Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
J Allergy Clin Immunol ; 146(3): 652-666.e11, 2020 09.
Article in English | MEDLINE | ID: mdl-32445838

ABSTRACT

BACKGROUND: Effector functions of IgG Abs are regulated by their Fc N-glycosylation pattern. IgG Fc glycans that lack galactose and terminal sialic acid residues correlate with the severity of inflammatory (auto)immune disorders and have also been linked to protection against viral infection and discussed in the context of vaccine-induced protection. In contrast, sialylated IgG Abs have shown immunosuppressive effects. OBJECTIVE: We sought to investigate IgG glycosylation programming during the germinal center (GC) reaction following immunization of mice with a foreign protein antigen and different adjuvants. METHODS: Mice were analyzed for GC T-cell, B-cell, and plasma cell responses, as well as for antigen-specific serum IgG subclass titers and Fc glycosylation patterns. RESULTS: Different adjuvants induce distinct IgG+ GC B-cell responses with specific transcriptomes and expression levels of the α2,6-sialyltransferase responsible for IgG sialylation that correspond to distinct serum IgG Fc glycosylation patterns. Low IgG Fc sialylation programming in GC B cells was overall highly dependent on the Foxp3- follicular helper T (TFH) cell-inducing cytokine IL-6, here in particular induced by water-in-oil adjuvants and Mycobacterium tuberculosis. Furthermore, low IgG Fc sialylation programming was dependent on adjuvants that induced IL-27 receptor-dependent IFN-γ+ TFH1 cells, IL-6/IL-23-dependent IL-17A+ TFH17 cells, and high ratios of TFH cells to Foxp3+ follicular regulatory T cells. Here, the 2 latter were dependent on M tuberculosis and its cord factor. CONCLUSION: This study's findings regarding adjuvant-dependent GC responses and IgG glycosylation programming may aid in the development of novel vaccination strategies to induce IgG Abs with both high affinity and defined Fc glycosylation patterns in the GC.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens/administration & dosage , Germinal Center/immunology , Immunoglobulin G/immunology , Alum Compounds/administration & dosage , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cytokines/immunology , Female , Freund's Adjuvant/administration & dosage , Glycosylation , Lipopolysaccharides/administration & dosage , Mice, Inbred C57BL , Mice, Knockout , Mineral Oil/administration & dosage , Mycobacterium tuberculosis/immunology , Ovalbumin/administration & dosage , Polysorbates/administration & dosage , Squalene/administration & dosage , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Vaccination
2.
J Exp Med ; 216(4): 807-830, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30819724

ABSTRACT

The lectin-like oxidized LDL receptor 1 (LOX-1) is a key player in the development of atherosclerosis. LOX-1 promotes endothelial activation and dysfunction by mediating uptake of oxidized LDL and inducing pro-atherogenic signaling. However, little is known about modulators of LOX-1-mediated responses. Here, we show that the function of LOX-1 is controlled proteolytically. Ectodomain shedding by the metalloprotease ADAM10 and lysosomal degradation generate membrane-bound N-terminal fragments (NTFs), which we identified as novel substrates of the intramembrane proteases signal peptide peptidase-like 2a and b (SPPL2a/b). SPPL2a/b control cellular LOX-1 NTF levels which, following self-association via their transmembrane domain, can activate MAP kinases in a ligand-independent manner. This leads to an up-regulation of several pro-atherogenic and pro-fibrotic targets including ICAM-1 and the connective tissue growth factor CTGF. Consequently, SPPL2a/b-deficient mice, which accumulate LOX-1 NTFs, develop larger and more advanced atherosclerotic plaques than controls. This identifies intramembrane proteolysis by SPPL2a/b as a novel atheroprotective mechanism via negative regulation of LOX-1 signaling.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Membrane Proteins/metabolism , Proteolysis , Scavenger Receptors, Class E/metabolism , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/genetics , Atherosclerosis/metabolism , Dipeptides/pharmacology , Endothelial Cells/metabolism , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Scavenger Receptors, Class E/genetics , Transfection
3.
J Mol Cell Biol ; 11(11): 979-993, 2019 12 23.
Article in English | MEDLINE | ID: mdl-30753537

ABSTRACT

Dysregulation of the disintegrin-metalloproteinase ADAM10 may contribute to the development of diseases including tumorigenesis and Alzheimer's disease. The mechanisms underlying ADAM10 sheddase activation are incompletely understood. Here, we show that transient exposure of the negatively charged phospholipid phosphatidylserine (PS) is necessarily required. The soluble PS headgroup was found to act as competitive inhibitor of substrate cleavage. Overexpression of the Ca2+-dependent phospholipid scramblase Anoctamin-6 (ANO6) led to increased PS externalization and substrate release. Transfection with a constitutively active form of ANO6 resulted in maximum sheddase activity in the absence of any stimulus. Calcium-dependent ADAM10 activation could not be induced in lymphocytes of patients with Scott syndrome harbouring a missense mutation in ANO6. A putative PS-binding motif was identified in the conserved stalk region. Replacement of this motif resulted in strong reduction of sheddase activity. In conjunction with the recently described 3D structure of the ADAM10 extracellular domain, a model is advanced to explain how surface-exposed PS triggers ADAM10 sheddase function.


Subject(s)
ADAM10 Protein/metabolism , Cell Membrane/metabolism , Enzyme Activation , ADAM10 Protein/chemistry , Amino Acid Sequence , Animals , Anoctamins/metabolism , Biomarkers , COS Cells , Cell Line , Chlorocebus aethiops , Erythrocytes/metabolism , Humans , Membrane Proteins/metabolism , Models, Biological , Phosphoserine/metabolism , Rabbits , Structure-Activity Relationship
4.
Gastroenterology ; 156(1): 145-159.e19, 2019 01.
Article in English | MEDLINE | ID: mdl-30273559

ABSTRACT

BACKGROUND & AIMS: RNase H2 is a holoenzyme, composed of 3 subunits (ribonuclease H2 subunits A, B, and C), that cleaves RNA:DNA hybrids and removes mis-incorporated ribonucleotides from genomic DNA through ribonucleotide excision repair. Ribonucleotide incorporation by eukaryotic DNA polymerases occurs during every round of genome duplication and produces the most frequent type of naturally occurring DNA lesion. We investigated whether intestinal epithelial proliferation requires RNase H2 function and whether RNase H2 activity is disrupted during intestinal carcinogenesis. METHODS: We generated mice with epithelial-specific deletion of ribonuclease H2 subunit B (H2bΔIEC) and mice that also had deletion of tumor-suppressor protein p53 (H2b/p53ΔIEC); we compared phenotypes with those of littermate H2bfl/fl or H2b/p53fl/fl (control) mice at young and old ages. Intestinal tissues were collected and analyzed by histology. We isolated epithelial cells, generated intestinal organoids, and performed RNA sequence analyses. Mutation signatures of spontaneous tumors from H2b/p53ΔIEC mice were characterized by exome sequencing. We collected colorectal tumor specimens from 467 patients, measured levels of ribonuclease H2 subunit B, and associated these with patient survival times and transcriptome data. RESULTS: The H2bΔIEC mice had DNA damage to intestinal epithelial cells and proliferative exhaustion of the intestinal stem cell compartment compared with controls and H2b/p53ΔIEC mice. However, H2b/p53ΔIEC mice spontaneously developed small intestine and colon carcinomas. DNA from these tumors contained T>G base substitutions at GTG trinucleotides. Analyses of transcriptomes of human colorectal tumors associated lower levels of RNase H2 with shorter survival times. CONCLUSIONS: In analyses of mice with disruption of the ribonuclease H2 subunit B gene and colorectal tumors from patients, we provide evidence that RNase H2 functions as a colorectal tumor suppressor. H2b/p53ΔIEC mice can be used to study the roles of RNase H2 in tissue-specific carcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Epithelial Cells/enzymology , Genomic Instability , Intestinal Neoplasms/prevention & control , Intestine, Small/enzymology , Ribonuclease H/metabolism , Animals , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colitis/chemically induced , Colitis/enzymology , Colitis/genetics , Colitis/pathology , DNA Damage , Dextran Sulfate , Disease Models, Animal , Epithelial Cells/pathology , Female , Genetic Predisposition to Disease , Humans , Intestinal Neoplasms/enzymology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Male , Mice, Knockout , Phenotype , Ribonuclease H/deficiency , Ribonuclease H/genetics , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
6.
J Exp Med ; 215(11): 2868-2886, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30254094

ABSTRACT

A coding variant of the inflammatory bowel disease (IBD) risk gene ATG16L1 has been associated with defective autophagy and deregulation of endoplasmic reticulum (ER) function. IL-22 is a barrier protective cytokine by inducing regeneration and antimicrobial responses in the intestinal mucosa. We show that ATG16L1 critically orchestrates IL-22 signaling in the intestinal epithelium. IL-22 stimulation physiologically leads to transient ER stress and subsequent activation of STING-dependent type I interferon (IFN-I) signaling, which is augmented in Atg16l1 ΔIEC intestinal organoids. IFN-I signals amplify epithelial TNF production downstream of IL-22 and contribute to necroptotic cell death. In vivo, IL-22 treatment in Atg16l1 ΔIEC and Atg16l1 ΔIEC/Xbp1 ΔIEC mice potentiates endogenous ileal inflammation and causes widespread necroptotic epithelial cell death. Therapeutic blockade of IFN-I signaling ameliorates IL-22-induced ileal inflammation in Atg16l1 ΔIEC mice. Our data demonstrate an unexpected role of ATG16L1 in coordinating the outcome of IL-22 signaling in the intestinal epithelium.


Subject(s)
Autophagy-Related Proteins/immunology , Carrier Proteins/immunology , Interleukins/immunology , Intestinal Mucosa/immunology , Membrane Proteins/immunology , Nucleotidyltransferases/immunology , Signal Transduction/immunology , Animals , Autophagy-Related Proteins/genetics , Caco-2 Cells , Carrier Proteins/genetics , Genetic Variation , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Interleukins/genetics , Intestinal Mucosa/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Nucleotidyltransferases/genetics , Signal Transduction/genetics , Interleukin-22
7.
Front Immunol ; 9: 587, 2018.
Article in English | MEDLINE | ID: mdl-29662492

ABSTRACT

Aicardi-Goutières syndrome (AGS) is a rare early onset childhood encephalopathy caused by persistent neuroinflammation of autoimmune origin. AGS is a genetic disorder and >50% of affected individuals bear hypomorphic mutations in ribonuclease H2 (RNase H2). All available RNase H2 mouse models so far fail to mimic the prominent CNS involvement seen in AGS. To establish a mouse model recapitulating the human disease, we deleted RNase H2 specifically in the brain, the most severely affected organ in AGS. Although RNase H2ΔGFAP mice lacked the nuclease in astrocytes and a majority of neurons, no disease signs were apparent in these animals. We additionally confirmed these results in a second, neuron-specific RNase H2 knockout mouse line. However, when astrocytes were isolated from brains of RNase H2ΔGFAP mice and cultured under mitogenic conditions, they showed signs of DNA damage and premature senescence. Enhanced expression of interferon-stimulated genes (ISGs) represents the most reliable AGS biomarker. Importantly, primary RNase H2ΔGFAP astrocytes displayed significantly increased ISG transcript levels, which we failed to detect in in vivo in brains of RNase H2ΔGFAP mice. Isolated astrocytes primed by DNA damage, including RNase H2-deficiency, exhibited a heightened innate immune response when exposed to bacterial or viral antigens. Taken together, we established a valid cellular AGS model that utilizes the very cell type responsible for disease pathology, the astrocyte, and phenocopies major molecular defects observed in AGS patient cells.


Subject(s)
Astrocytes/immunology , Astrocytes/metabolism , Autoimmune Diseases/etiology , Inflammation/etiology , Nucleic Acids/immunology , Ribonuclease H/deficiency , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Autoimmune Diseases of the Nervous System/etiology , Autoimmune Diseases of the Nervous System/metabolism , Autoimmune Diseases of the Nervous System/pathology , Biomarkers , Brain/metabolism , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental , Female , Fluorescent Antibody Technique , Immunohistochemistry , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Knockout , Nervous System Malformations/etiology , Nervous System Malformations/metabolism , Nervous System Malformations/pathology , Phenotype
8.
Cell Rep ; 21(8): 2090-2103, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29166602

ABSTRACT

The host metalloprotease meprin ß is required for mucin 2 (MUC2) cleavage, which drives intestinal mucus detachment and prevents bacterial overgrowth. To gain access to the cleavage site in MUC2, meprin ß must be proteolytically shed from epithelial cells. Hence, regulation of meprin ß shedding and activation is important for physiological and pathophysiological conditions. Here, we demonstrate that meprin ß activation and shedding are mutually exclusive events. Employing ex vivo small intestinal organoid and cell culture experiments, we found that ADAM-mediated shedding is restricted to the inactive pro-form of meprin ß and is completely inhibited upon its conversion to the active form at the cell surface. This strict regulation of meprin ß activity can be overridden by pathogens, as demonstrated for the bacterial protease Arg-gingipain (RgpB). This secreted cysteine protease potently converts membrane-bound meprin ß into its active form, impairing meprin ß shedding and its function as a mucus-detaching protease.


Subject(s)
Adhesins, Bacterial/metabolism , Cysteine Endopeptidases/metabolism , Metalloendopeptidases/metabolism , Metalloproteases/metabolism , Amino Acid Sequence/genetics , Animals , Cell Membrane/metabolism , Epithelial Cells/metabolism , Female , Gingipain Cysteine Endopeptidases , HEK293 Cells , Humans , Male , Metalloendopeptidases/genetics , Mice, Transgenic , Mucin-2/genetics , Mucin-2/metabolism
9.
Hum Mol Genet ; 26(20): 3960-3972, 2017 10 15.
Article in English | MEDLINE | ID: mdl-29016854

ABSTRACT

Hypomorphic mutations in the DNA repair enzyme RNase H2 cause the neuroinflammatory autoimmune disorder Aicardi-Goutières syndrome (AGS). Endogenous nucleic acids are believed to accumulate in patient cells and instigate pathogenic type I interferon expression. However, the underlying nucleic acid species amassing in the absence of RNase H2 has not been established yet. Here, we report that murine RNase H2 knockout cells accumulated cytosolic DNA aggregates virtually indistinguishable from micronuclei. RNase H2-dependent micronuclei were surrounded by nuclear lamina and most of them contained damaged DNA. Importantly, they induced expression of interferon-stimulated genes (ISGs) and co-localized with the nucleic acid sensor cGAS. Moreover, micronuclei associated with RNase H2 deficiency were cleared by autophagy. Consequently, induction of autophagy by pharmacological mTOR inhibition resulted in a significant reduction of cytosolic DNA and the accompanied interferon signature. Autophagy induction might therefore represent a viable therapeutic option for RNase H2-dependent disease. Endogenous retroelements have previously been proposed as a source of self-nucleic acids triggering inappropriate activation of the immune system in AGS. We used human RNase H2-knockout cells generated by CRISPR/Cas9 to investigate the impact of RNase H2 on retroelement propagation. Surprisingly, replication of LINE-1 and Alu elements was blunted in cells lacking RNase H2, establishing RNase H2 as essential host factor for the mobilisation of endogenous retrotransposons.


Subject(s)
Autoimmune Diseases of the Nervous System/enzymology , Micronucleus, Germline/enzymology , Nervous System Malformations/enzymology , Ribonuclease H/deficiency , Animals , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/metabolism , Autoimmune Diseases of the Nervous System/pathology , Autophagy/genetics , DNA/genetics , DNA Damage , DNA Replication , Mice , Mice, Knockout , Micronucleus, Germline/genetics , Micronucleus, Germline/immunology , Mutation , Nervous System Malformations/genetics , Nervous System Malformations/metabolism , Nervous System Malformations/pathology , Ribonuclease H/genetics , Ribonuclease H/metabolism
10.
Oncotarget ; 8(33): 54873-54888, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28903388

ABSTRACT

Transendothelial cell migration (TEM) is crucial for inflammation and metastasis. The adhesion molecule CD99 was shown to be important for correct immune cell extravasation and is highly expressed on certain cancer cells. Recently, we demonstrated that ectodomain shedding of CD99 by the metalloprotease meprin ß promotes TEM in vitro. In this study, we employed an acute inflammation model (air pouch/carrageenan) and found significantly less infiltrated cells in meprin ß knock-out animals validating the previously observed pro-inflammatory activity. To further analyze the impact of meprin ß on CD99 shedding with regard to cell adhesion and proliferation we characterized two lung cancer associated CD99 variants (D92H, D92Y), carrying point mutations at the main cleavage site. Interestingly, ectodomain shedding of these variants by meprin ß was still detectable. However the cleavage site shifted to adjacent positions. Nevertheless, expression of CD99 variants D92H and D92Y revealed partial misfolding and proteasomal degradation. A previously observed influence of CD99 on Src activation and increased proliferation could not be confirmed in this study, independent of wild-type CD99 or the variants D92H and D92Y. However, we identified meprin ß as a potent inducer of Src phosphorylation. Importantly, we found significantly increased cell migration when expressing the cancer-associated CD99 variant D92H compared to the wild-type protein.

11.
Sci Rep ; 7: 44053, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28276471

ABSTRACT

Soluble Interleukin-6 receptor (sIL-6R) mediated trans-signaling is an important pro-inflammatory stimulus associated with pathological conditions, such as arthritis, neurodegeneration and inflammatory bowel disease. The sIL-6R is generated proteolytically from its membrane bound form and A Disintegrin And Metalloprotease (ADAM) 10 and 17 were shown to perform ectodomain shedding of the receptor in vitro and in vivo. However, under certain conditions not all sIL-6R could be assigned to ADAM10/17 activity. Here, we demonstrate that the IL-6R is a shedding substrate of soluble meprin α and membrane bound meprin ß, resulting in bioactive sIL-6R that is capable of inducing IL-6 trans-signaling. We determined cleavage within the N-terminal part of the IL-6R stalk region, distinct from the cleavage site reported for ADAM10/17. Interestingly, meprin ß can be shed from the cell surface by ADAM10/17 and the observation that soluble meprin ß is not capable of shedding the IL-6R suggests a regulatory mechanism towards trans-signaling. Additionally, we observed a significant negative correlation of meprin ß expression and IL-6R levels on human granulocytes, providing evidence for in vivo function of this proteolytic interaction.


Subject(s)
Metalloendopeptidases/metabolism , Receptors, Interleukin-6/metabolism , Signal Transduction , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , ADAM17 Protein/genetics , ADAM17 Protein/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Metalloendopeptidases/genetics , Receptors, Interleukin-6/genetics , Solubility
12.
Biochim Biophys Acta Mol Cell Res ; 1864(1): 217-230, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27818272

ABSTRACT

Despite existing knowledge about the role of the A Disintegrin and Metalloproteinase 10 (ADAM10) as the α-secretase involved in the non-amyloidogenic processing of the amyloid precursor protein (APP) and Notch signalling we have only limited information about its regulation. In this study, we have identified ADAM10 interactors using a split ubiquitin yeast two hybrid approach. Tetraspanin 3 (Tspan3), which is highly expressed in the murine brain and elevated in brains of Alzheimer´s disease (AD) patients, was identified and confirmed to bind ADAM10 by co-immunoprecipitation experiments in mammalian cells in complex with APP and the γ-secretase protease presenilin. Tspan3 expression increased the cell surface levels of its interacting partners and was mainly localized in early and late endosomes. In contrast to the previously described ADAM10-binding tetraspanins, Tspan3 did not affect the endoplasmic reticulum to plasma membrane transport of ADAM10. Heterologous Tspan3 expression significantly increased the appearance of carboxy-terminal cleavage products of ADAM10 and APP, whereas N-cadherin ectodomain shedding appeared unaffected. Inhibiting the endocytosis of Tspan3 by mutating a critical cytoplasmic tyrosine-based internalization motif led to increased surface expression of APP and ADAM10. After its downregulation in neuroblastoma cells and in brains of Tspan3-deficient mice, ADAM10 and APP levels appeared unaltered possibly due to a compensatory increase in the expression of Tspans 5 and 7, respectively. In conclusion, our data suggest that Tspan3 acts in concert with other tetraspanins as a stabilizing factor of active ADAM10, APP and the γ-secretase complex at the plasma membrane and within the endocytic pathway.


Subject(s)
ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/genetics , Endosomes/metabolism , Membrane Proteins/genetics , Presenilins/genetics , Tetraspanins/genetics , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Brain Chemistry , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Endocytosis , Endosomes/chemistry , Gene Expression Regulation , HEK293 Cells , Humans , Membrane Proteins/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Presenilins/metabolism , Protein Binding , Protein Transport , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Tetraspanins/metabolism , Two-Hybrid System Techniques
13.
Hepatology ; 65(1): 89-103, 2017 01.
Article in English | MEDLINE | ID: mdl-27770462

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most frequent tumors worldwide with rising incidence. The inflammatory cytokine, interleukin-6 (IL-6), is a critical mediator of HCC development. It can signal through two distinct pathways: the IL-6 classic and the IL-6 trans-signaling pathway. Whereas IL-6 classic signaling is important for innate and acquired immunity, IL-6 trans-signaling has been linked to accelerated liver regeneration and several chronic inflammatory pathologies. However, its implication in liver tumorigenesis has not been addressed yet. Here, we show that IL-6 trans-signaling, but not IL-6 classic signaling, is essential to promote hepatocellular carcinogenesis by two mechanisms: First, it prevents DNA-damage-induced hepatocyte apoptosis through suppression of p53 and enhances ß-catenin activation and tumor proliferation. Second, IL-6 trans-signaling directly induces endothelial cell proliferation to promote tumor angiogenesis. Consequently, soluble gp130 fused to Fc transgenic mice lacking IL-6 trans-signaling are largely protected from tumor formation in a diethylnitrosamine/3,3',5,5'-tetrachloro-1,4-bis(pyridyloxy)benzene model of HCC. CONCLUSION: IL-6 trans-signaling, and not IL-6 classic signaling, is mandatory for development of hepatocellular carcinogenesis. Therefore, specific inhibition of IL-6 trans-signaling, rather than total inhibition of IL-6 signaling, is sufficient to blunt tumor initiation and impair tumor progression without compromising IL-6 classic signaling-driven protective immune responses. (Hepatology 2017;65:89-103).


Subject(s)
Carcinoma, Hepatocellular/etiology , Interleukin-6/physiology , Liver Neoplasms/etiology , Animals , Male , Mice , Signal Transduction
14.
J Immunol ; 197(9): 3705-3715, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27698010

ABSTRACT

Neutrophil and mononuclear cell infiltration during inflammatory processes is highly regulated. The first cells at the site of infection or inflammation are neutrophils, followed by mononuclear cells. IL-6 plays an important role during inflammatory states. It has been shown in several models that the soluble form of IL-6R (sIL-6R) is involved in the recruitment of mononuclear cells by a mechanism called IL-6 trans-signaling. It had been speculated that sIL-6R was generated at the site of inflammation by shedding from neutrophils via activation of the metalloprotease ADAM17. Attempts to genetically delete the floxed ADAM17 gene selectively in myeloid cells infiltrating an air pouch cavity upon injection of carrageenan failed because in transgenic mice, LysMcre did not lead to appreciable loss of the ADAM17 protein in these cells. We therefore used ADAM17 hypomorphic mice, which only express ∼5% of ADAM17 wild-type levels in all tissues and show virtually no shedding of all tested ADAM17 substrates, to clarify the role of ADAM17 during local inflammation in the murine air pouch model. In the present study, we demonstrate that although IL-6 and the trans-signaling mechanism is mandatory for cellular infiltration in this model, it is not ADAM17-mediated shedding of IL-6R within the pouch that orchestrates this inflammatory process. Instead, we demonstrate that sIL-6R is infiltrating from the circulation in an ADAM17-independent process. Our data suggest that this infiltrating sIL-6R, which is needed for IL-6 trans-signaling, is involved in the controlled resolution of an acute inflammatory episode.


Subject(s)
ADAM17 Protein/metabolism , Inflammation/immunology , Macrophages/immunology , Neutrophils/immunology , Receptors, Interleukin-6/metabolism , ADAM17 Protein/genetics , Animals , Carrageenan , Cell Movement , Cells, Cultured , Inflammation/chemically induced , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
15.
Oncotarget ; 7(32): 51096-51106, 2016 Aug 09.
Article in English | MEDLINE | ID: mdl-27191495

ABSTRACT

The role of STAT1 and STAT3 for colorectal carcinoma (CRC) development and progression is controversial. We evaluated 414 CRC patient samples on tissue microarrays for differential expression of STAT1 and STAT3 protein levels and correlated ratios with clinical parameters. Concomitant absence of nuclear STAT1 and STAT3 expression was associated with significantly reduced median survival by ≥33 months (p=0.003). To gain insight into underlying mechanisms, we generated four CRC cell lines with STAT3 knockdown. The cell lines harbor different known mutational drivers and were xenografted into SCID mice to analyze the influence of STAT3 on their tumor growth behavior. Experimental downregulation of STAT3 expression had differential, cell-line specific effects on STAT1 expression levels. STAT1 consistently showed nuclear localization irrespective of its tyrosine phosphorylation status. Two characteristic STAT1/3 expression patterns with opposite growth behavior could be distinguished: cell lines with a low STAT1/high STAT3 ratio showed faster tumor growth in xenografts. In contrast, xenografts of cell lines showing high STAT1 and low STAT3 levels grew slower. Importantly, these ratios reflected clinical outcome in CRC patients as well. We conclude that the ratio of STAT1 to STAT3 expression is a key determinant of CRC progression and that STAT1 counteracts pro-tumorigenic STAT3 signaling. Thus, we suggest that the STAT3/STAT1 ratios are better clinical predictors in CRC as compared to STAT3 or STAT1 levels alone.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , Animals , Biomarkers, Tumor/genetics , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/genetics , Colorectal Neoplasms/diagnosis , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, SCID , Prognosis
16.
Sci Rep ; 6: 25550, 2016 05 06.
Article in English | MEDLINE | ID: mdl-27151651

ABSTRACT

Limited proteolysis of the Interleukin-6 Receptor (IL-6R) leads to the release of the IL-6R ectodomain. Binding of the cytokine IL-6 to the soluble IL-6R (sIL-6R) results in an agonistic IL-6/sIL-6R complex, which activates cells via gp130 irrespective of whether the cells express the IL-6R itself. This signaling pathway has been termed trans-signaling and is thought to mainly account for the pro-inflammatory properties of IL-6. A Disintegrin And Metalloprotease 10 (ADAM10) and ADAM17 are the major proteases that cleave the IL-6R. We have previously shown that deletion of a ten amino acid long stretch within the stalk region including the cleavage site prevents ADAM17-mediated cleavage, whereas the receptor retained its full biological activity. In the present study, we show that deletion of a triple serine (3S) motif (Ser-359 to Ser-361) adjacent to the cleavage site is sufficient to prevent IL-6R cleavage by ADAM17, but not ADAM10. We find that the impaired shedding is caused by the reduced distance between the cleavage site and the plasma membrane. Positioning of the cleavage site in greater distance towards the plasma membrane abrogates ADAM17-mediated shedding and reveals a novel cleavage site of ADAM10. Our findings underline functional differences in IL-6R proteolysis by ADAM10 and ADAM17.


Subject(s)
ADAM10 Protein/metabolism , ADAM17 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Membrane Proteins/metabolism , Proteolysis , Receptors, Interleukin-6/metabolism , ADAM10 Protein/genetics , ADAM17 Protein/genetics , Amyloid Precursor Protein Secretases/genetics , Cell Line , DNA Mutational Analysis , Humans , Membrane Proteins/genetics , Sequence Deletion
17.
J Biol Chem ; 291(20): 10551-61, 2016 May 13.
Article in English | MEDLINE | ID: mdl-26961870

ABSTRACT

IL-23 (interleukin 23) regulates immune responses against pathogens and plays a major role in the differentiation and maintenance of TH17 cells and the development of autoimmune diseases and cancer. The IL-23 receptor (IL-23R) complex consists of the unique IL-23R and the common IL-12 receptor ß1 (IL-12Rß1). Differential splicing generates antagonistic soluble IL-23R (sIL-23R) variants, which might limit IL-23-mediated immune responses. Here, ectodomain shedding of human and murine IL-23R was identified as an alternative pathway for the generation of sIL-23R. Importantly, proteolytically released sIL-23R has IL-23 binding activity. Shedding of IL-23R was induced by stimulation with the phorbol ester phorbol 12-myristate 13-acetate (PMA), but not by ionomycin. PMA-induced shedding was abrogated by an ADAM (A disintegrin and metalloprotease) 10 and 17 selective inhibitor, but not by an ADAM10 selective inhibitor. ADAM17-deficient but not ADAM10-deficient HEK293 cells failed to shed IL-23R after PMA stimulation, demonstrating that ADAM17 but not ADAM10 cleaves the IL-23R. Constitutive shedding was, however, inhibited by an ADAM10 selective inhibitor. Using deletions and specific amino acid residue exchanges, we identified critical determinants of ectodomain shedding within the stalk region of the IL-23R. Finally, interaction studies identified domains 1 and 3 of the IL-23R as the main ADAM17 binding sites. In summary, we describe human and murine IL-23R as novel targets for protein ectodomain shedding by ADAM10 and ADAM17.


Subject(s)
ADAM10 Protein/metabolism , ADAM17 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Membrane Proteins/metabolism , Receptors, Interleukin/metabolism , Alternative Splicing , Animals , Cell-Derived Microparticles/metabolism , HEK293 Cells , Half-Life , Humans , Interleukin-23/metabolism , Mice , Receptors, Interleukin/chemistry , Receptors, Interleukin/genetics , Solubility , Substrate Specificity , Tetradecanoylphorbol Acetate/pharmacology
18.
EMBO J ; 35(8): 831-44, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26903602

ABSTRACT

Aicardi-Goutières syndrome (AGS) provides a monogenic model of nucleic acid-mediated inflammation relevant to the pathogenesis of systemic autoimmunity. Mutations that impair ribonuclease (RNase) H2 enzyme function are the most frequent cause of this autoinflammatory disorder of childhood and are also associated with systemic lupus erythematosus. Reduced processing of eitherRNA:DNAhybrid or genome-embedded ribonucleotide substrates is thought to lead to activation of a yet undefined nucleic acid-sensing pathway. Here, we establishRnaseh2b(A174T/A174T)knock-in mice as a subclinical model of disease, identifying significant interferon-stimulated gene (ISG) transcript upregulation that recapitulates theISGsignature seen inAGSpatients. The inflammatory response is dependent on the nucleic acid sensor cyclicGMP-AMPsynthase (cGAS) and its adaptorSTINGand is associated with reduced cellular ribonucleotide excision repair activity and increasedDNAdamage. This suggests thatcGAS/STINGis a key nucleic acid-sensing pathway relevant toAGS, providing additional insight into disease pathogenesis relevant to the development of therapeutics for this childhood-onset interferonopathy and adult systemic autoimmune disorders.


Subject(s)
Autoimmune Diseases of the Nervous System/genetics , Immunity, Innate/genetics , Membrane Proteins/immunology , Mutation, Missense , Nervous System Malformations/genetics , Nucleotidyltransferases/immunology , Ribonuclease H/genetics , Ribonucleases/genetics , Animals , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/metabolism , Autoimmunity/genetics , DNA Damage , Gene Expression Regulation , Humans , Interferons/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Mutant Strains , Nervous System Malformations/immunology , Nervous System Malformations/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Ribonuclease H/metabolism
19.
J Leukoc Biol ; 99(5): 749-60, 2016 05.
Article in English | MEDLINE | ID: mdl-26561568

ABSTRACT

The cytokine IL-6 is part of a regulatory signaling network that controls immune responses. IL-6 binds either to the membrane-bound IL-6 receptor-α (classic signaling) or to the soluble IL-6 receptor-α (trans-signaling) to initiate signal transduction via gp130 activation. Because classic and trans-signaling of IL-6 fulfill different tasks during immune responses, controlled shedding of the membrane-bound IL-6 receptor-α from the surface of immune cells can be considered a central regulator of IL-6 function. The results from cell culture-based experiments have implicated both a disintegrin and metalloprotease 10 and a disintegrin and metalloprotease 17 in IL-6 receptor-α shedding. However, the nature of the protease mediating IL-6 receptor-α release in vivo is not yet known. We used hypomorphic a disintegrin and metalloprotease 17 mice and conditional a disintegrin and metalloprotease 10 knock-out mice to identify the natural protease of the murine IL-6 receptor-α. Circulating homeostatic soluble IL-6 receptor-α levels are not dependent on a disintegrin and metalloprotease 10 or 17 activity. However, during Listeria monocytogenes infection, IL-6 receptor-α cleavage by the α-secretase a disintegrin and metalloprotease 17 is rapidly induced from the surface of different leukocyte populations. In contrast, CD4-Cre-driven a disintegrin and metalloprotease 10 deletion in T cells did not influence IL-6 receptor-α shedding from these cells after L. monocytogenes infection. A disintegrin and metalloprotease 17 was also required for IL-6 receptor-α ectodomain cleavage and release during endotoxemia. These results demonstrate a novel physiologic role for a disintegrin and metalloprotease 17 in regulating murine IL-6 signals during inflammatory processes.


Subject(s)
ADAM17 Protein/metabolism , Cell Membrane/metabolism , Inflammation/metabolism , Inflammation/pathology , Interleukin-6/metabolism , Leukocytes/metabolism , Receptors, Interleukin-6/metabolism , Signal Transduction , ADAM10 Protein/metabolism , Animals , Cells, Cultured , Endotoxemia/immunology , Endotoxemia/pathology , Fibroblasts/metabolism , Inflammation/blood , Interleukin-6/blood , Listeria monocytogenes/physiology , Listeriosis/blood , Listeriosis/immunology , Listeriosis/pathology , Mice, Inbred C57BL , Receptors, Interleukin-6/blood , T-Lymphocytes/metabolism
20.
J Biol Chem ; 290(43): 26059-71, 2015 Oct 23.
Article in English | MEDLINE | ID: mdl-26359498

ABSTRACT

Generation of the soluble interleukin-6 receptor (sIL-6R) is a prerequisite for pathogenic IL-6 trans-signaling, which constitutes a distinct signaling pathway of the pleiotropic cytokine interleukin-6 (IL-6). Although in vitro experiments using ectopically overexpressed IL-6R and candidate proteases revealed major roles for the metalloproteinases ADAM10 and ADAM17 in IL-6R shedding, the identity of the protease(s) cleaving IL-6R in more physiological settings, or even in vivo, remains unknown. By taking advantage of specific pharmacological inhibitors and primary cells from ADAM-deficient mice we established that endogenous IL-6R of both human and murine origin is shed by ADAM17 in an induced manner, whereas constitutive release of endogenous IL-6R is largely mediated by ADAM10. Although circulating IL-6R levels are altered in various diseases, the origin of blood-borne IL-6R is still poorly understood. It has been shown previously that ADAM17 hypomorphic mice exhibit unaltered levels of serum sIL-6R. Here, by quantification of serum sIL-6R in protease-deficient mice as well as human patients we also excluded ADAM10, ADAM8, neutrophil elastase, cathepsin G, and proteinase 3 from contributing to circulating sIL-6R. Furthermore, we ruled out alternative splicing of the IL-6R mRNA as a potential source of circulating sIL-6R in the mouse. Instead, we found full-length IL-6R on circulating microvesicles, establishing microvesicle release as a novel mechanism for sIL-6R generation.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Membrane Proteins/metabolism , Protein Isoforms/metabolism , Receptors, Interleukin-6/metabolism , ADAM10 Protein , ADAM17 Protein , Animals , Cell Line , Humans , Lipopolysaccharides/pharmacology , Mice , Monocytes/drug effects , Monocytes/metabolism , Proteolysis , RNA Splicing , Receptors, Interleukin-6/genetics , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...