Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.009
Filter
Add more filters

Publication year range
1.
Mol Cell ; 67(3): 387-399.e5, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28712728

ABSTRACT

The DNA-mediated innate immune response underpins anti-microbial defenses and certain autoimmune diseases. Here we used immunoprecipitation, mass spectrometry, and RNA sequencing to identify a ribonuclear complex built around HEXIM1 and the long non-coding RNA NEAT1 that we dubbed the HEXIM1-DNA-PK-paraspeckle components-ribonucleoprotein complex (HDP-RNP). The HDP-RNP contains DNA-PK subunits (DNAPKc, Ku70, and Ku80) and paraspeckle proteins (SFPQ, NONO, PSPC1, RBM14, and MATRIN3). We show that binding of HEXIM1 to NEAT1 is required for its assembly. We further demonstrate that the HDP-RNP is required for the innate immune response to foreign DNA, through the cGAS-STING-IRF3 pathway. The HDP-RNP interacts with cGAS and its partner PQBP1, and their interaction is remodeled by foreign DNA. Remodeling leads to the release of paraspeckle proteins, recruitment of STING, and activation of DNAPKc and IRF3. Our study establishes the HDP-RNP as a key nuclear regulator of DNA-mediated activation of innate immune response through the cGAS-STING pathway.


Subject(s)
DNA/immunology , Herpesvirus 8, Human/immunology , Immunity, Innate , RNA, Long Noncoding/immunology , RNA-Binding Proteins/immunology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/immunology , Calcium-Binding Proteins/metabolism , DNA/genetics , DNA/metabolism , DNA-Binding Proteins , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/virology , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Ku Autoantigen/genetics , Ku Autoantigen/immunology , Ku Autoantigen/metabolism , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Multiprotein Complexes , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/immunology , Nuclear Matrix-Associated Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/immunology , Nucleotidyltransferases/metabolism , Octamer Transcription Factors/genetics , Octamer Transcription Factors/immunology , Octamer Transcription Factors/metabolism , PTB-Associated Splicing Factor/genetics , PTB-Associated Splicing Factor/immunology , PTB-Associated Splicing Factor/metabolism , Protein Binding , RNA Interference , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction , Transcription Factors , Transfection
2.
PLoS Pathog ; 18(1): e1009718, 2022 01.
Article in English | MEDLINE | ID: mdl-35073381

ABSTRACT

Salmonella enterica serovar Typhimurium is a Gram-negative pathogen that uses two distinct type III secretion systems (T3SSs), termed Salmonella pathogenicity island (SPI)-1 and SPI-2, to deliver virulence factors into the host cell. The SPI-1 T3SS enables Salmonella to invade host cells, while the SPI-2 T3SS facilitates Salmonella's intracellular survival. In mice, a family of cytosolic immune sensors, including NAIP1, NAIP2, and NAIP5/6, recognizes the SPI-1 T3SS needle, inner rod, and flagellin proteins, respectively. Ligand recognition triggers assembly of the NAIP/NLRC4 inflammasome, which mediates caspase-1 activation, IL-1 family cytokine secretion, and pyroptosis of infected cells. In contrast to mice, humans encode a single NAIP that broadly recognizes all three ligands. The role of NAIP/NLRC4 or other inflammasomes during Salmonella infection of human macrophages is unclear. We find that although the NAIP/NLRC4 inflammasome is essential for detecting T3SS ligands in human macrophages, it is partially required for responses to infection, as Salmonella also activated the NLRP3 and CASP4/5 inflammasomes. Importantly, we demonstrate that combinatorial NAIP/NLRC4 and NLRP3 inflammasome activation restricts Salmonella replication in human macrophages. In contrast to SPI-1, the SPI-2 T3SS inner rod is not sensed by human or murine NAIPs, which is thought to allow Salmonella to evade host recognition and replicate intracellularly. Intriguingly, we find that human NAIP detects the SPI-2 T3SS needle protein. Critically, in the absence of both flagellin and the SPI-1 T3SS, the NAIP/NLRC4 inflammasome still controlled intracellular Salmonella burden. These findings reveal that recognition of Salmonella SPI-1 and SPI-2 T3SSs and engagement of both the NAIP/NLRC4 and NLRP3 inflammasomes control Salmonella infection in human macrophages.


Subject(s)
Inflammasomes/immunology , Macrophages/immunology , Macrophages/microbiology , Salmonella Infections/immunology , Type III Secretion Systems/immunology , CARD Signaling Adaptor Proteins/immunology , Calcium-Binding Proteins/immunology , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Neuronal Apoptosis-Inhibitory Protein/immunology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Virulence
3.
Nat Immunol ; 13(2): 162-9, 2012 Jan 08.
Article in English | MEDLINE | ID: mdl-22231517

ABSTRACT

Memory T cells exert antigen-independent effector functions, but how these responses are regulated is unclear. We discovered an in vivo link between flagellin-induced NLRC4 inflammasome activation in splenic dendritic cells (DCs) and host protective interferon-γ (IFN-γ) secretion by noncognate memory CD8(+) T cells, which could be activated by Salmonella enterica serovar Typhimurium, Yersinia pseudotuberculosis and Pseudomonas aeruginosa. We show that CD8α(+) DCs were particularly efficient at sensing bacterial flagellin through NLRC4 inflammasomes. Although this activation released interleukin 18 (IL-18) and IL-1ß, only IL-18 was required for IFN-γ production by memory CD8(+) T cells. Conversely, only the release of IL-1ß, but not IL-18, depended on priming signals mediated by Toll-like receptors. These findings provide a comprehensive mechanistic framework for the regulation of noncognate memory T cell responses during bacterial immunity.


Subject(s)
Apoptosis Regulatory Proteins/immunology , CD8-Positive T-Lymphocytes/immunology , Calcium-Binding Proteins/immunology , Dendritic Cells/immunology , Immunologic Memory , Inflammasomes/immunology , Interferon-gamma/immunology , Animals , Flagellin/immunology , Interleukin-18/immunology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Mice , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Salmonella Infections, Animal/immunology , Salmonella typhimurium/immunology , Signal Transduction/immunology , Spleen/immunology , Toll-Like Receptors/immunology , Yersinia pseudotuberculosis Infections/immunology
4.
Nat Immunol ; 13(5): 449-56, 2012 May.
Article in English | MEDLINE | ID: mdl-22484733

ABSTRACT

Intestinal phagocytes transport oral antigens and promote immune tolerance, but their role in innate immune responses remains unclear. Here we found that intestinal phagocytes were anergic to ligands for Toll-like receptors (TLRs) or commensals but constitutively expressed the precursor to interleukin 1ß (pro-IL-1ß). After infection with pathogenic Salmonella or Pseudomonas, intestinal phagocytes produced mature IL-1ß through the NLRC4 inflammasome but did not produce tumor necrosis factor (TNF) or IL-6. BALB/c mice deficient in NLRC4 or the IL-1 receptor were highly susceptible to orogastric but not intraperitoneal infection with Salmonella. That enhanced lethality was preceded by impaired expression of endothelial adhesion molecules, lower neutrophil recruitment and poor intestinal pathogen clearance. Thus, NLRC4-dependent production of IL-1ß by intestinal phagocytes represents a specific response that discriminates pathogenic bacteria from commensal bacteria and contributes to host defense in the intestine.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/immunology , Clonal Anergy , Host-Pathogen Interactions/immunology , Interleukin-1beta/metabolism , Intestines/immunology , Intestines/microbiology , Phagocytes/immunology , Animals , Apoptosis Regulatory Proteins/genetics , Calcium-Binding Proteins/genetics , Caspase 1/metabolism , Flagellin/immunology , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-6/biosynthesis , Interleukin-6/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/pathology , Phagocytes/microbiology , Pseudomonas/immunology , Pseudomonas Infections/immunology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Salmonella/genetics , Salmonella/immunology , Salmonella Infections/genetics , Salmonella Infections/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
5.
Immunity ; 43(5): 987-97, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26572063

ABSTRACT

Defective neutrophils in patients with chronic granulomatous disease (CGD) cause susceptibility to extracellular and intracellular infections. Microbes must first be ejected from intracellular niches to expose them to neutrophil attack, so we hypothesized that inflammasomes detect certain CGD pathogens upstream of neutrophil killing. Here, we identified one such ubiquitous environmental bacterium, Chromobacterium violaceum, whose extreme virulence was fully counteracted by the NLRC4 inflammasome. Caspase-1 protected via two parallel pathways that eliminated intracellular replication niches. Pyroptosis was the primary bacterial clearance mechanism in the spleen, but both pyroptosis and interleukin-18 (IL-18)-driven natural killer (NK) cell responses were required for liver defense. NK cells cleared hepatocyte replication niches via perforin-dependent cytotoxicity, whereas interferon-γ was not required. These insights suggested a therapeutic approach: exogenous IL-18 restored perforin-dependent cytotoxicity during infection by the inflammasome-evasive bacterium Listeria monocytogenes. Therefore, inflammasomes can trigger complementary programmed cell death mechanisms, directing sterilizing immunity against intracellular bacterial pathogens.


Subject(s)
Bacterial Infections/immunology , Inflammasomes/immunology , Killer Cells, Natural/immunology , Pyroptosis/immunology , Animals , Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/immunology , Caspase 1/immunology , Cell Death/immunology , Chromobacterium/immunology , Granulomatous Disease, Chronic/immunology , Interferon-gamma/immunology , Interleukin-18/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Liver/immunology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Spleen/immunology
6.
Clin Immunol ; 234: 108894, 2022 01.
Article in English | MEDLINE | ID: mdl-34843986

ABSTRACT

Cupressaceae pollen causes allergic reactions worldwide with long-lasting symptomatic periods. Currently, no cypress polcalcin is available for diagnostic purposes. With the aim to investigate the pattern of sensitization to a cypress polcalcin, a synthetic gene of Jun o 4, the Juniperus oxycedrus 4EF-hand polcalcin, was cloned and expressed in Escherichia coli. Its features were investigated in comparison with the grass 2EF-hand Phl p 7. Rhinitis was the symptom most frequently reported in a cohort of Italian patients sensitized to rJun o 4 and/or rPhl p 7. The detection of many pollen allergic patients sensitized to the cypress polcalcin, but negative to Phl p 7, indicates that Phl p 7 cannot be further considered a marker of sensitization towards all the polcalcins. A 4EF-hand cypress polcalcin claims the inclusion in allergy diagnostic tests. In addition, the sensitivity of polcalcins to gastrointestinal digestion is reported and discussed for the first time.


Subject(s)
Allergens/immunology , Antigens, Plant/immunology , Calcium-Binding Proteins/immunology , Juniperus/immunology , Rhinitis, Allergic, Seasonal/etiology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Cloning, Molecular , Female , Humans , Infant , Male , Middle Aged , Proteolysis , Rhinitis, Allergic, Seasonal/immunology , Young Adult
7.
Nat Immunol ; 11(12): 1136-42, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21057511

ABSTRACT

Macrophages mediate crucial innate immune responses via caspase-1-dependent processing and secretion of interleukin 1ß (IL-1ß) and IL-18. Although infection with wild-type Salmonella typhimurium is lethal to mice, we show here that a strain that persistently expresses flagellin was cleared by the cytosolic flagellin-detection pathway through the activation of caspase-1 by the NLRC4 inflammasome; however, this clearance was independent of IL-1ß and IL-18. Instead, caspase-1-induced pyroptotic cell death released bacteria from macrophages and exposed the bacteria to uptake and killing by reactive oxygen species in neutrophils. Similarly, activation of caspase-1 cleared unmanipulated Legionella pneumophila and Burkholderia thailandensis by cytokine-independent mechanisms. This demonstrates that activation of caspase-1 clears intracellular bacteria in vivo independently of IL-1ß and IL-18 and establishes pyroptosis as an efficient mechanism of bacterial clearance by the innate immune system.


Subject(s)
Apoptosis/immunology , Caspase 1/immunology , Immunity, Innate/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Animals , Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/immunology , Calcium-Binding Proteins/metabolism , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunohistochemistry , Inflammasomes/immunology , Inflammasomes/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL
8.
FASEB J ; 35(1): e21213, 2021 01.
Article in English | MEDLINE | ID: mdl-33368614

ABSTRACT

Preclinical studies have demonstrated that activation of the NOTCH pathway plays a key role in the pathogenesis of kidney damage. There is currently no information on the role of the Delta-like homologue 1 (DLK1), a NOTCH inhibitor, in the regulation of renal damage. Here, we investigated the contribution of DLK1 to experimental renal damage and the underlying molecular mechanisms. Using a Dlk1-null mouse model in the experimental renal damage of unilateral ureteral obstruction, we found activation of NOTCH, as shown by increased nuclear translocation of the NOTCH1 intracellular domain, and upregulation of Dlk2/hey-1 expression compared to wild-type (WT) littermates. NOTCH1 over-activation in Dlk1-null injured kidneys was associated with a higher inflammatory response, characterized by infiltration of inflammatory cells, mainly CD4/IL17A + lymphocytes, and activation of the Th17 immune response. Furthermore, pharmacological NOTCH blockade inhibited the transcription factors controlling Th17 differentiation and gene expression of the Th17 effector cytokine IL-17A and other related-inflammatory factors, linked to a diminution of inflammation in the injured kidneys. We propose that the non-canonical NOTCH ligand DLK1 acts as a NOTCH antagonist in renal injury regulating the Th17-mediated inflammatory response.


Subject(s)
Calcium-Binding Proteins/deficiency , Gene Deletion , Immunity, Cellular , Kidney Diseases/immunology , Kidney/immunology , Th17 Cells/immunology , Animals , Calcium-Binding Proteins/immunology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Mice , Th17 Cells/pathology , Ureteral Obstruction/genetics , Ureteral Obstruction/immunology , Ureteral Obstruction/pathology
9.
Immunity ; 39(3): 432-41, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-24054327

ABSTRACT

A major function of a subfamily of NLR (nucleotide-binding domain, leucine-rich repeat containing, or NOD-like receptor) proteins is in inflammasome activation, which has been implicated in a multitude of disease models and human diseases. This work will highlight key progress in understanding the mechanisms that activate the best-studied NLRs (NLRP3, NLRC4, NAIP, and NLRP1) and in uncovering inflammasome NLRs.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/metabolism , Carrier Proteins/metabolism , Inflammasomes/immunology , Neuronal Apoptosis-Inhibitory Protein/metabolism , Adaptor Proteins, Signal Transducing/immunology , Animals , Apoptosis Regulatory Proteins/immunology , CARD Signaling Adaptor Proteins/immunology , Calcium-Binding Proteins/immunology , Carrier Proteins/immunology , Caspase 1/metabolism , Enzyme Activation , Humans , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , NLR Proteins , Neuronal Apoptosis-Inhibitory Protein/immunology , Receptors, Cytoplasmic and Nuclear/immunology , Signal Transduction
10.
J Immunol ; 205(3): 811-821, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32591398

ABSTRACT

Some strains of lactic acid bacteria (LAB) have anti-inflammatory effects, but the mechanism underlying the alleviation of inflammation by LAB is not fully understood. In this study, we examined the inhibitory effect of a certain strain of LAB, Lactobacillus paracasei, on inflammasome activation, which is associated with various inflammatory disorders. Using bone marrow-derived macrophages from BALB/c mice, we found that L. paracasei, but not L. rhamnosus, suppressed NLRP3 inflammasome activation and inhibited subsequent caspase-1 activation and IL-1ß secretion. L. paracasei also had inhibitory effects on AIM2 and NLRC4 inflammasome activation as well as the NLRP3 inflammasome. These inhibitory effects of L. paracasei on inflammasome activation were dependent on autocrine IL-10 induced by L. paracasei-stimulated macrophages. Furthermore, IL-10 production by L. paracasei-stimulated macrophages was involved with phagocytosis and the NOD2 signaling pathway in macrophages. In addition to in vitro studies, oral administration of L. paracasei in C57BL/6 mice reduced monosodium urate crystal-induced peritoneal inflammation in vivo. Moreover, continuous intake of L. paracasei in C57BL/6 mice alleviated high fat diet-induced insulin resistance and aging-induced expression of biomarkers for T cell senescence. Taken together, we demonstrated that L. paracasei inhibits inflammasome activation in vitro and exhibits an anti-inflammatory function in vivo. These results indicate that LAB that have inhibitory effects on inflammasome activation might contribute to the alleviation of inflammation-related disorders.


Subject(s)
Inflammasomes/immunology , Lacticaseibacillus paracasei/immunology , Macrophages/immunology , Signal Transduction/immunology , Animals , Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/immunology , Caspase 1/immunology , DNA-Binding Proteins/immunology , Inflammation/chemically induced , Inflammation/immunology , Inflammation/prevention & control , Interleukin-10/immunology , Mice , Mice, Inbred BALB C , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Nod2 Signaling Adaptor Protein/immunology
11.
J Immunol ; 204(5): 1214-1224, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31980574

ABSTRACT

Leukocytes are rapidly recruited to sites of inflammation via interactions with the vascular endothelium. The steroid hormone dehydroepiandrosterone (DHEA) exerts anti-inflammatory properties; however, the underlying mechanisms are poorly understood. In this study, we show that an anti-inflammatory mechanism of DHEA involves the regulation of developmental endothelial locus 1 (DEL-1) expression. DEL-1 is a secreted homeostatic factor that inhibits ß2-integrin-dependent leukocyte adhesion, and the subsequent leukocyte recruitment and its expression is downregulated upon inflammation. Similarly, DHEA inhibited leukocyte adhesion to the endothelium in venules of the inflamed mouse cremaster muscle. Importantly, in a model of lung inflammation, DHEA limited neutrophil recruitment in a DEL-1-dependent manner. Mechanistically, DHEA counteracted the inhibitory effect of inflammation on DEL-1 expression. Indeed, whereas TNF reduced DEL-1 expression and secretion in endothelial cells by diminishing C/EBPß binding to the DEL-1 gene promoter, DHEA counteracted the inhibitory effect of TNF via activation of tropomyosin receptor kinase A (TRKA) and downstream PI3K/AKT signaling that restored C/EBPß binding to the DEL-1 promoter. In conclusion, DHEA restrains neutrophil recruitment by reversing inflammation-induced downregulation of DEL-1 expression. Therefore, the anti-inflammatory DHEA/DEL-1 axis could be harnessed therapeutically in the context of inflammatory diseases.


Subject(s)
Calcium-Binding Proteins/immunology , Cell Adhesion Molecules/immunology , Dehydroepiandrosterone/pharmacology , Leukocytes/immunology , Signal Transduction/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/immunology , CD18 Antigens/immunology , Cell Adhesion/immunology , Endothelium, Vascular/immunology , Female , Gene Expression Regulation/immunology , Leukocytes/cytology , Mice , Phosphatidylinositol 3-Kinases/immunology , Promoter Regions, Genetic/immunology , Proto-Oncogene Proteins c-akt/immunology , Receptor, trkA/immunology
12.
J Am Soc Nephrol ; 32(2): 268-278, 2021 02.
Article in English | MEDLINE | ID: mdl-33380523

ABSTRACT

Membranous nephropathy (MN) occurs due to deposition of immune complexes along the subepithelial region of glomerular basement membrane. Two previously identified target antigens for the immune complexes, PLA2R (identified in 2009) and THSD7A (in 2014), account for approximately 60% of all MN, both primary and secondary. In the remaining MN, target antigens were unknown. Use of laser microdissection and mass spectrometry enabled identification of new "antigens." This approach led to the identification of four novel types of MN: exotosin 1 (EXT1)- and exotosin 2 (EXT2)-associated MN, NELL1-associated MN, Sema3B-associated MN, and PCDH7-associated MN. Each of these represents a distinct disease entity, with different clinical and pathologic findings. In this review, the structure of the proteins and the clinical and pathologic findings of the new types of MN are discussed. The role of mass spectrometry for accurate diagnosis of MN cannot be overemphasized. Finally, any classification of MN should be made on the basis of the antigens that are detected. Further studies are required to understand the pathophysiology, response to treatment, and outcomes of these new MNs.


Subject(s)
Glomerulonephritis, Membranous/diagnosis , Cadherins/immunology , Calcium-Binding Proteins/immunology , Glomerulonephritis, Membranous/etiology , Humans , Laser Capture Microdissection , Membrane Glycoproteins/immunology , N-Acetylglucosaminyltransferases/immunology , Protocadherins , Semaphorins/immunology , Tandem Mass Spectrometry
13.
J Allergy Clin Immunol ; 147(4): 1442-1452, 2021 04.
Article in English | MEDLINE | ID: mdl-32791164

ABSTRACT

BACKGROUND: The immunoinhibitory receptor Siglec-8 on the surface of human eosinophils and mast cells binds to sialic acid-containing ligands in the local milieu, resulting in eosinophil apoptosis, inhibition of mast cell degranulation, and suppression of inflammation. Siglec-8 ligands were found on postmortem human trachea and bronchi and on upper airways in 2 compartments, cartilage and submucosal glands, but they were surprisingly absent from the epithelium. We hypothesized that Siglec-8 ligands in submucosal glands and ducts are normally transported to the airway mucus layer, which is lost during tissue preparation. OBJECTIVE: Our aim was to identify the major Siglec-8 sialoglycan ligand on the mucus layer of human airways. METHODS: Human upper airway mucus layer proteins were recovered during presurgical nasal lavage of patients at a sinus clinic. Proteins were resolved by gel electrophoresis and blotted, and Siglec-8 ligands detected. Ligands were purified by size exclusion and affinity chromatography, identified by proteomic mass spectrometry, and validated by electrophoretic and histochemical colocalization. The affinity of Siglec-8 binding to purified human airway ligand was determined by inhibition of glycan binding. RESULTS: A Siglec-8-ligand with a molecular weight of approximately 1000 kDa was found in all patient nasal lavage samples. Purification and identification revealed deleted in malignant brain tumors 1 (DMBT1) (also known by the aliases GP340 and SALSA), a large glycoprotein with multiple O-glycosylation repeats. Immunoblotting, immunohistochemistry, and enzyme treatments confirmed that Siglec-8 ligand on the human airway mucus layer is an isoform of DMBT1 carrying O-linked sialylated keratan sulfate chains (DMBT1S8). Quantitative inhibition revealed that DMBT1S8 has picomolar affinity for Siglec-8. CONCLUSION: A distinct DMBT1 isoform, DMBT1S8, is the major high-avidity ligand for Siglec-8 on human airways.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Calcium-Binding Proteins/immunology , DNA-Binding Proteins/immunology , Lectins/immunology , Tumor Suppressor Proteins/immunology , Bronchi/immunology , Calcium-Binding Proteins/chemistry , DNA-Binding Proteins/chemistry , Eosinophils/immunology , Humans , Ligands , Mast Cells/immunology , Nasal Lavage Fluid/immunology , Proteoglycans/immunology , Trachea/immunology , Tumor Suppressor Proteins/chemistry
14.
Am J Physiol Lung Cell Mol Physiol ; 321(3): L533-L544, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34231388

ABSTRACT

Store-operated calcium entry (SOCE) is involved in the pathogenesis of airway inflammation and remodeling in asthma. Store-operated calcium entry-associated regulatory factor (SARAF) can downregulate SOCE. We sought to investigate the role of SARAF in the regulation of airway inflammation and remodeling in asthma mice models, as well as in the functional regulation of human airway smooth muscle cells (hASMCs). Balb/c mice were sensitized and challenged with ovalbumin to establish the asthma mice models. Mice were transfected with lentivirus, which expressed the SARAF gene + GFP (green fluorescence protein) or the negative control gene + GFP. Airway resistance was measured with the animal pulmonary function system. Airway inflammation and remodeling were evaluated via histological staining. In vitro cultured hASMCs were transfected with scrambled small interfering RNA (siRNA) or SARAF-specific siRNA, respectively. The proliferation, migration rate, hypertrophy, and SOCE activity of hASMCs were examined with Cell Counting Kit-8, wound healing test, bright field imaging, and Ca2+ fluorescence imaging, respectively. SARAF expression was measured by quantitative real-time PCR. Asthma mice models showed decreased SARAF mRNA expression in the lungs. SARAF overexpression attenuated airway inflammation, resistance, and also remodeling. Downregulation of SARAF expression with siRNA promoted the proliferation, migration, hypertrophy, and SOCE activity in hASMCs. SARAF plays a protective role against airway inflammation and remodeling in asthma mice models by blunting SOCE; SARAF may also be a functional regulating factor of hASMCs.


Subject(s)
Airway Remodeling/immunology , Asthma/immunology , Calcium-Binding Proteins/immunology , Gene Expression Regulation/immunology , Lung/immunology , Membrane Proteins/immunology , Myocytes, Smooth Muscle/immunology , Airway Remodeling/drug effects , Airway Remodeling/genetics , Airway Resistance/drug effects , Airway Resistance/genetics , Airway Resistance/immunology , Animals , Asthma/chemically induced , Asthma/genetics , Calcium-Binding Proteins/genetics , Female , Gene Expression Regulation/drug effects , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Lung/pathology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Myocytes, Smooth Muscle/pathology
15.
PLoS Pathog ; 15(6): e1007886, 2019 06.
Article in English | MEDLINE | ID: mdl-31251782

ABSTRACT

Inflammasomes are cytosolic multi-protein complexes that detect infection or cellular damage and activate the Caspase-1 (CASP1) protease. The NAIP5/NLRC4 inflammasome detects bacterial flagellin and is essential for resistance to the flagellated intracellular bacterium Legionella pneumophila. The effectors required downstream of NAIP5/NLRC4 to restrict bacterial replication remain unclear. Upon NAIP5/NLRC4 activation, CASP1 cleaves and activates the pore-forming protein Gasdermin-D (GSDMD) and the effector caspase-7 (CASP7). However, Casp1-/- (and Casp1/11-/-) mice are only partially susceptible to L. pneumophila and do not phenocopy Nlrc4-/-mice, because NAIP5/NLRC4 also activates CASP8 for restriction of L. pneumophila infection. Here we show that CASP8 promotes the activation of CASP7 and that Casp7/1/11-/- and Casp8/1/11-/- mice recapitulate the full susceptibility of Nlrc4-/- mice. Gsdmd-/- mice exhibit only mild susceptibility to L. pneumophila, but Gsdmd-/-Casp7-/- mice are as susceptible as the Nlrc4-/- mice. These results demonstrate that GSDMD and CASP7 are the key substrates downstream of NAIP5/NLRC4/CASP1/8 required for resistance to L. pneumophila.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/immunology , Caspase 1/immunology , Caspase 7/immunology , Caspase 8/immunology , Inflammasomes/immunology , Legionella pneumophila/immunology , Legionnaires' Disease/immunology , Neuronal Apoptosis-Inhibitory Protein/immunology , Animals , Apoptosis Regulatory Proteins/genetics , Calcium-Binding Proteins/genetics , Caspase 1/genetics , Caspase 7/genetics , Caspase 8/genetics , Inflammasomes/genetics , Intracellular Signaling Peptides and Proteins , Legionnaires' Disease/genetics , Legionnaires' Disease/pathology , Mice , Mice, Knockout , Neuronal Apoptosis-Inhibitory Protein/genetics , Phosphate-Binding Proteins
16.
PLoS Pathog ; 15(6): e1007795, 2019 06.
Article in English | MEDLINE | ID: mdl-31170267

ABSTRACT

Infection with the Streptococcus suis (S. suis) epidemic strain can cause Streptococcal toxic shock-like syndrome (STSLS), which is characterized by a cytokine storm, dysfunction of multiple organs and a high incidence of mortality despite adequate treatment. Despite some progress concerning the contribution of the inflammatory response to STSLS, the precise mechanism underlying STSLS development remains elusive. Here, we use a murine model to demonstrate that caspase-1 activity is critical for STSLS development. Furthermore, we show that inflammasome activation by S. suis is mainly dependent on NLRP3 but not on NLRP1, AIM2 or NLRC4. The important role of NLRP3 activation in STSLS is further confirmed in vivo with the NLRP3 inhibitor MCC950 and nlrp3-knockout mice. By comparison of WT strain with isogenic strains with mutation of various virulence genes for inflammasome activation, Suilysin is essential for inflammasome activation, which is dependent on the membrane perforation activity to cause cytosolic K+ efflux. Moreover, the mutant strain msly (P353L) expressing mutagenic SLY without hemolytic activity was unable to activate the inflammasome and does not cause STSLS. In summary, we demonstrate that the high membrane perforation activity of the epidemic strain induces a high level of NLRP3 inflammasome activation, which is essential for the development of the cytokine storm and multi-organ dysfunction in STSLS and suggests NLRP3 inflammasome as an attractive target for the treatment of STSLS.


Subject(s)
Cytokines/immunology , Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Shock, Septic/immunology , Streptococcal Infections/immunology , Streptococcus suis/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/immunology , Cytokines/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Inflammasomes/genetics , Mice , Mice, Inbred BALB C , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Shock, Septic/genetics , Shock, Septic/pathology , Streptococcal Infections/genetics , Streptococcal Infections/pathology
17.
Nat Immunol ; 10(3): 241-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19221555

ABSTRACT

The inflammasome is a multiprotein complex that mediates the activation of caspase-1, which promotes secretion of the proinflammatory cytokines interleukin 1beta (IL-1beta) and IL-18, as well as 'pyroptosis', a form of cell death induced by bacterial pathogens. Members of the Nod-like receptor family, including NLRP1, NLRP3 and NLRC4, and the adaptor ASC are critical components of the inflammasome that link microbial and endogenous 'danger' signals to caspase-1 activation. Several diseases are associated with dysregulated activation of caspase-1 and secretion of IL-1beta. Thus, understanding inflammasome pathways may provide insight into disease pathogenesis that might identify potential targets for therapeutic intervention.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Apoptosis Regulatory Proteins/immunology , CARD Signaling Adaptor Proteins/immunology , Calcium-Binding Proteins/immunology , Carrier Proteins/immunology , Caspase 1/immunology , Interleukin-1beta/immunology , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/metabolism , Carrier Proteins/metabolism , Caspase 1/metabolism , Humans , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , NLR Proteins , Protein Interaction Domains and Motifs , Uric Acid/immunology , Uric Acid/metabolism
18.
Am J Kidney Dis ; 77(3): 440-453, 2021 03.
Article in English | MEDLINE | ID: mdl-33487481

ABSTRACT

The understanding and management of membranous nephropathy, a common cause of nephrotic syndrome that is more frequently encountered in adults than in children, has rapidly evolved over the past decade. Identification of target antigens has allowed for more precise molecular diagnoses, and the ability to monitor circulating autoantibodies has added a new vantage point in terms of disease monitoring and decisions about immunosuppression. Although immunosuppression with alkylating agents combined with corticosteroids, or with calcineurin inhibitor-based regimens, has been the historical mainstay of treatment, observational and now randomized controlled trials with the B-cell-depleting agent rituximab have moved this agent to the forefront of therapy for primary membranous nephropathy. In this Core Curriculum, we discuss the typical features of primary and secondary disease; highlight the target antigens such as the phospholipase A2 receptor, thrombospondin type 1 domain-containing 7A, neural epidermal growth factor-like 1, and semaphorin-3B; describe the relationship between the immunologic and clinical courses of disease; and review modern management with supportive care or immunosuppressive treatment based on these composite parameters.


Subject(s)
Autoantibodies/immunology , Glomerulonephritis, Membranous/immunology , Calcium-Binding Proteins/immunology , Glomerulonephritis, Membranous/drug therapy , Glomerulonephritis, Membranous/etiology , Glomerulonephritis, Membranous/pathology , Glucocorticoids/therapeutic use , Graft vs Host Disease/complications , Graft vs Host Disease/immunology , Hepatitis B/complications , Hepatitis C/complications , Humans , Immunologic Factors/therapeutic use , Immunosuppressive Agents , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/immunology , Malaria/complications , Membrane Glycoproteins/immunology , Mixed Connective Tissue Disease/complications , Mixed Connective Tissue Disease/immunology , Neoplasms/complications , Receptors, Phospholipase A2/immunology , Rituximab/therapeutic use , Semaphorins/immunology , Sjogren's Syndrome/complications , Sjogren's Syndrome/immunology , Thrombospondins/immunology
19.
Histopathology ; 79(2): 252-259, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33657658

ABSTRACT

AIMS: Because serous cystadenoma (SCA) does not usually require excision, it is critical to distinguish it from differential diagnoses which do, especially neuroendocrine tumour (NET). The gold standard for diagnosing SCA is assessment of endoscopic ultrasound-guided fine needle aspiration/biopsy (EUS-FNAB) material. Inhibin immunohistochemistry aids this assessment, but such positivity is not absolutely sensitive or specific to SCA. The following is the largest known study of SCA EUS-FNAB specimens and the first to compare four potential SCA immunomarkers between themselves and inhibin, compared against NET. METHODS AND RESULTS: Immunohistochemistry for calponin, mucin 6 (MUC6), glucose transporter 1 (GLUT1) and vascular endothelial growth factor A (VEGFA) was performed on 30 EUS-FNAB and three resection specimens of SCA and 32 EUS-FNAB specimens of NET. GLUT1 and VEGFA were suboptimal as diagnostic immunomarkers of SCA, being expressed by 10 and 44% of NETs, respectively. Further, their expression by cellular constituents of blood which often contaminate EUS-FNAB specimens hampered identification of neoplastic cells, especially in hypocellular samples. While 19% of NETs showed nuclear MUC6 positivity, cytoplasmic expression of the protein showed 100% specificity and sensitivity as an SCA marker. However, assessing MUC6 in EUS-FNAB specimens must also consider the protein's focal expression in physiological pancreatic, gastric or duodenal tissues, which can contaminate these specimens. Calponin was less sensitive (71% versus 100%) but more specific (100% versus 91%) than inhibin, although easier to assess in EUS-FNAB specimens than MUC6. CONCLUSIONS: Of the four potential immunomarkers of SCA suggested by the existing literature, calponin and MUC6 are useful complementary studies to inhibin for application to EUS-FNAB specimens.


Subject(s)
Calcium-Binding Proteins/metabolism , Cystadenoma, Serous/diagnosis , Cystadenoma, Serous/immunology , Endoscopic Ultrasound-Guided Fine Needle Aspiration , Inhibins/metabolism , Microfilament Proteins/metabolism , Mucin-6/metabolism , Adolescent , Adult , Aged , Biomarkers, Tumor , Calcium-Binding Proteins/immunology , Cohort Studies , Cystadenoma, Serous/pathology , Duodenum/pathology , Endoscopic Ultrasound-Guided Fine Needle Aspiration/instrumentation , Female , Glucose Transporter Type 1/metabolism , Humans , Immunohistochemistry , Inhibins/immunology , Male , Microfilament Proteins/immunology , Middle Aged , Neuroendocrine Tumors/pathology , Pancreas/pathology , Stomach/pathology , Synaptophysin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Calponins
20.
Int Arch Allergy Immunol ; 182(6): 465-473, 2021.
Article in English | MEDLINE | ID: mdl-33461195

ABSTRACT

INTRODUCTION: Polcalcins belong to the family of calcium-binding proteins. They are ubiquitous in the plant kingdom and highly conserved, which leads to these panallergens showing a high degree of inter-cross-reactivity. They are responsible for allergic polysensitization, and therefore, their diagnosis is necessary for correct selection of immunotherapy. The objectives were to develop a method to purify native polcalcin with intact allergenic properties and to validate its use for diagnosis of polcalcin sensitization. METHODS: Ole e 3 was purified by immunoaffinity chromatography using anti-rChe a 3 polyclonal antibodies and identified by mass spectrometry. Calcium-binding assays were performed in immunoblot and ELISA assays. Diagnostic capacity of Ole e 3 was analyzed by ELISA and compared to ImmunoCAP with sera from a pollen-sensitized population. Cross-reactivity with other polcalcins was investigated by ImmunoCAP inhibition. RESULTS: Immunogenicity of purified Ole e 3 was not affected by the addition of calcium. However, the presence of a calcium chelator agent completely inhibited IgG binding by immunoblot and produced a 32.3% reduction in IgE binding by ELISA. Ole e 3 enabled diagnosis of polcalcin-sensitized patients, and a good correlation was revealed with ImmunoCAP. A 50% inhibition in IgE binding was obtained with 2.8 ng of Ole e 3 for rBet v 4 and 3.9 ng for rPhl p 7. DISCUSSION/CONCLUSION: Native Ole e 3 was purified by maintaining its allergenic properties. This innovative method enables obtaining this active native allergen to be used for in vivo diagnosis of polcalcin sensitization.


Subject(s)
Allergens/immunology , Antigens, Plant/immunology , Calcium-Binding Proteins/immunology , Hypersensitivity/diagnosis , Hypersensitivity/immunology , Plant Proteins/immunology , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Humans , Immunization , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology
SELECTION OF CITATIONS
SEARCH DETAIL