy Clin Immunol ; 153(5): 1344-1354.e5, 2024 May.
Article in English | MEDLINE | ID: mdl-38336257

ABSTRACT

BACKGROUND: Atopic dermatitis skin lesions exhibit increased infiltration by basophils. Basophils produce IL-4, which plays an important role in the pathogenesis of atopic dermatitis. OBJECTIVE: We sought to determine the role of basophils in a mouse model of antigen-driven allergic skin inflammation. METHODS: Wild-type mice, mice with selective and inducible depletion of basophils, and mice expressing Il4-driven enhanced green fluorescent protein were subjected to epicutaneous sensitization with ovalbumin or saline. Sensitized skin was examined by histology for epidermal thickening. Cells were analyzed for surface markers and intracellular expression of enhanced green fluorescent protein by flow cytometry. Gene expression was evaluated by real-time reverse transcription-quantitative PCR. RESULTS: Basophils were important for epidermal hyperplasia, dermal infiltration by CD4+ T cells, mast cells, and eosinophils in ovalbumin-sensitized mouse skin and for the local and systemic TH2 response to epicutaneous sensitization. Moreover, basophils were the major source of IL-4 in epicutaneous-sensitized mouse skin and promote the ability of dendritic cells to drive TH2 polarization of naive T cells. CONCLUSION: Basophils play an important role in the development of allergic skin inflammation induced by cutaneous exposure to antigen in mice.


Subject(s)
Basophils , Dermatitis, Atopic , Interleukin-4 , Ovalbumin , Th2 Cells , Animals , Basophils/immunology , Mice , Interleukin-4/immunology , Interleukin-4/genetics , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Ovalbumin/immunology , Th2 Cells/immunology , Skin/immunology , Skin/pathology , Mice, Inbred C57BL , Mice, Inbred BALB C , Disease Models, Animal , Dendritic Cells/immunology , Mice, Transgenic , Mast Cells/immunology
10.
Adv Exp Med Biol ; 1444: 97-108, 2024.
Article in English | MEDLINE | ID: mdl-38467975

ABSTRACT

Nucleic acid (NA)-sensing Toll-like receptors (TLRs) reside in the endosomal compartment of innate immune cells, such as macrophages and dendritic cells. NAs transported to the endosomal compartment are degraded by DNases and RNases. Degradation products, including single-stranded DNA, oligoRNA, and nucleosides, are recognized by TLR7, TLR8, and TLR9 to drive the defense responses against pathogens. NA degradation influences endosomal TLR responses by generating and degrading TLR ligands. TLR ligand accumulation because of impaired NA degradation causes constitutive TLR activation, leading to autoinflammatory and autoimmune diseases. Furthermore, some genes associated with these diseases promote endosomal TLR responses. Therefore, endosomal TLRs are promising therapeutic targets for TLR-mediated inflammatory diseases, and novel drugs targeting TLRs are being developed.


Subject(s)
Autoimmune Diseases , Nucleic Acids , Humans , Toll-Like Receptors , Autoimmune Diseases/drug therapy , Nucleic Acids/metabolism , Macrophages/metabolism
11.
J Virol ; 96(10): e0030622, 2022 05 25.
Article in English | MEDLINE | ID: mdl-35475666

ABSTRACT

This study developed a system consisting of two rounds of screening cellular proteins involved in the nuclear egress of herpes simplex virus 1 (HSV-1). Using this system, we first screened cellular proteins that interacted with the HSV-1 nuclear egress complex (NEC) consisting of UL34 and UL31 in HSV-1-infected cells, which are critical for the nuclear egress of HSV-1, by tandem affinity purification coupled with mass spectrometry-based proteomics technology. Next, we performed CRISPR/Cas9-based screening of live HSV-1-infected reporter cells under fluorescence microscopy using single guide RNAs targeting the cellular proteins identified in the first proteomic screening to detect the mislocalization of the lamin-associated protein emerin, which is a phenotype for defects in HSV-1 nuclear egress. This study focused on a cellular orphan transporter SLC35E1, one of the cellular proteins identified by the screening system. Knockout of SLC35E1 reduced HSV-1 replication and induced membranous invaginations containing perinuclear enveloped virions (PEVs) adjacent to the nuclear membrane (NM), aberrant accumulation of PEVs in the perinuclear space between the inner and outer NMs and the invagination structures, and mislocalization of the NEC. These effects were similar to those of previously reported mutation(s) in HSV-1 proteins and depletion of cellular proteins that are important for HSV-1 de-envelopment, one of the steps required for HSV-1 nuclear egress. Our newly established screening system enabled us to identify a novel cellular protein required for efficient HSV-1 de-envelopment. IMPORTANCE The identification of cellular protein(s) that interact with viral effector proteins and function in important viral procedures is necessary for enhancing our understanding of the mechanics of various viral processes. In this study, we established a new system consisting of interactome screening for the herpes simplex virus 1 (HSV-1) nuclear egress complex (NEC), followed by loss-of-function screening to target the identified putative NEC-interacting cellular proteins to detect a defect in HSV-1 nuclear egress. This newly established system identified SLC35E1, an orphan transporter, as a novel cellular protein required for efficient HSV-1 de-envelopment, providing an insight into the mechanisms involved in this viral procedure.


Subject(s)
Herpesvirus 1, Human , Membrane Transport Proteins , Virus Release , Animals , CRISPR-Cas Systems , Chlorocebus aethiops , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Humans , Membrane Transport Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins , Proteomics , Vero Cells , Viral Proteins/metabolism
12.
Ann Rheum Dis ; 82(9): 1153-1161, 2023 09.
Article in English | MEDLINE | ID: mdl-37400117

ABSTRACT

OBJECTIVES: Recent studies demonstrate that extracellular-released aminoacyl-tRNA synthetases (aaRSs) play unique roles in immune responses and diseases. This study aimed to understand the role of extracellular aaRSs in the pathogenesis of rheumatoid arthritis (RA). METHODS: Primary macrophages and fibroblast-like synoviocytes were cultured with aaRSs. aaRS-induced cytokine production including IL-6 and TNF-α was detected by ELISA. Transcriptomic features of aaRS-stimulated macrophages were examined using RNA-sequencing. Serum and synovial fluid (SF) aaRS levels in patients with RA were assessed using ELISA. Peptidyl arginine deiminase (PAD) 4 release from macrophages stimulated with aaRSs was detected by ELISA. Citrullination of aaRSs by themselves was examined by immunoprecipitation and western blotting. Furthermore, aaRS inhibitory peptides were used for inhibition of arthritis in two mouse RA models, collagen-induced arthritis and collagen antibody-induced arthritis. RESULTS: All 20 aaRSs functioned as alarmin; they induced pro-inflammatory cytokines through the CD14-MD2-TLR4 axis. Stimulation of macrophages with aaRSs displayed persistent innate inflammatory responses. Serum and SF levels of many aaRSs increased in patients with RA compared with control subjects. Furthermore, aaRSs released PAD4 from living macrophages, leading to their citrullination. We demonstrate that aaRS inhibitory peptides suppress cytokine production and PAD4 release by aaRSs and alleviate arthritic symptoms in a mouse RA model. CONCLUSIONS: Our findings uncovered the significant role of aaRSs as a novel alarmin in RA pathogenesis, indicating that their blocking agents are potent antirheumatic drugs.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Animals , Mice , Alarmins , Cells, Cultured , Cytokines , Disease Models, Animal , Fibroblasts/pathology , Inflammation , Synovial Fluid , Humans
13.
Immunity ; 40(4): 530-41, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24726878

ABSTRACT

Mast cells (MCs) mature locally, thus possessing tissue-dependent phenotypes for their critical roles in both protective immunity against pathogens and the development of allergy or inflammation. We previously reported that MCs highly express P2X7, a receptor for extracellular ATP, in the colon but not in the skin. The ATP-P2X7 pathway induces MC activation and consequently exacerbates the inflammation. Here, we identified the mechanisms by which P2X7 expression on MCs is reduced by fibroblasts in the skin, but not in the other tissues. The retinoic-acid-degrading enzyme Cyp26b1 is highly expressed in skin fibroblasts, and its inhibition resulted in the upregulation of P2X7 on MCs. We also noted the increased expression of P2X7 on skin MCs and consequent P2X7- and MC-dependent dermatitis (so-called retinoid dermatitis) in the presence of excessive amounts of retinoic acid. These results demonstrate a unique skin-barrier homeostatic network operating through Cyp26b1-mediated inhibition of ATP-dependent MC activation by fibroblasts.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Dermatitis/immunology , Fibroblasts/immunology , Mast Cells/immunology , Receptors, Purinergic P2X7/metabolism , Skin/metabolism , Adenosine Triphosphate/immunology , Animals , Cell Degranulation/drug effects , Cell Degranulation/genetics , Cytochrome P-450 Enzyme System/genetics , Imidazoles/administration & dosage , Immunity, Innate/drug effects , Immunity, Innate/genetics , Mast Cells/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microbiota/immunology , Receptors, Purinergic P2X7/genetics , Retinoic Acid 4-Hydroxylase , Skin/immunology , Skin/microbiology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Tretinoin/immunology
14.
Clin Exp Allergy ; 52(1): 149-161, 2022 01.
Article in English | MEDLINE | ID: mdl-34418187

ABSTRACT

BACKGROUND: Myeloid differentiation protein-2 (MD-2) is a lipopolysaccharide-binding protein involved in lipopolysaccharide signalling via Toll-like receptor 4 (TLR4). TLR4 plays an essential role in HDM-mediated allergic airway inflammation. Moreover, MD-2 is structurally similar to Der f 2, a major allergen from house dust mite (HDM). OBJECTIVES: We aimed to clarify the role of MD-2 in the pathogenesis of HDM-mediated allergic airway inflammation. METHODS: Wild-type (WT), TLR4 knockout and MD-2 knockout mice were subjected to intranasal instillation of HDM extract, and asthmatic features were evaluated. We also evaluated gene sets regulated by MD-2 in HDM-treated airway epithelial cells and examined the function of dendritic cells from lymph nodes and from lungs. RESULTS: Aggravated allergic airway inflammation with increased airway hyperresponsiveness was observed in MD-2 knockout mice compared with WT and TLR4 knockout mice. Global gene expression analysis revealed an MD-2 regulated proinflammatory response and reconstituted TLR4 signalling in airway epithelial cells. The ability of dendritic cells to evoke an allergic immune response was enhanced in MD-2 knockout mice. CONCLUSIONS & CLINICAL RELEVANCE: MD-2 plays a protective role in HDM-induced airway allergy with the proinflammatory regulation of airway epithelial cells and dendritic cells. MD-2 may serve as a therapeutic target in the treatment of asthma.


Subject(s)
Asthma , Pyroglyphidae , Animals , Asthma/genetics , Dendritic Cells , Disease Models, Animal , Epithelial Cells/metabolism , Humans , Lung , Mice , Mice, Knockout
15.
Int Immunol ; 33(12): 809-813, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34038539

ABSTRACT

Basophils, the rarest granulocytes, were identified by Paul Ehrlich more than 140 years ago, much earlier than the discovery of T and B cells. Unfortunately, basophils were often mixed up with tissue-resident mast cells because of some phenotypic similarities between them and considered erroneously as minor relatives or blood-circulating precursors of mast cells. Moreover, basophil research was hindered by the rarity of basophils and the paucity of useful analytical tools, and therefore basophils had often been neglected in immunological studies. A series of studies using newly developed tools, including basophil-depleting antibodies and genetically engineered mice deficient only in basophils, have clearly defined previously unrecognized roles of basophils, that are distinct from those played by tissue-resident mast cells. In this mini-review, we highlight recent advances in our understanding of basophil functions, particularly focusing on their roles in the regulation of innate and acquired immunity, allergic reactions, autoimmunity and protective immunity against parasitic infections, mainly based on animal studies. Further studies on human basophils would facilitate the development of new strategies for the treatment of basophil-associated disorders.


Subject(s)
Basophils/immunology , Animals , Humans
16.
Int Immunol ; 33(12): 835-840, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34223897

ABSTRACT

Nucleic-acid (NA)-sensing Toll-like receptors (TLRs) are synthesized in the endoplasmic reticulum and mature with chaperones, such as Unc93B1 and the protein associated with TLR4 A (PRAT4A)-gp96 complex. The TLR-Unc93B1 complexes move to the endosomal compartment, where proteases such as cathepsins activate their responsiveness through proteolytic cleavage of the extracellular domain of TLRs. Without proteolytic cleavage, ligand-dependent dimerization of NA-sensing TLRs is prevented by the uncleaved loop in the extracellular domains. Additionally, the association of Unc93B1 inhibits ligand-dependent dimerization of TLR3 and TLR9 and, therefore, Unc93B1 is released from these TLRs before dimerization. Ligand-activated NA-sensing TLRs induce the production of pro-inflammatory cytokines and act on the endosomal compartment to initiate anterograde trafficking to the cell periphery for type I interferon production. In the endosomal compartment, DNA and RNA are degraded by DNases and RNases, respectively, generating degradation products. DNase 2A and RNase T2 generate ligands for TLR9 and TLR8, respectively. In this mechanism, DNases and RNases control innate immune responses to NAs in endosomal compartments. NA-sensing TLRs and the endosomal compartment work together to monitor environmental cues through endosomes and decide to launch innate immune responses.


Subject(s)
Endosomes/immunology , Nucleic Acids/immunology , Toll-Like Receptors/immunology , Animals , Humans
17.
Int Immunol ; 33(9): 479-490, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34161582

ABSTRACT

RNase T2, a ubiquitously expressed RNase, degrades RNAs in the endosomal compartments. RNA sensors, double-stranded RNA (dsRNA)-sensing Toll-like receptor 3 (TLR3) and single-stranded RNA (ssRNA)-sensing TLR7, are localized in the endosomal compartment in mouse macrophages. We here studied the role of RNase T2 in TLR3 and TLR7 responses in macrophages. Macrophages expressed RNase T2 and a member of the RNase A family RNase 4. RNase T2 was also expressed in plasmacytoid and conventional dendritic cells. Treatment with dsRNAs or type I interferon (IFN) up-regulated expression of RNase T2 but not RNase 4. RNase T2-deficiency in macrophages up-regulated TLR3 responses but impaired TLR7 responses. Mechanistically, RNase T2 degraded both dsRNAs and ssRNAs in vitro, and its mutants showed a positive correlation between RNA degradation and the rescue of altered TLR3 and TLR7 responses. H122A and C188R RNase T2 mutations, not H69A and E118V mutations, impaired both RNA degradation and the rescue of altered TLR3 and TLR7 responses. RNase T2 in bone marrow-derived macrophages was broadly distributed from early endosomes to lysosomes, and colocalized with the internalized TLR3 ligand poly(I:C). These results suggest that RNase T2-dependent RNA degradation in endosomes/lysosomes negatively and positively regulates TLR3 and TLR7 responses, respectively, in macrophages.


Subject(s)
Endoribonucleases/metabolism , Endosomes/metabolism , Macrophages/metabolism , Membrane Glycoproteins/metabolism , RNA, Double-Stranded/metabolism , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 7/metabolism , Animals , Cell Line , Cytokines/metabolism , Dendritic Cells/metabolism , HEK293 Cells , Humans , Lysosomes/metabolism , Mice , Mice, Inbred C57BL
18.
Immunity ; 38(6): 1187-97, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23791646

ABSTRACT

The small intestine harbors a substantial number of commensal bacteria and is sporadically invaded by pathogens, but the response to these microorganisms is fundamentally different. We identified a discriminatory sensor by using Toll-like receptor 3 (TLR3). Double-stranded RNA (dsRNA) of one major commensal species, lactic acid bacteria (LAB), triggered interferon-ß (IFN-ß) production, which protected mice from experimental colitis. The LAB-induced IFN-ß response was diminished by dsRNA digestion and treatment with endosomal inhibitors. Pathogenic bacteria contained less dsRNA and induced much less IFN-ß than LAB, and dsRNA was not involved in pathogen-induced IFN-ß induction. These results identify TLR3 as a sensor to small intestinal commensal bacteria and suggest that dsRNA in commensal bacteria contributes to anti-inflammatory and protective immune responses.


Subject(s)
Colitis/prevention & control , Enterococcaceae/immunology , Gram-Positive Bacterial Infections/immunology , Interferon-beta/metabolism , Lactobacillus/immunology , Macrophages/immunology , Toll-Like Receptor 3/metabolism , Animals , Cells, Cultured , Colitis/etiology , Colitis/immunology , Colitis/microbiology , Disease Models, Animal , Enterococcaceae/pathogenicity , Female , Gram-Positive Bacterial Infections/complications , Gram-Positive Bacterial Infections/microbiology , Intestines/immunology , Intestines/microbiology , Macrophages/microbiology , Membrane Transport Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , RNA, Double-Stranded/immunology
19.
Am J Hum Genet ; 102(2): 321-329, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29394991

ABSTRACT

Early-onset epileptic encephalopathies, including West syndrome (WS), are a group of neurological disorders characterized by developmental impairments and intractable seizures from early infancy. We have now identified biallelic CNPY3 variants in three individuals with WS; these include compound-heterozygous missense and frameshift variants in a family with two affected siblings (individuals 1 and 2) and a homozygous splicing variant in a consanguineous family (individual 3). All three individuals showed hippocampal malrotation. In individuals 1 and 2, electroencephalography (EEG) revealed characteristic fast waves and diffuse sharp- and slow-wave complexes. The fast waves were clinically associated with seizures. CNPY3 encodes a co-chaperone in the endoplasmic reticulum and regulates the subcellular distribution and responses of multiple Toll-like receptors. The amount of CNPY3 in lymphoblastoid cells derived from individuals 1 and 2 was severely lower than that in control cells. Cnpy3-knockout mice exhibited spastic or dystonic features under resting conditions and hyperactivity and anxiolytic behavior during the open field test. Also, their resting EEG showed enhanced activity in the fast beta frequency band (20-35 Hz), which could mimic the fast waves in individuals 1 and 2. These data suggest that CNPY3 and Cnpy3 perform essential roles in brain function in addition to known Toll-like receptor-dependent immune responses.


Subject(s)
Molecular Chaperones/genetics , Mutation , Seizures/genetics , Spasms, Infantile/genetics , Adolescent , Amino Acid Sequence , Animals , Child , Consanguinity , Electroencephalography , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Family , Female , Gene Expression , Heterozygote , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Infant , Magnetic Resonance Imaging , Male , Mice , Mice, Knockout , Seizures/diagnostic imaging , Seizures/physiopathology , Sequence Alignment , Sequence Homology, Amino Acid , Siblings , Spasms, Infantile/diagnostic imaging , Spasms, Infantile/physiopathology
20.
Allergy ; 76(6): 1693-1706, 2021 06.
Article in English | MEDLINE | ID: mdl-33205439

ABSTRACT

Basophils are the rarest granulocytes which represent <1% of peripheral blood leukocytes. Basophils bear several phenotypic similarities to tissue-resident mast cells and therefore had been erroneously considered as blood-circulating mast cells. However, recent researches have revealed that basophils play nonredundant roles in allergic inflammation, protective immunity against parasitic infections and regulation of innate and acquired immunity. Basophils are recruited to inflamed tissues and activated in an IgE-dependent or IgE-independent manner to release a variety of effector molecules. Such molecules, including IL-4, act on various types of cells and play versatile roles, including the induction and termination of allergic inflammation and the regulation of immune responses. Recent development of novel therapeutic agents has enabled us to gain further insights into basophil biology in human disorders. In this review, we highlight the recent advances in the field of basophil biology with a particular focus on the role of basophils in allergic inflammation. Further studies on basophils and their effector molecules will help us identify novel therapeutic targets for treating allergic disorders.


Subject(s)
Hypersensitivity , Parasitic Diseases , Basophils , Humans , Inflammation , Mast Cells
SELECTION OF CITATIONS
SEARCH DETAIL