Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cell ; 183(3): 636-649.e18, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33031745

ABSTRACT

Cytoplasmic accumulation of TDP-43 is a disease hallmark for many cases of amyotrophic lateral sclerosis (ALS), associated with a neuroinflammatory cytokine profile related to upregulation of nuclear factor κB (NF-κB) and type I interferon (IFN) pathways. Here we show that this inflammation is driven by the cytoplasmic DNA sensor cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS) when TDP-43 invades mitochondria and releases DNA via the permeability transition pore. Pharmacologic inhibition or genetic deletion of cGAS and its downstream signaling partner STING prevents upregulation of NF-κB and type I IFN induced by TDP-43 in induced pluripotent stem cell (iPSC)-derived motor neurons and in TDP-43 mutant mice. Finally, we document elevated levels of the specific cGAS signaling metabolite cGAMP in spinal cord samples from patients, which may be a biomarker of mtDNA release and cGAS/STING activation in ALS. Our results identify mtDNA release and cGAS/STING activation as critical determinants of TDP-43-associated pathology and demonstrate the potential for targeting this pathway in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , Membrane Proteins/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Nucleotidyltransferases/metabolism , Alarmins/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cytoplasm/metabolism , Disease Models, Animal , Disease Progression , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Inflammation/metabolism , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , NF-kappa B/metabolism , Nerve Degeneration/pathology , Phosphotransferases (Alcohol Group Acceptor) , Protein Subunits/metabolism , Signal Transduction
2.
J Clin Immunol ; 42(2): 325-335, 2022 02.
Article in English | MEDLINE | ID: mdl-34783940

ABSTRACT

PURPOSE: NLRC4-associated autoinflammatory disease (NLRC4-AID) is an autosomal dominant condition presenting with a range of clinical manifestations which can include macrophage activation syndrome (MAS) and severe enterocolitis. We now report the first homozygous mutation in NLRC4 (c.478G > A, p.A160T) causing autoinflammatory disease with immune dysregulation and find that heterozygous carriers in the general population are at increased risk of developing ulcerative colitis. METHODS: Circulating immune cells and inflammatory markers were profiled and historical clinical data interrogated. DNA was extracted and sequenced using standard procedures. Inflammasome activation assays for ASC speck formation, pyroptosis, and IL-1ß/IL-18 secretion confirmed pathogenicity of the mutation in vitro. Genome-wide association of NLRC4 (A160T) with ulcerative colitis was examined using data from the IBD exomes portal. RESULTS: A 60-year-old Brazilian female patient was evaluated for recurrent episodes of systemic inflammation from six months of age. Episodes were characterized by recurrent low-grade fever, chills, oral ulceration, uveitis, arthralgia, and abdominal pain, followed by diarrhea with mucus and variable skin rash. High doses of corticosteroids were somewhat effective in controlling disease and anti-IL-1ß therapy partially controlled symptoms. While on treatment, serum IL-1ß and IL-18 levels remained elevated. Genetic investigations identified a homozygous mutation in NLRC4 (A160T), inherited in a recessive fashion. Increased ASC speck formation and IL-1ß/IL-18 secretion confirmed pathogenicity when NLRC4 (A160T) was analyzed in human cell lines. This allele is significantly enriched in patients with ulcerative colitis: OR 2.546 (95% 1.778-3.644), P = 0.01305. CONCLUSION: NLRC4 (A160T) can either cause recessively inherited autoinflammation and immune dysregulation, or function as a heterozygous risk factor for the development of ulcerative colitis.


Subject(s)
Colitis, Ulcerative , Hereditary Autoinflammatory Diseases , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/genetics , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/genetics , Female , Genome-Wide Association Study , Humans , Inflammasomes/metabolism , Middle Aged
3.
J Allergy Clin Immunol ; 147(6): 2134-2145.e20, 2021 06.
Article in English | MEDLINE | ID: mdl-33378691

ABSTRACT

BACKGROUND: NLRP1 is an innate immune sensor that can form cytoplasmic inflammasome complexes. Polymorphisms in NLRP1 are linked to asthma; however, there is currently no functional or mechanistic explanation for this. OBJECTIVE: We sought to clarify the role of NLRP1 in asthma pathogenesis. METHODS: Results from the GALA II cohort study were used to identify a link between NLRP1 and asthma in Mexican Americans. In vitro and in vivo models for NLRP1 activation were applied to investigate the role of this inflammasome in asthma at the molecular level. RESULTS: We document the association of an NLRP1 haplotype with asthma for which the single nucleotide polymorphism rs11651270 (M1184V) individually is the most significant. Surprisingly, M1184V increases NLRP1 activation in the context of N-terminal destabilization, but decreases NLRP1 activation on dipeptidyl peptidase 9 inhibition. In vitro studies demonstrate that M1184V increases binding to dipeptidyl peptidase 9, which can account for its inhibitory role in this context. In addition, in vivo data from a mouse model of airway inflammation reveal a protective role for NLRP1 inflammasome activation reducing eosinophilia in this setting. CONCLUSIONS: Linking our in vitro and in vivo results, we found that the NLRP1 variant M1184V reduces inflammasome activation in the context of dipeptidyl peptidase 9 inhibition and could thereby increase asthma severity. Our studies may have implications for the treatment of asthma in patients carrying this variant of NLRP1.


Subject(s)
Alleles , Asthma/etiology , Asthma/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Inflammasomes/metabolism , Mutation , NLR Proteins/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Asthma/diagnosis , Cell Line , Disease Models, Animal , Disease Susceptibility , Eosinophils/immunology , Eosinophils/metabolism , Eosinophils/pathology , Genetic Predisposition to Disease , Humans , Mice , Mice, Knockout , NLR Proteins/chemistry , NLR Proteins/metabolism , Polymorphism, Single Nucleotide , Structure-Activity Relationship , Trauma Severity Indices
4.
Curr Rheumatol Rep ; 20(7): 38, 2018 05 30.
Article in English | MEDLINE | ID: mdl-29846818

ABSTRACT

PURPOSE OF REVIEW: Type I interferons (IFNαß) induce the expression of hundreds of genes; thus, it is unsurprising that the initiation, transmission, and resolution of the IFNαß-mediated immune response is tightly controlled. Mutations that alter nucleic acid processing and recognition, ablate IFNαß-specific negative feedback mechanisms, or result in dysfunction of the proteasome system can all induce pathogenic IFNαß signalling and are the focus of this review. RECENT FINDINGS: Recent advances have delineated the precise cytoplasmic mechanisms that facilitate self-DNA to be recognised by cGAS and self-RNA to be recognised by RIG-I or MDA-5. This helps clarify interferonopathies associated with mutations in genes which code for DNase-II and ADAR1, among others. Similarly, loss of function mutations in Pol α, which lowers the presence of antagonistic ligands in the cytosol, or gain of function mutations in RIG-I and MDA-5, result in increased propensity for receptor activation and therefore IFNαß induction. As the aetiology of monogenic autoinflammatory diseases are uncovered, novel and sometimes unsuspected molecular interactions and signalling pathways are being defined. This review covers developments that have come to light over the past 3 years, with reference to the study of interferonopathies.


Subject(s)
Autoimmune Diseases/immunology , Hereditary Autoinflammatory Diseases/immunology , Interferon Type I/immunology , Autoimmune Diseases/genetics , Hereditary Autoinflammatory Diseases/genetics , Humans , Mutation , Signal Transduction/genetics , Signal Transduction/immunology
5.
Curr Rheumatol Rep ; 20(7): 40, 2018 05 30.
Article in English | MEDLINE | ID: mdl-29846819

ABSTRACT

PURPOSE OF REVIEW: Autoinflammatory diseases are driven by abnormal innate immune activation. In the case of inflammasomopathies, these are all attributable to activation of an inflammasome complex, nucleated by an innate immune sensor such as NLRP3. This review will focus on recent advances that have helped to elucidate the role of three other sensors (NLRP1, NLRC4 and pyrin) which can also cause inflammasomopathies. RECENT FINDINGS: Mutations in pyrin (S242R or E244K) destroy an inhibitory 14-3-3 binding site and result in the newly characterised disease pyrin-associated autoinflammation with neutrophilic dermatosis (PAAND). Moreover, a separate autoinflammatory disease driven by mevalonate kinase deficiency leads to defective RhoGTPase prenylation and subsequent loss of pyrin S242R phosphorylation, suggesting a shared mechanism of disease. Other inflammasomes such as NLRP1 and NLRC4 have had novel mutations described recently, which inform about the specific domains required for activation and autoinhibition. This review covers recent advances in the study of inflammasomopathies, focussing on gene discoveries that elucidate new pathogenic mechanisms.


Subject(s)
Hereditary Autoinflammatory Diseases/genetics , Inflammasomes/genetics , Mutation , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , CARD Signaling Adaptor Proteins/genetics , Calcium-Binding Proteins/genetics , Hereditary Autoinflammatory Diseases/immunology , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Inflammasomes/immunology , NLR Proteins , Pyrin/genetics
6.
Curr Rheumatol Rep ; 20(7): 39, 2018 05 30.
Article in English | MEDLINE | ID: mdl-29846841

ABSTRACT

PURPOSE OF REVIEW: The nuclear factor κB (NF-κB) pathway is tightly regulated through multiple posttranslational mechanisms including ubiquitination. Mutations in these regulatory pathways can cause disease and are the focus of this review. RECENT FINDINGS: The linear ubiquitin chain assembly complex (LUBAC) is a trimer made up of HOIL-1L, SHARPIN, and the catalytic subunit HOIP. Loss of function mutations in HOIL-1L and HOIP result in largely overlapping phenotypes, characterized by multi-organ autoinflammation, immunodeficiency, and amylopectinosis. Interestingly, patient fibroblasts exhibited diminished IL-1ß- and TNF-induced NF-κB activation, yet monocytes were hyper-responsive to IL-1ß, hinting at cell type or target specific roles of LUBAC-mediated ubiquitination. Ubiquitin-driven signaling is counterbalanced by deubiquitinase enzymes (DUBs), such as OTULIN and A20. Hypomorphic mutations in OTULIN result in elevated NF-κB signaling causing an autoinflammatory syndrome. Similarly, patients with high-penetrance heterozygous mutations in the gene encoding A20 (haploinsufficiency of A20 (HA20)) display excessive ubiquitination and increased activity of NF-κB and of NLRP3 inflammasome activation. HA20 patients present with Behçet-like characteristics or an autoimmune lymphoproliferative syndrome (ALPS)-like phenotype, indicating diverse protein functions. This review summarizes recent discoveries in the field of NF-kB-related autoinflammatory diseases (relopathies) within the past 3 years and points to several questions that still remain unanswered.


Subject(s)
Hereditary Autoinflammatory Diseases/genetics , Endopeptidases/genetics , Endopeptidases/physiology , Hereditary Autoinflammatory Diseases/metabolism , Humans , Mutation , NF-kappa B/metabolism , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/physiology , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/physiology
7.
EMBO Rep ; 16(9): 1203-18, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26265006

ABSTRACT

Streptococcus pneumoniae coinfection is a major cause of influenza-associated mortality; however, the mechanisms underlying pathogenesis or protection remain unclear. Using a clinically relevant mouse model, we identify immune-mediated damage early during coinfection as a new mechanism causing susceptibility. Coinfected CCR2(-/-) mice lacking monocytes and monocyte-derived cells control bacterial invasion better, show reduced epithelial damage and are overall more resistant than wild-type controls. In influenza-infected wild-type lungs, monocytes and monocyte-derived cells are the major cell populations expressing the apoptosis-inducing ligand TRAIL. Accordingly, anti-TRAIL treatment reduces bacterial load and protects against coinfection if administered during viral infection, but not following bacterial exposure. Post-influenza bacterial outgrowth induces a strong proinflammatory cytokine response and massive inflammatory cell infiltrate. Depletion of neutrophils or blockade of TNF-α facilitate bacterial outgrowth, leading to increased mortality, demonstrating that these factors aid bacterial control. We conclude that inflammatory monocytes recruited early, during the viral phase of coinfection, induce TRAIL-mediated lung damage, which facilitates bacterial invasion, while TNF-α and neutrophil responses help control subsequent bacterial outgrowth. We thus identify novel determinants of protection versus pathology in influenza-Streptococcus pneumoniae coinfection.


Subject(s)
Coinfection/immunology , Influenza A Virus, H3N2 Subtype , Monocytes/immunology , Orthomyxoviridae Infections/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/pathogenicity , Animals , Coinfection/physiopathology , Cytokines/immunology , Disease Models, Animal , Disease Susceptibility , Humans , Lung/immunology , Lung/microbiology , Lung/physiopathology , Lung/virology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Orthomyxoviridae Infections/complications , Pneumococcal Infections/complications , Receptors, CCR2/deficiency , Receptors, CCR2/genetics , Streptococcus pneumoniae/growth & development , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor-alpha/immunology
8.
J Immunol ; 193(2): 700-7, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24907343

ABSTRACT

Themis1 is a protein implicated in transducing signals from the TCR. Mice deficient in Themis1 show a strong impairment in T cell selection in the thymus and defective T cell activation. The related Themis2 protein is expressed in B cells where it associates with signaling proteins Grb2 and Vav1, and is tyrosine phosphorylated after BCR stimulation. Thus, it has been proposed that Themis2 may transduce BCR signals, and hence play important roles in B cell development and activation. In this article, we show that Themis2 is expressed in all developing subsets of B cells, in mature follicular and marginal zone B cells, and in activated B cells, including germinal center B cells and plasma cells. In contrast, B lineage cells express no other Themis-family genes. Activation of B cells leads to reduced Themis2 expression, although it remains the only Themis-family protein expressed. To analyze the physiological function of Themis2, we generated a Themis2-deficient mouse strain. Surprisingly, we found that Themis2 is not required for B cell development, for activation, or for Ab responses either to model Ags or to influenza viral infection.


Subject(s)
Antibody Formation/immunology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Intracellular Signaling Peptides and Proteins/immunology , Lymphocyte Activation/immunology , Animals , Antibody Formation/genetics , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B-Lymphocytes/metabolism , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Lineage/immunology , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Female , Flow Cytometry , Gene Expression/immunology , Immunoblotting , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spleen/immunology , Spleen/metabolism
9.
J Immunol ; 192(6): 2677-88, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24534532

ABSTRACT

NK cells contribute to antitumor and antiviral immunosurveillance. Their development in the bone marrow (BM) requires the transcription factor E4BP4/NFIL3, but requirements in other organs are less well defined. In this study, we show that CD3(-)NK1.1(+)NKp46(+)CD122(+) NK cells of immature phenotype and expressing low eomesodermin levels are found in thymus, spleen, and liver of E4BP4-deficient mice, whereas numbers of mature, eomesodermin(high) conventional NK cells are drastically reduced. E4BP4-deficient CD44(+)CD25(-) double-negative 1 thymocytes efficiently develop in vitro into NK cells with kinetics, phenotype, and functionality similar to wild-type controls, whereas no NK cells develop from E4BP4-deficient BM precursors. In E4BP4/Rag-1 double-deficient (DKO) mice, NK cells resembling those in Rag-1-deficient controls are found in similar numbers in the thymus and liver. However, NK precursors are reduced in DKO BM, and no NK cells develop from DKO BM progenitors in vitro. DKO thymocyte precursors readily develop into NK cells, but DKO BM transfers into nude recipients and NK cells in E4BP4/Rag-1/IL-7 triple-KO mice indicated thymus-independent NK cell development. In the presence of T cells or E4BP4-sufficient NK cells, DKO NK cells have a selective disadvantage, and thymic and hepatic DKO NK cells show reduced survival when adoptively transferred into lymphopenic hosts. This correlates with higher apoptosis rates and lower responsiveness to IL-15 in vitro. In conclusion, we demonstrate E4BP4-independent development of NK cells of immature phenotype, reduced fitness, short t1/2, and potential extramedullary origin. Our data identify E4BP4-independent NK cell developmental pathways and a role for E4BP4 in NK cell homeostasis.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Killer Cells, Natural/immunology , Liver/immunology , Spleen/immunology , Thymus Gland/immunology , Animals , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Cells, Cultured , Flow Cytometry , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Homeodomain Proteins/metabolism , Interleukin-7/deficiency , Interleukin-7/genetics , Interleukin-7/immunology , Killer Cells, Natural/metabolism , Liver/cytology , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Signal Transduction/immunology , Spleen/cytology , Spleen/metabolism , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
10.
PLoS Pathog ; 9(11): e1003773, 2013.
Article in English | MEDLINE | ID: mdl-24278020

ABSTRACT

Interferons (IFNs) are a group of cytokines with a well-established antiviral function. They can be induced by viral infection, are secreted and bind to specific receptors on the same or neighbouring cells to activate the expression of hundreds of IFN stimulated genes (ISGs) with antiviral function. Type I IFN has been known for more than half a century. However, more recently, type III IFN (IFNλ, IL-28/29) was shown to play a similar role and to be particularly important at epithelial surfaces. Here we show that airway epithelia, the primary target of influenza A virus, produce both IFN I and III upon infection, and that induction of both depends on the RIG-I/MAVS pathway. While IRF3 is generally regarded as the transcription factor required for initiation of IFN transcription and the so-called "priming loop", we find that IRF3 deficiency has little impact on IFN expression. In contrast, lack of IRF7 reduced IFN production significantly, and only IRF3(-/-)IRF7(-/-) double deficiency completely abolished it. The transcriptional response to influenza infection was largely dependent on IFNs, as it was reduced to a few upregulated genes in epithelia lacking receptors for both type I and III IFN (IFNAR1(-/-)IL-28Rα(-/-)). Wild-type epithelia and epithelia deficient in either the type I IFN receptor or the type III IFN receptor exhibit similar transcriptional profiles in response to virus, indicating that none of the induced genes depends selectively on only one IFN system. In chimeric mice, the lack of both IFN I and III signalling in the stromal compartment alone significantly increased the susceptibility to influenza infection. In conclusion, virus infection of airway epithelia induces, via a RIG-I/MAVS/IRF7 dependent pathway, both type I and III IFNs which drive two completely overlapping and redundant amplification loops to upregulate ISGs and protect from influenza infection.


Subject(s)
Epithelial Cells/metabolism , Influenza A virus/metabolism , Interferon Type I/metabolism , Interleukins/metabolism , Orthomyxoviridae Infections/metabolism , Respiratory Mucosa/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Epithelial Cells/immunology , Epithelial Cells/pathology , Epithelial Cells/virology , Influenza A virus/genetics , Influenza A virus/immunology , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Regulatory Factor-7/metabolism , Interferon Type I/genetics , Interferon Type I/immunology , Interleukins/genetics , Interleukins/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Receptors, Cell Surface , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Respiratory Mucosa/virology
11.
J Allergy Clin Immunol ; 131(5): 1331-9.e10, 2013 May.
Article in English | MEDLINE | ID: mdl-23561801

ABSTRACT

BACKGROUND: Respiratory tract viruses are a major environmental risk factor for both the inception and exacerbations of asthma. Genetic defects in Toll-like receptor (TLR) 7-mediated signaling, impaired type I interferon responses, or both have been reported in asthmatic patients, although their contribution to the onset and exacerbation of asthma remains poorly understood. OBJECTIVE: We sought to determine whether Pneumovirus infection in the absence of TLR7 predisposes to bronchiolitis and the inception of asthma. METHODS: Wild-type and TLR7-deficient (TLR7(-/-)) mice were inoculated with the rodent-specific pathogen pneumonia virus of mice at 1 (primary), 7 (secondary), and 13 (tertiary) weeks of age, and pathologic features of bronchiolitis or asthma were assessed. In some experiments infected mice were exposed to low-dose cockroach antigen. RESULTS: TLR7 deficiency increased viral load in the airway epithelium, which became sloughed and necrotic, and promoted an IFN-α/ß(low), IL-12p70(low), IL-1ß(high), IL-25(high), and IL-33(high) cytokine microenvironment that was associated with the recruitment of type 2 innate lymphoid cells/nuocytes and increased TH2-type cytokine production. Viral challenge of TLR7(-/-) mice induced all of the cardinal pathophysiologic features of asthma, including tissue eosinophilia, mast cell hyperplasia, IgE production, airway smooth muscle alterations, and airways hyperreactivity in a memory CD4(+) T cell-dependent manner. Importantly, infections with pneumonia virus of mice promoted allergic sensitization to inhaled cockroach antigen in the absence but not the presence of TLR7. CONCLUSION: TLR7 gene defects and Pneumovirus infection interact to establish an aberrant adaptive response that might underlie virus-induced asthma exacerbations in later life.


Subject(s)
Asthma/immunology , Asthma/pathology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Murine pneumonia virus , Pneumovirus Infections/complications , Toll-Like Receptor 7/deficiency , Toll-Like Receptor 7/genetics , Animals , Animals, Newborn , Asthma/etiology , Disease Models, Animal , Lung/pathology , Lung/physiopathology , Lung/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Murine pneumonia virus/pathogenicity , Pneumovirus Infections/immunology , Pneumovirus Infections/pathology , Viral Load
12.
J Exp Med ; 221(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38630025

ABSTRACT

OTU deubiquitinase with linear linkage specificity (OTULIN) regulates inflammation and cell death by deubiquitinating linear ubiquitin chains generated by the linear ubiquitin chain assembly complex (LUBAC). Biallelic loss-of-function mutations causes OTULIN-related autoinflammatory syndrome (ORAS), while OTULIN haploinsuffiency has not been associated with spontaneous inflammation. However, herein, we identify two patients with the heterozygous mutation p.Cys129Ser in OTULIN. Consistent with ORAS, we observed accumulation of linear ubiquitin chains, increased sensitivity to TNF-induced death, and dysregulation of inflammatory signaling in patient cells. While the C129S mutation did not affect OTULIN protein stability or binding capacity to LUBAC and linear ubiquitin chains, it did ablate OTULIN deubiquitinase activity. Loss of activity facilitated the accumulation of autoubiquitin chains on LUBAC. Altered ubiquitination of LUBAC inhibits its recruitment to the TNF receptor signaling complex, promoting TNF-induced cell death and disease pathology. By reporting the first dominant negative mutation driving ORAS, this study expands our clinical understanding of OTULIN-associated pathology.


Subject(s)
Inflammation , Ubiquitin , Humans , Cell Death , Cell Membrane , Deubiquitinating Enzymes , Inflammation/genetics , Syndrome , Ubiquitin-Protein Ligase Complexes
13.
Am J Respir Cell Mol Biol ; 49(3): 481-90, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23614768

ABSTRACT

Allergic asthma is a chronic inflammatory disease predominately associated with the activation of CD4(+) T helper Type 2 (Th2) cells. Innate pattern recognition receptors are widely acknowledged to shape the adaptive immune response. For example, the activation of airway epithelial Toll-like receptor-4 (TLR4) is necessary for the generation of house dust mite (HDM)-specific Th2 responses and the development of asthma in mice. Here we sought to determine whether the absence of Toll-interleukin-1 receptor (TIR)-8, a negative regulator of TLR4 signaling that is highly expressed in airway epithelial cells, would exacerbate HDM-induced asthma in a murine model. We found that Th2 but not Th1 or Th17 cytokine expression was significantly reduced in the lung and draining lymph nodes in HDM-sensitized/challenged TIR8 gene-deleted mice. Mucus-producing goblet cells, HDM-specific IgG1, and airway hyperreactivity were also significantly reduced in HDM-exposed, TIR8-deficient mice. Consistent with the attenuated Th2 response, eotaxin-2/CCL24 expression and airway and peribronchial eosinophils were significantly reduced in the absence of TIR8. In contrast, IL-17A-responsive chemokines and neutrophil numbers were unaffected. Similar findings were obtained for cockroach allergen. HDM sensitization alone up-regulated the expression of IL-1F5, a putative TIR8 ligand and inducer of IL-4. Of note, innate IL-4, IL-5, IL-13, and IL-33 cytokine expression was reduced during HDM sensitization in the absence of TIR8, as was the recruitment of conventional dendritic cells and basophils to the draining lymph nodes. Our findings suggest that TIR8 enhances the development of HDM-induced innate and adaptive Th2, but not Th1 or Th17 type immunity.


Subject(s)
Allergens/immunology , Antigens, Dermatophagoides/immunology , Insect Proteins/immunology , Receptors, Interleukin-1/immunology , Respiratory Hypersensitivity/immunology , Adaptive Immunity , Allergens/administration & dosage , Animals , Antigens, Dermatophagoides/administration & dosage , Chemokine CCL24/genetics , Chemokine CCL24/immunology , Eosinophils/immunology , Gene Expression Regulation/immunology , Immunity, Innate , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Insect Proteins/administration & dosage , Interleukin-1/genetics , Interleukin-1/immunology , Lung/immunology , Lymph Nodes/immunology , Male , Mice , Mice, Knockout , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/pathology , Severity of Illness Index , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology
14.
J Immunol ; 186(10): 5938-48, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21482736

ABSTRACT

Human respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection in infants. In human infants, plasmacytoid dendritic cells (pDC) are recruited to the nasal compartment during infection and initiate host defense through the secretion of type I IFN, IL-12, and IL-6. However, RSV-infected pDC are refractory to TLR7-mediated activation. In this study, we used the rodent-specific pathogen, pneumonia virus of mice (PVM), to determine the contribution of pDC and TLR7 signaling to the development of the innate inflammatory and early adaptive immune response. In wild-type, but not TLR7- or MyD88-deficient mice, PVM inoculation led to a marked infiltration of pDC and increased expression of type I, II, and III IFNs. The delayed induction of IFNs in the absence of TLR7 or MyD88 was associated with a diminished innate inflammatory response and augmented virus recovery from lung tissue. In the absence of TLR7, PVM-specific CD8(+) T cell cytokine production was abrogated. The adoptive transfer of TLR7-sufficient, but not TLR7-deficient pDC to TLR7 gene-deleted mice recapitulated the antiviral responses observed in wild-type mice and promoted virus clearance. In summary, TLR7-mediated signaling by pDC is required for appropriate innate responses to acute pneumovirus infection. It is conceivable that as-yet-unidentified defects in the TLR7 signaling pathway may be associated with elevated levels of RSV-associated morbidity and mortality among otherwise healthy human infants.


Subject(s)
Dendritic Cells/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Murine pneumonia virus/immunology , Myeloid Differentiation Factor 88/metabolism , Pneumovirus Infections/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Adaptive Immunity , Adoptive Transfer , Animals , Interferons/genetics , Interferons/immunology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Signal Transduction , Toll-Like Receptor 7/genetics
15.
Cell Death Dis ; 14(2): 123, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36792599

ABSTRACT

Necroptosis is a lytic and inflammatory form of cell death that is highly constrained to mitigate detrimental collateral tissue damage and impaired immunity. These constraints make it difficult to define the relevance of necroptosis in diseases such as chronic and persistent viral infections and within individual organ systems. The role of necroptotic signalling is further complicated because proteins essential to this pathway, such as receptor interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like (MLKL), have been implicated in roles outside of necroptotic signalling. We sought to address this issue by individually defining the role of RIPK3 and MLKL in chronic lymphocytic choriomeningitis virus (LCMV) infection. We investigated if necroptosis contributes to the death of LCMV-specific CD8+ T cells or virally infected target cells during infection. We provide evidence showing that necroptosis was redundant in the pathogenesis of acute forms of LCMV (Armstrong strain) and the early stages of chronic (Docile strain) LCMV infection in vivo. The number of immune cells, their specificity and reactivity towards viral antigens and viral loads are not altered in the absence of either MLKL or RIPK3 during acute and during the early stages of chronic LCMV infection. However, we identified that RIPK3 promotes immune dysfunction and prevents control of infection at later stages of chronic LCMV disease. This was not phenocopied by the loss of MLKL indicating that the phenotype was driven by a necroptosis-independent function of RIPK3. We provide evidence that RIPK3 signaling evoked a dysregulated type 1 interferone response which we linked to an impaired antiviral immune response and abrogated clearance of chronic LCMV infection.


Subject(s)
Lymphocytic choriomeningitis virus , Protein Kinases , Protein Kinases/genetics , Protein Kinases/metabolism , Lymphocytic choriomeningitis virus/metabolism , Necroptosis , CD8-Positive T-Lymphocytes/metabolism , Cell Death , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
16.
Nat Rev Immunol ; 22(9): 535-549, 2022 09.
Article in English | MEDLINE | ID: mdl-35197578

ABSTRACT

A cell is delimited by numerous borders that define specific organelles. The walls of some organelles are particularly robust, such as in mitochondria or endoplasmic reticulum, but some are more fluid such as in phase-separated stress granules. Either way, all organelles can be damaged at times, leading their contents to leak out into the surrounding environment. Therefore, an elegant way to construct an innate immune defence system is to recognize host molecules that do not normally reside within a particular compartment. Here, we provide several examples where organellar homeostasis is lost, leading to the activation of a specific innate immune sensor; these include NLRP3 activation owing to a disrupted trans-Golgi network, Pyrin activation due to cytoskeletal damage, and cGAS-STING activation following the leakage of nuclear or mitochondrial DNA. Frequently, organelle damage is observed downstream of pathogenic infection but it can also occur in sterile settings as associated with auto-inflammatory disease. Therefore, understanding organellar homeostasis is central to efforts that will identify new innate immune pathways, and therapeutics that balance organellar homeostasis, or target the breakdown pathways that trigger innate immune sensors, could be useful treatments for infection and chronic inflammatory diseases.


Subject(s)
Mitochondria , Nucleotidyltransferases , DNA, Mitochondrial/metabolism , Endoplasmic Reticulum/metabolism , Homeostasis , Humans , Immunity, Innate , Mitochondria/metabolism , Nucleotidyltransferases/genetics
17.
Nat Commun ; 13(1): 2321, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35484149

ABSTRACT

Coatomer complex I (COPI) mediates retrograde vesicular trafficking from Golgi to the endoplasmic reticulum (ER) and within Golgi compartments. Deficiency in subunit alpha causes COPA syndrome and is associated with type I IFN signalling, although the upstream innate immune sensor involved was unknown. Using in vitro models we find aberrant activation of the STING pathway due to deficient retrograde but probably not intra-Golgi transport. Further we find the upstream cytosolic DNA sensor cGAS as essentially required to drive type I IFN signalling. Genetic deletion of COPI subunits COPG1 or COPD similarly induces type I IFN activation in vitro, which suggests that inflammatory diseases associated with mutations in other COPI subunit genes may exist. Finally, we demonstrate that inflammation in COPA syndrome patient peripheral blood mononuclear cells and COPI-deficient cell lines is ameliorated by treatment with the small molecule STING inhibitor H-151, suggesting targeted inhibition of the cGAS/STING pathway as a promising therapeutic approach.


Subject(s)
Leukocytes, Mononuclear , Nucleotidyltransferases , COP-Coated Vesicles/metabolism , Coat Protein Complex I/metabolism , Electron Transport Complex I/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Signal Transduction
18.
Sci Immunol ; 7(68): eabi6763, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35148201

ABSTRACT

Proteasome dysfunction can lead to autoinflammatory disease associated with elevated type I interferon (IFN-αß) and NF-κB signaling; however, the innate immune pathway driving this is currently unknown. Here, we identified protein kinase R (PKR) as an innate immune sensor for proteotoxic stress. PKR activation was observed in cellular models of decreased proteasome function and in multiple cell types from patients with proteasome-associated autoinflammatory disease (PRAAS). Furthermore, genetic deletion or small-molecule inhibition of PKR in vitro ameliorated inflammation driven by proteasome deficiency. In vivo, proteasome inhibitor-induced inflammatory gene transcription was blunted in PKR-deficient mice compared with littermate controls. PKR also acted as a rheostat for proteotoxic stress by triggering phosphorylation of eIF2α, which can prevent the translation of new proteins to restore homeostasis. Although traditionally known as a sensor of RNA, under conditions of proteasome dysfunction, PKR sensed the cytoplasmic accumulation of a known interactor, interleukin-24 (IL-24). When misfolded IL-24 egress into the cytosol was blocked by inhibition of the endoplasmic reticulum-associated degradation pathway, PKR activation and subsequent inflammatory signaling were blunted. Cytokines such as IL-24 are normally secreted from cells; therefore, cytoplasmic accumulation of IL-24 represents an internal danger-associated molecular pattern. Thus, we have identified a mechanism by which proteotoxic stress is detected, causing inflammation observed in the disease PRAAS.


Subject(s)
Immunity, Innate/immunology , Interleukins/immunology , eIF-2 Kinase/immunology , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , eIF-2 Kinase/deficiency
19.
Sci Immunol ; 7(75): eabi4611, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36112693

ABSTRACT

Dipeptidyl peptidase 9 (DPP9) is a direct inhibitor of NLRP1, but how it affects inflammasome regulation in vivo is not yet established. Here, we report three families with immune-associated defects, poor growth, pancytopenia, and skin pigmentation abnormalities that segregate with biallelic DPP9 rare variants. Using patient-derived primary cells and biochemical assays, these variants were shown to behave as hypomorphic or knockout alleles that failed to repress NLRP1. The removal of a single copy of Nlrp1a/b/c, Asc, Gsdmd, or Il-1r, but not Il-18, was sufficient to rescue the lethality of Dpp9 mutant neonates in mice. Similarly, dpp9 deficiency was partially rescued by the inactivation of asc, an obligate downstream adapter of the NLRP1 inflammasome, in zebrafish. These experiments suggest that the deleterious consequences of DPP9 deficiency were mostly driven by the aberrant activation of the canonical NLRP1 inflammasome and IL-1ß signaling. Collectively, our results delineate a Mendelian disorder of DPP9 deficiency driven by increased NLRP1 activity as demonstrated in patient cells and in two animal models of the disease.


Subject(s)
Apoptosis Regulatory Proteins , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases , Inflammasomes , Animals , Mice , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Inflammasomes/metabolism , Interleukin-1/metabolism , NLR Proteins/genetics , Zebrafish
20.
J Child Orthop ; 15(2): 114-121, 2021 Apr 19.
Article in English | MEDLINE | ID: mdl-34040657

ABSTRACT

PURPOSE: Comparison of two hexapod frame systems in paediatric tibial deformity correction; the Taylor Spatial Frame (TSF) and Orthex Hexapod System. METHODS: Paediatric patients with congenital and acquired tibial deformities treated with either TSF (between 2014 and 2016) or Orthex (between 2017 and 2019) frames were included in a retrospective comparative study. Outcome measures were healing index, pin infection rate, regenerate quality and density, software residual rate, deformity correction accuracy, strut exchanges and quality of life (QoL). RESULTS: The TSF group had 17 patients (18 frames) and the Orthex group had 21 patients (25 frames). The most common indications for tibial deformity correction were fibular hemimelia (14) and septic or traumatic growth arrest (8). The median time in frame was 230 days (TSF) versus 203 days (Orthex) (p= 0.06). The mean lengthening achieved was 54 mm (TSF) and 51 mm (Orthex) (p = 0.41). The healing index was 41 days/cm (TSF) versus 43 days/cm (Orthex) (p = 0.70). Pin site infections occurred more in the TSF cohort (40%) than in the Orthex cohort (18%) (p < 0.001). The regenerate in the Orthex group showed higher density at three months (p = 0.029) and was more homogenous (p = 0.023) at six months after frame application. Strut exchanges were less frequent with the Orthex system (p < 0.0001). QoL measures were similar in both cohorts (p = 0.92). CONCLUSIONS: This is the first study to compare two hexapod designs in paediatric orthopaedics. The Orthex system showed superiority in regenerate quality and a significant reduction in pin site infection rates. Both systems delivered predictable and accurate limb deformity correction. LEVEL OF EVIDENCE: III.

SELECTION OF CITATIONS
SEARCH DETAIL