Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nat Immunol ; 21(4): 381-387, 2020 04.
Article in English | MEDLINE | ID: mdl-32205881

ABSTRACT

Protein ubiquitination regulates protein stability and modulates the composition of signaling complexes. A20 is a negative regulator of inflammatory signaling, but the molecular mechanisms involved are ill understood. Here, we generated Tnfaip3 gene-targeted A20 mutant mice bearing inactivating mutations in the zinc finger 7 (ZnF7) and ZnF4 ubiquitin-binding domains, revealing that binding to polyubiquitin is essential for A20 to suppress inflammatory disease. We demonstrate that a functional ZnF7 domain was required for recruiting A20 to the tumor necrosis factor receptor 1 (TNFR1) signaling complex and to suppress inflammatory signaling and cell death. The combined inactivation of ZnF4 and ZnF7 phenocopied the postnatal lethality and severe multiorgan inflammation of A20-deficient mice. Conditional tissue-specific expression of mutant A20 further revealed the key role of ubiquitin-binding in myeloid and intestinal epithelial cells. Collectively, these results demonstrate that the anti-inflammatory and cytoprotective functions of A20 are largely dependent on its ubiquitin-binding properties.


Subject(s)
Inflammation/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Animals , Epithelial Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Polyubiquitin/metabolism , Protein Binding/physiology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin/metabolism , Ubiquitination/physiology , Zinc Fingers/physiology
2.
EMBO Rep ; 23(12): e55233, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36194667

ABSTRACT

The anti-inflammatory protein A20 serves as a critical brake on NF-κB signaling and NF-κB-dependent inflammation. In humans, polymorphisms in or near the TNFAIP3/A20 gene have been associated with several inflammatory disorders, including rheumatoid arthritis (RA), and experimental studies in mice have demonstrated that myeloid-specific A20 deficiency causes the development of a severe polyarthritis resembling human RA. Myeloid A20 deficiency also promotes osteoclastogenesis in mice, suggesting a role for A20 in the regulation of osteoclast differentiation and bone formation. We show here that osteoclast-specific A20 knockout mice develop severe osteoporosis, but not inflammatory arthritis. In vitro, osteoclast precursor cells from A20 deficient mice are hyper-responsive to RANKL-induced osteoclastogenesis. Mechanistically, we show that A20 is recruited to the RANK receptor complex within minutes of ligand binding, where it restrains NF-κB activation independently of its deubiquitinating activity but through its zinc finger (ZnF) 4 and 7 ubiquitin-binding functions. Together, these data demonstrate that A20 acts as a regulator of RANK-induced NF-κB signaling to control osteoclast differentiation, assuring proper bone development and turnover.


Subject(s)
NF-kappa B , Humans , Animals , Mice
3.
Eur Respir J ; 59(5)2022 05.
Article in English | MEDLINE | ID: mdl-34588194

ABSTRACT

BACKGROUND: Chronic airway inflammation is the main driver of pathogenesis in respiratory diseases such as severe asthma, chronic obstructive pulmonary disease, cystic fibrosis (CF) and bronchiectasis. While the role of common pathogens in airway inflammation is widely recognised, the influence of other microbiota members is still poorly understood. METHODS: We hypothesised that the lung microbiota contains bacteria with immunomodulatory activity which modulate net levels of immune activation by key respiratory pathogens. Therefore, we assessed the immunomodulatory effect of several members of the lung microbiota frequently reported as present in CF lower respiratory tract samples. RESULTS: We show that Rothia mucilaginosa, a common resident of the oral cavity that is also often detectable in the lower airways in chronic disease, has an inhibitory effect on pathogen- or lipopolysaccharide-induced pro-inflammatory responses, in vitro (three-dimensional cell culture model) and in vivo (mouse model). Furthermore, in a cohort of adults with bronchiectasis, the abundance of Rothia species was negatively correlated with pro-inflammatory markers (interleukin (IL)-8 and IL-1ß) and matrix metalloproteinase (MMP)-1, MMP-8 and MMP-9 in sputum. Mechanistic studies revealed that R. mucilaginosa inhibits NF-κB pathway activation by reducing the phosphorylation of IκBα and consequently the expression of NF-κB target genes. CONCLUSIONS: These findings indicate that the presence of R. mucilaginosa in the lower airways potentially mitigates inflammation, which could in turn influence the severity and progression of chronic respiratory disorders.


Subject(s)
Bronchiectasis , Cystic Fibrosis , Animals , Anti-Inflammatory Agents/pharmacology , Bacteria , Bronchiectasis/microbiology , Humans , Inflammation , Lung , Mice , NF-kappa B , Sputum/microbiology
4.
Immunity ; 35(1): 82-96, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21723156

ABSTRACT

Dendritic cells (DCs) regulate both immunity and tolerance. Here we have shown that the ubiquitin editing enzyme A20 (Tnfaip3) determines the activation threshold of DCs, via control of canonical NF-κB activation. Tnfaip3(fl/fl)Cd11c-cre(+) mice lacking A20 in DCs demonstrated spontaneous proliferation of conventional and double-negative T cells, their conversion to interferon-γ (IFN-γ)-producing effector cells, and expansion of plasma cells. They developed ds-DNA antibodies, nephritis, the antiphospholipid syndrome, and lymphosplenomegaly-features of systemic lupus erythematosus-and extramedullary hematopoiesis. A20-deficient DCs were resistant to apoptosis, caused by increased sensitivity to CD40L and RANKL prosurvival signals and upregulation of antiapoptotic proteins Bcl-2 and Bcl-x. They captured injected apoptotic cells more efficiently, resisted the inhibitory effects of apoptotic cells, and induced self-reactive effector lymphocytes. Because genetic polymorphisms in TNFAIP3 are associated with human autoimmune disorders, these findings identify A20-mediated control of DC activation as a crucial checkpoint in the development of systemic autoimmunity.


Subject(s)
Cysteine Endopeptidases/metabolism , Dendritic Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lupus Erythematosus, Systemic/immunology , Plasma Cells/metabolism , T-Lymphocytes/metabolism , Animals , Antibodies, Antinuclear/blood , Apoptosis/genetics , Autoimmunity/genetics , CD40 Ligand/metabolism , Cell Proliferation , Cells, Cultured , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Humans , Interferon-gamma/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Lupus Erythematosus, Systemic/blood , Mice , Mice, Mutant Strains , Mutation/genetics , Plasma Cells/immunology , Plasma Cells/pathology , RANK Ligand/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Necrosis Factor alpha-Induced Protein 3
5.
Nature ; 513(7516): 95-9, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25186904

ABSTRACT

Receptor interacting protein kinase 1 (RIPK1) has an essential role in the signalling triggered by death receptors and pattern recognition receptors. RIPK1 is believed to function as a node driving NF-κB-mediated cell survival and inflammation as well as caspase-8 (CASP8)-dependent apoptotic or RIPK3/MLKL-dependent necroptotic cell death. The physiological relevance of this dual function has remained elusive because of the perinatal death of RIPK1 full knockout mice. To circumvent this problem, we generated RIPK1 conditional knockout mice, and show that mice lacking RIPK1 in intestinal epithelial cells (IECs) spontaneously develop severe intestinal inflammation associated with IEC apoptosis leading to early death. This early lethality was rescued by antibiotic treatment, MYD88 deficiency or tumour-necrosis factor (TNF) receptor 1 deficiency, demonstrating the importance of commensal bacteria and TNF in the IEC Ripk1 knockout phenotype. CASP8 deficiency, but not RIPK3 deficiency, rescued the inflammatory phenotype completely, indicating the indispensable role of RIPK1 in suppressing CASP8-dependent apoptosis but not RIPK3-dependent necroptosis in the intestine. RIPK1 kinase-dead knock-in mice did not exhibit any sign of inflammation, suggesting that RIPK1-mediated protection resides in its kinase-independent platform function. Depletion of RIPK1 in intestinal organoid cultures sensitized them to TNF-induced apoptosis, confirming the in vivo observations. Unexpectedly, TNF-mediated NF-κB activation remained intact in these organoids. Our results demonstrate that RIPK1 is essential for survival of IECs, ensuring epithelial homeostasis by protecting the epithelium from CASP8-mediated IEC apoptosis independently of its kinase activity and NF-κB activation.


Subject(s)
Apoptosis , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/metabolism , Homeostasis , Intestinal Mucosa/metabolism , Intestines/cytology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , Caspase 8/genetics , Caspase 8/metabolism , Cell Survival/drug effects , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelium/drug effects , Epithelium/pathology , Female , Gene Deletion , Homeostasis/drug effects , Inflammation/metabolism , Inflammation/pathology , Intestines/drug effects , Intestines/pathology , Male , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , NF-kappa B/metabolism , Necrosis , Organoids/cytology , Organoids/drug effects , Organoids/enzymology , Organoids/metabolism , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptors, Tumor Necrosis Factor, Type I/deficiency , Survival Analysis , Tumor Necrosis Factors/pharmacology
6.
PLoS Pathog ; 12(1): e1005410, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26815999

ABSTRACT

A20 negatively regulates multiple inflammatory signalling pathways. We here addressed the role of A20 in club cells (also known as Clara cells) of the bronchial epithelium in their response to influenza A virus infection. Club cells provide a niche for influenza virus replication, but little is known about the functions of these cells in antiviral immunity. Using airway epithelial cell-specific A20 knockout (A20AEC-KO) mice, we show that A20 in club cells critically controls innate immune responses upon TNF or double stranded RNA stimulation. Surprisingly, A20AEC-KO mice are better protected against influenza A virus challenge than their wild type littermates. This phenotype is not due to decreased viral replication. Instead host innate and adaptive immune responses and lung damage are reduced in A20AEC-KO mice. These attenuated responses correlate with a dampened cytotoxic T cell (CTL) response at later stages during infection, indicating that A20AEC-KO mice are better equipped to tolerate Influenza A virus infection. Expression of the chemokine CCL2 (also named MCP-1) is particularly suppressed in the lungs of A20AEC-KO mice during later stages of infection. When A20AEC-KO mice were treated with recombinant CCL2 the protective effect was abrogated demonstrating the crucial contribution of this chemokine to the protection of A20AEC-KO mice to Influenza A virus infection. Taken together, we propose a mechanism of action by which A20 expression in club cells controls inflammation and antiviral CTL responses in response to influenza virus infection.


Subject(s)
Cysteine Endopeptidases/immunology , Cytotoxicity, Immunologic/immunology , Intracellular Signaling Peptides and Proteins/immunology , Orthomyxoviridae Infections/immunology , Respiratory Mucosa/immunology , Animals , Cysteine Endopeptidases/deficiency , Flow Cytometry , Immunity, Innate/immunology , Immunoblotting , Immunohistochemistry , Influenza A virus , Intracellular Signaling Peptides and Proteins/deficiency , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor alpha-Induced Protein 3
7.
Eur J Immunol ; 46(4): 971-80, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26677802

ABSTRACT

Optineurin (OPTN) is an evolutionary conserved and ubiquitously expressed ubiquitin-binding protein that has been implicated in glaucoma, Paget bone disease, amyotrophic lateral sclerosis, and other neurodegenerative diseases. From in vitro studies, OPTN was shown to suppress TNF-induced NF-κB signaling and virus-induced IRF signaling, and was identified as an autophagy receptor required for the clearance of cytosolic Salmonella upon infection. To assess the in vivo functions of OPTN in inflammation and infection, we generated OPTN-deficient mice. OPTN knockout mice are born with normal Mendelian distribution and develop normally without any signs of spontaneous organ abnormality or inflammation. However, no differences in NF-κB activation could be observed in OPTN knockout mice or fibroblasts derived from these mice upon TNF or LPS treatment. Primary bone marrow-derived macrophages from OPTN-deficient mice had slightly impaired IRF signaling and reduced IFN type I production in response to LPS or poly(I,C). Finally, OPTN-deficient mice were more susceptible to infection with Salmonella, confirming in vivo the importance of OPTN in bacterial clearance.


Subject(s)
Eye Proteins/genetics , NF-kappa B/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Cycle Proteins , Fibroblasts/immunology , Influenza A Virus, H3N2 Subtype/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Type I/biosynthesis , Lipopolysaccharides/pharmacology , Macrophages/immunology , Membrane Transport Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Poly I-C/pharmacology , Salmonella Infections/microbiology , Signal Transduction/immunology
8.
PLoS Pathog ; 8(3): e1002570, 2012.
Article in English | MEDLINE | ID: mdl-22396652

ABSTRACT

The innate immune response provides the first line of defense against viruses and other pathogens by responding to specific microbial molecules. Influenza A virus (IAV) produces double-stranded RNA as an intermediate during the replication life cycle, which activates the intracellular pathogen recognition receptor RIG-I and induces the production of proinflammatory cytokines and antiviral interferon. Understanding the mechanisms that regulate innate immune responses to IAV and other viruses is of key importance to develop novel therapeutic strategies. Here we used myeloid cell specific A20 knockout mice to examine the role of the ubiquitin-editing protein A20 in the response of myeloid cells to IAV infection. A20 deficient macrophages were hyperresponsive to double stranded RNA and IAV infection, as illustrated by enhanced NF-κB and IRF3 activation, concomitant with increased production of proinflammatory cytokines, chemokines and type I interferon. In vivo this was associated with an increased number of alveolar macrophages and neutrophils in the lungs of IAV infected mice. Surprisingly, myeloid cell specific A20 knockout mice are protected against lethal IAV infection. These results challenge the general belief that an excessive host proinflammatory response is associated with IAV-induced lethality, and suggest that under certain conditions inhibition of A20 might be of interest in the management of IAV infections.


Subject(s)
DNA-Binding Proteins/deficiency , Influenza A virus/pathogenicity , Intracellular Signaling Peptides and Proteins/deficiency , Macrophages, Alveolar/enzymology , Neutrophils/enzymology , Ubiquitin-Protein Ligases/deficiency , Animals , Cysteine Endopeptidases , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation, Viral , Influenza A virus/physiology , Interferon Regulatory Factor-3/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lung/enzymology , Lung/immunology , Lung/virology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology , Mice , Mice, Knockout , NF-kappa B/biosynthesis , Neutrophils/immunology , Neutrophils/virology , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA, Messenger/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
9.
bioRxiv ; 2023 Aug 18.
Article in English | MEDLINE | ID: mdl-37645947

ABSTRACT

Various bacteria are suggested to contribute to colorectal cancer (CRC) development, including pks+ E. coli which produce the genotoxin colibactin that induces characteristic mutational signatures in host epithelial cells. It remains unclear how the highly unstable colibactin molecule is able to access host epithelial cells and its DNA to cause harm. Using the microbiota-dependent ZEB2-transgenic mouse model of invasive CRC, we found that pks+ E. coli drives CRC exacerbation and tissue invasion in a colibactin-dependent manner. Using isogenic mutant strains, we further demonstrate that CRC exacerbation critically depends on expression of the E. coli type-1 pilus adhesin FimH and the F9-pilus adhesin FmlH. Blocking bacterial adhesion using a pharmacological FimH inhibitor attenuates colibactin-mediated genotoxicity and CRC exacerbation. Together, we show that the oncogenic potential of pks+ E. coli critically depends on bacterial adhesion to host epithelial cells and is critically mediated by specific bacterial adhesins. Adhesin-mediated epithelial binding subsequently allows production of the genotoxin colibactin in close proximity to host epithelial cells, which promotes DNA damage and drives CRC development. These findings present promising therapeutic avenues for the development of anti-adhesive therapies aiming at mitigating colibactin-induced DNA damage and inhibiting the initiation and progression of CRC, particularly in individuals at risk for developing CRC.

10.
Cell Death Dis ; 14(8): 534, 2023 08 19.
Article in English | MEDLINE | ID: mdl-37598207

ABSTRACT

The intestinal epithelium is a single cell layer that is constantly renewed and acts as a physical barrier that separates intestinal microbiota from underlying tissues. In inflammatory bowel disease (IBD) in humans, as well as in experimental mouse models of IBD, this barrier is impaired, causing microbial infiltration and inflammation. Deficiency in OTU deubiquitinase with linear linkage specificity (OTULIN) causes OTULIN-related autoinflammatory syndrome (ORAS), a severe inflammatory pathology affecting multiple organs including the intestine. We show that mice with intestinal epithelial cell (IEC)-specific OTULIN deficiency exhibit increased susceptibility to experimental colitis and are highly sensitive to TNF toxicity, due to excessive apoptosis of OTULIN deficient IECs. OTULIN deficiency also increases intestinal pathology in mice genetically engineered to secrete excess TNF, confirming that chronic exposure to TNF promotes epithelial cell death and inflammation in OTULIN deficient mice. Mechanistically we demonstrate that upon TNF stimulation, OTULIN deficiency impairs TNF receptor complex I formation and LUBAC recruitment, and promotes the formation of the cytosolic complex II inducing epithelial cell death. Finally, we show that OTULIN deficiency in IECs increases susceptibility to Salmonella infection, further confirming the importance of OTULIN for intestinal barrier integrity. Together, these results identify OTULIN as a major anti-apoptotic protein in the intestinal epithelium and provide mechanistic insights into how OTULIN deficiency drives gastrointestinal inflammation in ORAS patients.


Subject(s)
Inflammatory Bowel Diseases , Intestinal Mucosa , Animals , Humans , Mice , Apoptosis , Cell Death , Inflammation
11.
J Immunol ; 185(12): 7646-53, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21068410

ABSTRACT

Apoptosis of oligodendrocytes (ODCs), the myelin-producing glial cells in the CNS, plays a central role in demyelinating diseases such as multiple sclerosis and experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. To investigate the mechanism behind ODC apoptosis in EAE, we made use of conditional knockout mice lacking the adaptor protein FADD specifically in ODCs (FADD(ODC-KO)). FADD mediates apoptosis by coupling death receptors with downstream caspase activation. In line with this, ODCs from FADD(ODC-KO) mice were completely resistant to death receptor-induced apoptosis in vitro. In the EAE model, FADD(ODC-KO) mice followed an ameliorated clinical disease course in comparison with control littermates. Lymphocyte and macrophage infiltration into the spinal cord parenchyma was significantly reduced, as was the extent of demyelination and proinflammatory gene expression. Collectively, our data show that FADD is critical for ODC apoptosis and the development of autoimmune demyelinating disease.


Subject(s)
Apoptosis/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Fas-Associated Death Domain Protein/immunology , Gene Deletion , Multiple Sclerosis/immunology , Oligodendroglia/immunology , Animals , Apoptosis/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Fas-Associated Death Domain Protein/biosynthesis , Fas-Associated Death Domain Protein/genetics , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Myelin Sheath/immunology , Myelin Sheath/metabolism , Myelin Sheath/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology
12.
Proc Natl Acad Sci U S A ; 106(29): 12195-200, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19587238

ABSTRACT

Binding of the peptide hormone vasopressin to its type-2 receptor (V2R) in kidney triggers a cAMP-mediated translocation of Aquaporin-2 water channels to the apical membrane, resulting in water reabsorption and thereby preventing dehydration. Mutations in the V2R gene lead to Nephrogenic Diabetes Insipidus (NDI), a disorder in which this process is disturbed, because the encoded, often intrinsically functional mutant V2 receptors are misfolded and retained in the endoplasmic reticulum (ER). Since plasma membrane expression is thought to be essential for V2R activation, cell permeable V2R antagonists have been used to induce maturation and rescue cell surface expression of V2R mutants, after which they need to be displaced by vasopressin for activation. Here, however, we show that 3 novel nonpeptide V2R agonists, but not vasopressin, activate NDI-causing V2R mutants at their intracellular location, without changing their maturation and at a sufficient level to induce the translocation of aquaporin-2 to the apical membrane. Moreover, in contrast to plasma membrane V2R, degradation of intracellular V2R mutants is not increased by their activation. Our data reveal that G protein-coupled receptors (GPCRs) normally active at the plasma membrane can be activated intracellularly and that intracellular activation does not induce their degradation; the data also indicate that nonpeptide agonists constitute highly promising therapeutics for diseases caused by misfolded GPCRs in general, and NDI in particular.


Subject(s)
Diabetes Insipidus, Nephrogenic/metabolism , Intracellular Space/metabolism , Mutant Proteins/metabolism , Peptides/pharmacology , Receptors, Vasopressin/agonists , Receptors, Vasopressin/metabolism , Animals , Aquaporin 2/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Polarity/drug effects , Deamino Arginine Vasopressin/pharmacology , Dogs , Humans , Intracellular Space/drug effects , Protein Stability/drug effects , Protein Transport/drug effects , Receptors, Vasopressin/ultrastructure , Recombinant Fusion Proteins/metabolism , Subcellular Fractions/drug effects
13.
Cell Death Dis ; 13(5): 476, 2022 05 19.
Article in English | MEDLINE | ID: mdl-35589698

ABSTRACT

The transcription factor nuclear factor-κB (NF-κB) has a key role in the pathogenesis of diabetes and its complications. Although activation of the canonical NF-κB pathway in ß-cells is generally deleterious, little is known about the role of the non-canonical NF-κB signalling and its main regulator, the NF-κB-inducing kinase (NIK), on pancreatic ß-cell survival and function. Previous studies based on models of NIK overexpression in pancreatic islet cells showed that NIK induced either spontaneous ß-cell death due to islet inflammation or glucose intolerance during diet-induced obesity (DIO) in mice. Therefore, NIK has been proposed as a potential target for diabetes therapy. However, no clear studies showed whether inhibition of NIK improves diabetes development. Here we show that genetic silencing of NIK in pancreatic ß-cells neither modifies diabetes incidence nor inflammatory responses in a mouse model of immune-mediated diabetes. Moreover, NIK silencing in DIO mice did not influence body weight gain, nor glucose metabolism. In vitro studies corroborated the in vivo findings in terms of ß-cell survival, function, and downstream gene regulation. Taken together, our data suggest that NIK activation is dispensable for the development of diabetes.


Subject(s)
Diabetes Mellitus , Insulin-Secreting Cells , Animals , Diabetes Mellitus/pathology , Insulin-Secreting Cells/metabolism , Mice , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction/physiology , NF-kappaB-Inducing Kinase
14.
Nat Commun ; 12(1): 5913, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34625556

ABSTRACT

OTULIN is a deubiquitinase that specifically cleaves linear ubiquitin chains. Here we demonstrate that the ablation of Otulin selectively in keratinocytes causes inflammatory skin lesions that develop into verrucous carcinomas. Genetic deletion of Tnfr1, knockin expression of kinase-inactive Ripk1 or keratinocyte-specific deletion of Fadd and Mlkl completely rescues mice with OTULIN deficiency from dermatitis and tumorigenesis, thereby identifying keratinocyte cell death as the driving force for inflammation. Single-cell RNA-sequencing comparing non-lesional and lesional skin reveals changes in epidermal stem cell identity in OTULIN-deficient keratinocytes prior to substantial immune cell infiltration. Keratinocytes lacking OTULIN display a type-1 interferon and IL-1ß response signature, and genetic or pharmacologic inhibition of these cytokines partially inhibits skin inflammation. Finally, expression of a hypomorphic mutant Otulin allele, previously shown to cause OTULIN-related autoinflammatory syndrome in humans, induces a similar inflammatory phenotype, thus supporting the importance of OTULIN for restraining skin inflammation and maintaining immune homeostasis.


Subject(s)
Endopeptidases/metabolism , Keratinocytes/metabolism , Skin/metabolism , Animals , Cell Death/genetics , Cytokines/metabolism , Endopeptidases/genetics , Fas-Associated Death Domain Protein , Gene Knock-In Techniques , Homeostasis , Inflammation/pathology , Interferon Type I , Interleukin-1beta , Mice , Necroptosis , Peptide Fragments , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Skin/pathology , Stem Cells/metabolism , Systems Analysis , Ubiquitin/metabolism
15.
Cell Rep ; 36(12): 109748, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34551300

ABSTRACT

Obesity-induced inflammation is a major driving force in the development of insulin resistance, type 2 diabetes (T2D), and related metabolic disorders. During obesity, macrophages accumulate in the visceral adipose tissue, creating a low-grade inflammatory environment. Nuclear factor κB (NF-κB) signaling is a central coordinator of inflammatory responses and is tightly regulated by the anti-inflammatory protein A20. Here, we find that myeloid-specific A20-deficient mice are protected from diet-induced obesity and insulin resistance despite an inflammatory environment in their metabolic tissues. Macrophages lacking A20 show impaired mitochondrial respiratory function and metabolize more palmitate both in vitro and in vivo. We hypothesize that A20-deficient macrophages rely more on palmitate oxidation and metabolize the fat present in the diet, resulting in a lean phenotype and protection from metabolic disease. These findings reveal a role for A20 in regulating macrophage immunometabolism.


Subject(s)
Fatty Acids/metabolism , Obesity/pathology , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Adipose Tissue, White/metabolism , Animals , Cytokines/genetics , Cytokines/metabolism , Diet, High-Fat , Disease Models, Animal , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Insulin Resistance , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Obesity/metabolism , Oxygen Consumption , Palmitates/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/deficiency , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism
16.
Cell Rep ; 30(7): 2237-2247.e6, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32075762

ABSTRACT

Inflammatory signaling pathways are tightly regulated to avoid chronic inflammation and the development of disease. OTULIN is a deubiquitinating enzyme that controls inflammation by cleaving linear ubiquitin chains generated by the linear ubiquitin chain assembly complex. Here, we show that ablation of OTULIN in liver parenchymal cells in mice causes severe liver disease which is characterized by liver inflammation, hepatocyte apoptosis, and compensatory hepatocyte proliferation, leading to steatohepatitis, fibrosis, and hepatocellular carcinoma (HCC). Genetic ablation of Fas-associated death domain (FADD) completely rescues and knockin expression of kinase inactive receptor-interacting protein kinase 1 (RIPK1) significantly protects mice from developing liver disease, demonstrating that apoptosis of OTULIN-deficient hepatocytes triggers disease pathogenesis in this model. Finally, we demonstrate that type I interferons contribute to disease in hepatocyte-specific OTULIN-deficient mice. Our study reveals the critical importance of OTULIN in protecting hepatocytes from death, thereby preventing the development of chronic liver inflammation and HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Endopeptidases/metabolism , Fas-Associated Death Domain Protein/antagonists & inhibitors , Hepatitis/metabolism , Liver Neoplasms/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/physiology , CHO Cells , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cricetulus , Fas-Associated Death Domain Protein/metabolism , Hepatitis/genetics , Hepatitis/pathology , Humans , Interferon Type I/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction
17.
Nat Cancer ; 1(6): 620-634, 2020 06.
Article in English | MEDLINE | ID: mdl-35121975

ABSTRACT

Colorectal cancer (CRC) is highly prevalent in Western society, and increasing evidence indicates strong contributions of environmental factors and the intestinal microbiota to CRC initiation, progression and even metastasis. We have identified a synergistic inflammatory tumor-promoting mechanism through which the resident intestinal microbiota boosts invasive CRC development in an epithelial-to-mesenchymal transition-prone tissue environment. Intestinal epithelial cell (IEC)-specific transgenic expression of the epithelial-to-mesenchymal transition regulator Zeb2 in mice (Zeb2IEC-Tg/+) leads to increased intestinal permeability, myeloid cell-driven inflammation and spontaneous invasive CRC development. Zeb2IEC-Tg/+ mice develop a dysplastic colonic epithelium, which progresses to severely inflamed neoplastic lesions while the small intestinal epithelium remains normal. Zeb2IEC-Tg/+ mice are characterized by intestinal dysbiosis, and microbiota depletion with broad-spectrum antibiotics or germ-free rederivation completely prevents cancer development. Zeb2IEC-Tg/+ mice represent the first mouse model of spontaneous microbiota-dependent invasive CRC and will help us to better understand host-microbiome interactions driving CRC development in humans.


Subject(s)
Carcinoma , Microbiota , Animals , Carcinoma/metabolism , Colon/metabolism , Mice
18.
Nat Commun ; 10(1): 1834, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015422

ABSTRACT

Prevention of inflammatory bowel disease (IBD) relies on tight control of inflammatory, cell death and autophagic mechanisms, but how these pathways are integrated at the molecular level is still unclear. Here we show that the anti-inflammatory protein A20 and the critical autophagic mediator Atg16l1 physically interact and synergize to regulate the stability of the intestinal epithelial barrier. A proteomic screen using the WD40 domain of ATG16L1 (WDD) identified A20 as a WDD-interacting protein. Loss of A20 and Atg16l1 in mouse intestinal epithelium induces spontaneous IBD-like pathology, as characterized by severe inflammation and increased intestinal epithelial cell death in both small and large intestine. Mechanistically, absence of A20 promotes Atg16l1 accumulation, while elimination of Atg16l1 or expression of WDD-deficient Atg16l1 stabilizes A20. Collectively our data show that A20 and Atg16l1 cooperatively control intestinal homeostasis by acting at the intersection of inflammatory, autophagy and cell death pathways.


Subject(s)
Carrier Proteins/metabolism , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , WD40 Repeats/genetics , Animals , Autophagy/immunology , Autophagy-Related Proteins , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Line, Tumor , Disease Models, Animal , Endoscopy , Female , Homeostasis/immunology , Humans , Inflammatory Bowel Diseases/diagnostic imaging , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/cytology , Intestinal Mucosa/diagnostic imaging , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/immunology , Proteomics , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/immunology , WD40 Repeats/immunology
19.
Cell Rep ; 29(9): 2689-2701.e4, 2019 Nov 26.
Article in English | MEDLINE | ID: mdl-31775038

ABSTRACT

Regenerative responses predispose tissues to tumor formation by largely unknown mechanisms. High-mobility group box 1 (HMGB1) is a danger-associated molecular pattern contributing to inflammatory pathologies. We show that HMGB1 derived from keratinocytes, but not myeloid cells, delays cutaneous wound healing and drives tumor formation. In wounds of mice lacking HMGB1 selectively in keratinocytes, a marked reduction in neutrophil extracellular trap (NET) formation is observed. Pharmacological targeting of HMGB1 or NETs prevents skin tumorigenesis and accelerates wound regeneration. HMGB1-dependent NET formation and skin tumorigenesis is orchestrated by tumor necrosis factor (TNF) and requires RIPK1 kinase activity. NETs are present in the microenvironment of keratinocyte-derived tumors in mice and lesional and tumor skin of patients suffering from recessive dystrophic epidermolysis bullosa, a disease in which skin blistering predisposes to tumorigenesis. We conclude that tumorigenicity of the wound microenvironment depends on epithelial-derived HMGB1 regulating NET formation, thereby establishing a mechanism linking reparative inflammation to tumor initiation.


Subject(s)
Extracellular Traps/metabolism , Neutrophils/metabolism , Skin/pathology , HMGB1 Protein/metabolism , Humans , Tumor Microenvironment , Wound Healing
20.
Nat Commun ; 9(1): 2036, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29789522

ABSTRACT

Microglia, the mononuclear phagocytes of the central nervous system (CNS), are important for the maintenance of CNS homeostasis, but also critically contribute to CNS pathology. Here we demonstrate that the nuclear factor kappa B (NF-κB) regulatory protein A20 is crucial in regulating microglia activation during CNS homeostasis and pathology. In mice, deletion of A20 in microglia increases microglial cell number and affects microglial regulation of neuronal synaptic function. Administration of a sublethal dose of lipopolysaccharide induces massive microglia activation, neuroinflammation, and lethality in mice with microglia-confined A20 deficiency. Microglia A20 deficiency also exacerbates multiple sclerosis (MS)-like disease, due to hyperactivation of the Nlrp3 inflammasome leading to enhanced interleukin-1ß secretion and CNS inflammation. Finally, we confirm a Nlrp3 inflammasome signature and IL-1ß expression in brain and cerebrospinal fluid from MS patients. Collectively, these data reveal a critical role for A20 in the control of microglia activation and neuroinflammation.


Subject(s)
Inflammasomes/immunology , Microglia/immunology , Multiple Sclerosis/immunology , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Adult , Aged , Aged, 80 and over , Animals , Brain/immunology , Brain/pathology , Disease Models, Animal , Female , Humans , Interleukin-1beta/metabolism , Lipopolysaccharides/immunology , Male , Mice , Microglia/pathology , Middle Aged , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Signal Transduction/immunology , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/immunology
SELECTION OF CITATIONS
SEARCH DETAIL