Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Nat Immunol ; 21(1): 54-64, 2020 01.
Article in English | MEDLINE | ID: mdl-31819256

ABSTRACT

Ptpn6 is a cytoplasmic phosphatase that functions to prevent autoimmune and interleukin-1 (IL-1) receptor-dependent, caspase-1-independent inflammatory disease. Conditional deletion of Ptpn6 in neutrophils (Ptpn6∆PMN) is sufficient to initiate IL-1 receptor-dependent cutaneous inflammatory disease, but the source of IL-1 and the mechanisms behind IL-1 release remain unclear. Here, we investigate the mechanisms controlling IL-1α/ß release from neutrophils by inhibiting caspase-8-dependent apoptosis and Ripk1-Ripk3-Mlkl-regulated necroptosis. Loss of Ripk1 accelerated disease onset, whereas combined deletion of caspase-8 and either Ripk3 or Mlkl strongly protected Ptpn6∆PMN mice. Ptpn6∆PMN neutrophils displayed increased p38 mitogen-activated protein kinase-dependent Ripk1-independent IL-1 and tumor necrosis factor production, and were prone to cell death. Together, these data emphasize dual functions for Ptpn6 in the negative regulation of p38 mitogen-activated protein kinase activation to control tumor necrosis factor and IL-1α/ß expression, and in maintaining Ripk1 function to prevent caspase-8- and Ripk3-Mlkl-dependent cell death and concomitant IL-1α/ß release.


Subject(s)
Apoptosis/immunology , Caspase 8/immunology , Neutrophils/immunology , Protein Kinases/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Animals , Caspase 8/genetics , Cells, Cultured , Gene Deletion , Inflammation/immunology , Interleukin-1/immunology , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Receptors, Interleukin-1 Type I/immunology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Immunity ; 48(3): 570-583.e8, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29562203

ABSTRACT

Polymorphisms in NFKB1 that diminish its expression have been linked to human inflammatory diseases and increased risk for epithelial cancers. The underlying mechanisms are unknown, and the link is perplexing given that NF-κB signaling reportedly typically exerts pro-tumorigenic activity. Here we have shown that NF-κB1 deficiency, even loss of a single allele, resulted in spontaneous invasive gastric cancer (GC) in mice that mirrored the histopathological progression of human intestinal-type gastric adenocarcinoma. Bone marrow chimeras revealed that NF-κB1 exerted tumor suppressive functions in both epithelial and hematopoietic cells. RNA-seq analysis showed that NF-κB1 deficiency resulted in aberrant JAK-STAT signaling, which dysregulated expression of effectors of inflammation, antigen presentation, and immune checkpoints. Concomitant loss of STAT1 prevented these immune abnormalities and GC development. These findings provide mechanistic insight into how polymorphisms that attenuate NFKB1 expression predispose humans to epithelial cancers, highlighting the pro-tumorigenic activity of STAT1 and identifying targetable vulnerabilities in GC.


Subject(s)
Gene Expression Regulation, Neoplastic , Inflammation/genetics , Inflammation/metabolism , NF-kappa B/deficiency , STAT1 Transcription Factor/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Animals , Antigen Presentation/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Regulatory Networks , Humans , Inflammation/pathology , Mice , Mice, Knockout , STAT1 Transcription Factor/deficiency , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology
3.
Genes Dev ; 32(21-22): 1420-1429, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30366906

ABSTRACT

Mutations in Trp53, prevalent in human cancer, are reported to drive tumorigenesis through dominant-negative effects (DNEs) over wild-type TRP53 function as well as neomorphic gain-of-function (GOF) activity. We show that five TRP53 mutants do not accelerate lymphomagenesis on a TRP53-deficient background but strongly synergize with c-MYC overexpression in a manner that distinguishes the hot spot Trp53 mutations. RNA sequencing revealed that the mutant TRP53 DNE does not globally repress wild-type TRP53 function but disproportionately impacts a subset of wild-type TRP53 target genes. Accordingly, TRP53 mutant proteins impair pathways for DNA repair, proliferation, and metabolism in premalignant cells. This reveals that, in our studies of lymphomagenesis, mutant TRP53 drives tumorigenesis primarily through the DNE, which modulates wild-type TRP53 function in a manner advantageous for neoplastic transformation.


Subject(s)
Carcinogenesis/genetics , Mutation , Tumor Suppressor Protein p53/genetics , Animals , Lymphoma/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Suppressor Protein p53/metabolism
4.
Immunity ; 45(3): 513-526, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27523270

ABSTRACT

The kinases RIPK1 and RIPK3 and the pseudo-kinase MLKL have been identified as key regulators of the necroptotic cell death pathway, although a role for MLKL within the whole animal has not yet been established. Here, we have shown that MLKL deficiency rescued the embryonic lethality caused by loss of Caspase-8 or FADD. Casp8(-/-)Mlkl(-/-) and Fadd(-/-)Mlkl(-/-) mice were viable and fertile but rapidly developed severe lymphadenopathy, systemic autoimmune disease, and thrombocytopenia. These morbidities occurred more rapidly and with increased severity in Casp8(-/-)Mlkl(-/-) and Fadd(-/-)Mlkl(-/-) mice compared to Casp8(-/-)Ripk3(-/-) or Fadd(-/-)Ripk3(-/-) mice, respectively. These results demonstrate that MLKL is an essential effector of aberrant necroptosis in embryos caused by loss of Caspase-8 or FADD. Furthermore, they suggest that RIPK3 and/or MLKL may exert functions independently of necroptosis. It appears that non-necroptotic functions of RIPK3 contribute to the lymphadenopathy, autoimmunity, and excess cytokine production that occur when FADD or Caspase-8-mediated apoptosis is abrogated.


Subject(s)
Apoptosis/physiology , Autoimmune Diseases/metabolism , Cell Death/physiology , Fas-Associated Death Domain Protein/metabolism , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Caspase 8/metabolism , Mice , Mice, Inbred C57BL , Necrosis/metabolism
5.
Immunity ; 42(5): 890-902, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25979420

ABSTRACT

The mechanistic links between genetic variation and autoantibody production in autoimmune disease remain obscure. Autoimmune lymphoproliferative syndrome (ALPS) is caused by inactivating mutations in FAS or FASL, with autoantibodies thought to arise through failure of FAS-mediated removal of self-reactive germinal center (GC) B cells. Here we show that FAS is in fact not required for this process. Instead, FAS inactivation led to accumulation of a population of unconventional GC B cells that underwent somatic hypermutation, survived despite losing antigen reactivity, and differentiated into a large population of plasma cells that included autoantibody-secreting clones. IgE(+) plasma cell numbers, in particular, increased after FAS inactivation and a major cohort of ALPS-affected patients were found to have hyper-IgE. We propose that these previously unidentified cells, designated "rogue GC B cells," are a major driver of autoantibody production and provide a mechanistic explanation for the linked production of IgE and autoantibodies in autoimmune disease.


Subject(s)
Autoantibodies/immunology , B-Lymphocytes/cytology , Germinal Center/cytology , Germinal Center/immunology , Immunoglobulin E/immunology , fas Receptor/immunology , Animals , Autoantibodies/biosynthesis , B-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoglobulin E/biosynthesis , Mice , Polymerase Chain Reaction , fas Receptor/deficiency , fas Receptor/metabolism
6.
Health Expect ; 27(1): e13968, 2024 02.
Article in English | MEDLINE | ID: mdl-39102693

ABSTRACT

BACKGROUND AND CONTEXT: Involving people with lived experience of health conditions and the public (consumers) in health research is supported by policy, practice and research funding schemes. However, consumer involvement programmes in discovery-based preclinical research settings are uncommon. Few formal evaluations of these programmes are reported in the literature. OBJECTIVE: This study aimed to evaluate an established patient and public involvement programme operating in a major Australian Discovery-Based Medical Research Institute (DBMRI) to inform programme development and the wider field. DESIGN AND PARTICIPANTS: A multimethods programme evaluation incorporating demographic, descriptive and qualitative data obtained through consumer/researcher co-developed online surveys and semistructured virtual interviews. Programme participants (n = 111) were invited to complete an online survey seeking feedback on their experience of involvement, programme processes and perceived impacts. A purposive sample of 25 participants was interviewed. Descriptive data were analysed using explanatory statistics and qualitative data from surveys and interviews were thematically analysed. RESULTS: This consumer involvement programme was found to be useful and meaningful for most participants, with specific examples of perceived added value. Consumers most commonly engaged with researchers to inform research development, prepare funding applications or strengthen lay communication of science. Genuine consumer-researcher interactions, relationship development and mutual respect were key elements in a positive experience for participants. Opportunities to 'give back', to learn and to ground research in lived experience were identified programme strengths and benefits. Developing researcher training in how to work with consumers, increasing the diversity of the consumer group membership and expanding the range of consumer activities were identified opportunities for improvement. Organisational support and adequate programme resourcing were identified as key enablers. CONCLUSION: Discovery-based preclinical research is often viewed as being distant from clinical application; therefore, consumer involvement may be considered less relevant. However this study identified value in bringing a strong consumer voice to the discovery-based research process through a coordinated, organisation-wide approach with the potential for application in similar preclinical research settings. PATIENT OR PUBLIC CONTRIBUTION: Four consumer partners from the DBMRI Consumer Advisory Panel were actively engaged in developing this programme evaluation. Specifically, these consumer partners co-developed and pilot-tested surveys and interview guides, reviewed and commented on project data analysis and reporting and also contributed as co-authors by editing the manuscript.


Subject(s)
Biomedical Research , Community Participation , Patient Participation , Program Evaluation , Humans , Australia , Male , Female , Community Participation/methods , Middle Aged , Adult , Surveys and Questionnaires , Aged , Interviews as Topic
7.
Immunity ; 41(4): 646-56, 2014 Oct 16.
Article in English | MEDLINE | ID: mdl-25367576

ABSTRACT

Natural killer (NK) cells have been reported to control adaptive immune responses that occur in lymphoid organs at the early stages of immune challenge. The physiological purpose of such regulatory activity remains unclear, because it generally does not confer a survival advantage. We found that NK cells specifically eliminated activated CD4(+) T cells in the salivary gland during chronic murine cytomegalovirus (MCMV) infection. This was dependent on TNF-related apoptosis inducing ligand (TRAIL) expression by NK cells. Although NK cell-mediated deletion of CD4(+) T cells prolonged the chronicity of infection, it also constrained viral-induced autoimmunity. In the absence of this activity, chronic infection was associated with a Sjogren's-like syndrome characterized by focal lymphocytic infiltration into the glands, production of autoantibodies, and reduced saliva and tear secretion. Thus, NK cells are an important homeostatic control that balances the efficacy of adaptive immune responses with the risk of developing autoimmunity.


Subject(s)
Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Herpesviridae Infections/immunology , Killer Cells, Natural/immunology , Muromegalovirus/immunology , TNF-Related Apoptosis-Inducing Ligand/immunology , Adoptive Transfer , Animals , Apoptosis/immunology , Chronic Disease , Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/transplantation , Mice , Mice, Inbred BALB C , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Salivary Glands/immunology , Salivary Glands/pathology , Salivary Glands/virology
8.
Immunity ; 39(3): 573-83, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-24012421

ABSTRACT

Activation-induced cell death (AICD) plays a critical role in immune homeostasis and tolerance. In T-cell-dependent humoral responses, AICD of B cells is initiated by Fas ligand (FasL) on T cells, stimulating the Fas receptor on B cells. In contrast, T-cell-independent B cell responses involve innate-type B lymphocytes, such as marginal zone (MZ) B cells, and little is known about the mechanisms that control AICD during innate B cell responses to Toll-like receptor (TLR) activation. Here, we show that MZ B cells undergo AICD in response to TLR4 activation in vivo. The transmembrane activator, calcium modulator, and cyclophilin ligand interactor (TACI) receptor and TLR4 cooperate to upregulate expression of both FasL and Fas on MZ B cells and also to repress inhibitors of Fas-induced apoptosis signaling. These findings demonstrate an unappreciated role for TACI and its ligands in the regulation of AICD during T-cell-independent B cell responses.


Subject(s)
Apoptosis , Fas Ligand Protein/metabolism , Toll-Like Receptor 4/metabolism , Transmembrane Activator and CAML Interactor Protein/metabolism , fas Receptor/metabolism , Animals , B-Cell Activation Factor Receptor/biosynthesis , B-Lymphocytes/immunology , Enzyme Activation , Fas Ligand Protein/biosynthesis , Lipopolysaccharides , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Transmembrane Activator and CAML Interactor Protein/genetics
9.
Gastroenterology ; 159(4): 1444-1458.e15, 2020 10.
Article in English | MEDLINE | ID: mdl-32569771

ABSTRACT

BACKGROUND & AIMS: Activity of nuclear factor κB transcription factors and signaling via signal transducer and activator of transcription (STAT) are frequently altered in gastric cancer cells. Mice lacking NFKB1 (Nfkb1-/- mice) develop invasive gastric cancer, and their gastric tissues have increased levels of cytokines, such as interleukin (IL) 6, IL22, IL11, and tumor necrosis factor (TNF), as well as increased activation of STAT1. We investigated whether these cytokines were required for STAT1 activation in gastric tissues of mice and critical for gastric tumorigenesis. METHODS: We crossed Nfkb1-/- mice with Il6-/-, Il22-/-, Il11Rα-/-, and Tnf-/- mice. Stomach tissues from compound mutant mice were analyzed by histology, immunoblotting, and RNA sequencing. Lymphoid, myeloid, and epithelial cells were isolated from stomachs, and the levels of cytokines were determined by flow cytometric analysis. RESULTS: Nfkb1-/- mice developed gastritis, oxyntic atrophy, gastric dysplasia, and invasive tumors, whereas Nfkb1-/-Stat1-/- mice did not, even when followed for as long as 2 years. The levels of Il6, Il11, Il22, and Tnf messenger RNA were increased in the body and antrum of the stomachs from Nfkb1-/- mice, from 3-6 months of age. However, Nfkb1-/-Il6-/-, Nfkb1-/-Il22-/-, and Nfkb1-/-Il11Rα-/- mice still developed gastric tumors, although the absence of IL11 receptor (IL11R) significantly reduced development of invasive gastric tumors. Stomachs from Nfkb1-/-Tnf-/- mice exhibited significantly less gastritis and oxyntic atrophy and fewer tumors than Nfkb1-/- mice. This correlated with reduced activation of STAT1 and STAT3 and fewer numbers of T cells and B cells infiltrating the gastric body. Loss of STAT1 or TNF significantly reduced expression of PD-L1 on epithelial and myeloid (CD11b+) cells in the gastric mucosa of Nfkb1-/- mice-indeed, to the levels observed on the corresponding cells from wild-type mice. CONCLUSIONS: In studies of gastric tumor development in knockout mice, we found that loss of NFKB1 causes increased expression of TNF in the stomach and thereby drives activation of STAT1, resulting in an inflammatory immune response and the development of gastric cancer. IL11R appears to be required for the progression of gastric tumors to the invasive stage. These findings suggest that inhibitors of TNF, and possibly also inhibitors of IL11/IL11Rα, might be useful in the treatment of gastric cancer.


Subject(s)
Gastritis/pathology , NF-kappa B p50 Subunit/metabolism , STAT1 Transcription Factor/metabolism , Stomach Neoplasms/etiology , Stomach Neoplasms/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Carcinogenesis , Gastritis/etiology , Gastritis/metabolism , Interleukin-11/metabolism , Interleukin-6/metabolism , Mice , Signal Transduction , Stomach Neoplasms/metabolism
10.
Immunity ; 37(3): 451-62, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22960223

ABSTRACT

Although the proapoptotic BH3-only protein, Bim, is required for deletion of autoreactive thymocytes, Bim-deficient mice do not succumb to extensive organ-specific autoimmune disease. To determine whether other BH3-only proteins safeguard tolerance in the absence of Bim, we screened mice lacking Bim as well as other BH3-only proteins. Most strains showed no additional defects; however, mice deficient for both Puma and Bim spontaneously developed autoimmunity in multiple organs, and their T cells could transfer organ-specific autoimmunity. Puma- and Bim-double-deficient mice had a striking accumulation of mature, single-positive thymocytes, suggesting an additional defect in thymic deletion was the basis for disease. Transgenic mouse models of thymocyte deletion by peripheral neoantigens confirmed that the loss of Bim and Puma allowed increased numbers of autoreactive thymocytes to escape deletion. Our data show that Puma cooperates with Bim to impose a thymic-deletion checkpoint to peripheral self-antigens and cement the notion that defects in apoptosis alone are sufficient to cause autoimmune disease.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Autoantigens/immunology , Autoimmunity/immunology , Membrane Proteins/immunology , Proto-Oncogene Proteins/immunology , Thymocytes/immunology , Tumor Suppressor Proteins/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Apoptosis Regulatory Proteins/genetics , Autoimmunity/genetics , Bcl-2-Like Protein 11 , Cell Differentiation/genetics , Cell Differentiation/immunology , Female , Flow Cytometry , Male , Membrane Proteins/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/immunology , Self Tolerance/genetics , Self Tolerance/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymocytes/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Proteins/genetics
11.
Immunity ; 36(4): 646-57, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22483802

ABSTRACT

The immune system must distinguish viable cells from cells damaged by physical and infective processes. The damaged cell-recognition molecule Clec9A is expressed on the surface of the mouse and human dendritic cell subsets specialized for the uptake and processing of material from dead cells. Clec9A recognizes a conserved component within nucleated and nonnucleated cells, exposed when cell membranes are damaged. We have identified this Clec9A ligand as a filamentous form of actin in association with particular actin-binding domains of cytoskeletal proteins. We have determined the crystal structure of the human CLEC9A C-type lectin domain and propose a functional dimeric structure with conserved tryptophans in the ligand recognition site. Mutation of these residues ablated CLEC9A binding to damaged cells and to the isolated ligand complexes. We propose that Clec9A provides targeted recruitment of the adaptive immune system during infection and can also be utilized to enhance immune responses generated by vaccines.


Subject(s)
Actin Cytoskeleton/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lectins, C-Type/metabolism , Receptors, Immunologic/metabolism , Receptors, Mitogen/metabolism , Actins/metabolism , Adaptive Immunity , Animals , Binding Sites , Cell Line , Cell Membrane/metabolism , Dendritic Cells/cytology , Female , Humans , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Protein Structure, Secondary , Receptors, Immunologic/genetics , Receptors, Mitogen/chemistry , Receptors, Mitogen/genetics , Spectrin/metabolism
12.
Immunol Cell Biol ; 97(8): 740-752, 2019 09.
Article in English | MEDLINE | ID: mdl-31087793

ABSTRACT

FOXP3+ regulatory T (Treg) cells are essential for immunological tolerance and immune homeostasis. Despite a great deal of interest in modulating their number and function for the treatment of autoimmune disease or cancer, the precise mechanisms that control the homeostasis of Treg cells remain unclear. We report a new ENU-induced mutant mouse, lack of costimulation (loco), with atopic dermatitis and Treg cell deficiency typical of Card11 loss-of-function mutants. Three distinct single nucleotide variants were found in the Card11 introns 2, 10 and 20 that cause the loss of CARD11 expression in these mutant mice. These mutations caused the loss of thymic-derived, Neuropilin-1+ (NRP1+ ) Treg cells in neonatal and adult loco mice; however, residual peripherally induced NRP1- Treg cells remained. These peripherally generated Treg cells could be expanded in vivo by the administration of IL-2:anti-IL-2 complexes, indicating that this key homeostatic signaling axis remained intact in CARD11-deficient Treg cells. Furthermore, these expanded Treg cells could mediate near-normal suppression of activated, conventional CD4+ T cells, suggesting that CARD11 is dispensable for Treg cell function. In addition to shedding light on the requirements for CARD11 in Treg cell homeostasis and function, these data reveal novel noncoding Card11 loss-of-function mutations that impair the expression of this critical immune-regulatory protein.


Subject(s)
CARD Signaling Adaptor Proteins/deficiency , Dermatitis, Atopic/immunology , Homeostasis/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/immunology , Dermatitis, Atopic/genetics , Disease Models, Animal , Ethylnitrosourea/toxicity , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Homeostasis/genetics , Humans , Introns/drug effects , Introns/genetics , Introns/immunology , Loss of Function Mutation/drug effects , Loss of Function Mutation/immunology , Mice , Mice, Transgenic , Mutagenesis/immunology , Mutagens/toxicity , Neuropilin-1/immunology , Neuropilin-1/metabolism , Polymorphism, Single Nucleotide/drug effects , Polymorphism, Single Nucleotide/immunology , Signal Transduction/genetics , T-Lymphocytes, Regulatory/metabolism
13.
Nature ; 501(7466): 247-51, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24025841

ABSTRACT

Successful infection by enteric bacterial pathogens depends on the ability of the bacteria to colonize the gut, replicate in host tissues and disseminate to other hosts. Pathogens such as Salmonella, Shigella and enteropathogenic and enterohaemorrhagic (EPEC and EHEC, respectively) Escherichia coli use a type III secretion system (T3SS) to deliver virulence effector proteins into host cells during infection that promote colonization and interfere with antimicrobial host responses. Here we report that the T3SS effector NleB1 from EPEC binds to host cell death-domain-containing proteins and thereby inhibits death receptor signalling. Protein interaction studies identified FADD, TRADD and RIPK1 as binding partners of NleB1. NleB1 expressed ectopically or injected by the bacterial T3SS prevented Fas ligand or TNF-induced formation of the canonical death-inducing signalling complex (DISC) and proteolytic activation of caspase-8, an essential step in death-receptor-induced apoptosis. This inhibition depended on the N-acetylglucosamine transferase activity of NleB1, which specifically modified Arg 117 in the death domain of FADD. The importance of the death receptor apoptotic pathway to host defence was demonstrated using mice deficient in the FAS signalling pathway, which showed delayed clearance of the EPEC-like mouse pathogen Citrobacter rodentium and reversion to virulence of an nleB mutant. The activity of NleB suggests that EPEC and other attaching and effacing pathogens antagonize death-receptor-induced apoptosis of infected cells, thereby blocking a major antimicrobial host response.


Subject(s)
Enteropathogenic Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Gastrointestinal Tract/microbiology , Signal Transduction , Virulence Factors/metabolism , Animals , Caspase 8/metabolism , Cell Death , Citrobacter rodentium/pathogenicity , Citrobacter rodentium/physiology , Enteropathogenic Escherichia coli/pathogenicity , Enzyme Activation , Escherichia coli Infections/pathology , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/chemistry , Fas-Associated Death Domain Protein/metabolism , Female , HEK293 Cells , HeLa Cells , Humans , Male , Mice , N-Acetylglucosaminyltransferases/metabolism , Protein Structure, Tertiary , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Death Domain Protein/chemistry , TNF Receptor-Associated Death Domain Protein/metabolism , fas Receptor/deficiency , fas Receptor/metabolism
14.
Proc Natl Acad Sci U S A ; 112(31): 9698-703, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26195802

ABSTRACT

Rheumatoid arthritis (RA) and ankylosing spondylitis (AS) are chronic inflammatory diseases that together affect 2-3% of the population. RA and AS predominantly involve joints, but heart disease is also a common feature in RA and AS patients. Here we have studied a new spontaneous mutation that causes severe polyarthritis in bone phenotype spontaneous mutation 1 (BPSM1) mice. In addition to joint destruction, mutant mice also develop aortic root aneurism and aorto-mitral valve disease that can be fatal depending on the genetic background. The cause of the disease is the spontaneous insertion of a retrotransposon into the 3' untranslated region (3'UTR) of the tumor necrosis factor (TNF), which triggers its strong overexpression in myeloid cells. We found that several members of a family of RNA-binding, CCCH-containing zinc-finger proteins control TNF expression through its 3'UTR, and we identified a previously unidentified regulatory element in the UTR. The disease in BPSM1 mice is independent of the adaptive immune system and does not appear to involve inflammatory cytokines other than TNF. To our knowledge, this is the first animal model showing both polyarthritis and heart disease as a direct result of TNF deregulation. These results emphasize the therapeutic potential of anti-TNF drugs for the treatment of heart valve disease and identify potential therapeutic targets to control TNF expression and inflammation.


Subject(s)
3' Untranslated Regions/genetics , Arthritis/genetics , Heart Valve Diseases/genetics , Mutagenesis, Insertional/genetics , Mutation/genetics , Retroelements/genetics , Tumor Necrosis Factor-alpha/genetics , Aneurysm/pathology , Animals , Aortic Valve Insufficiency/complications , Aortic Valve Insufficiency/diagnostic imaging , Aortic Valve Insufficiency/pathology , Arthritis/blood , Arthritis/diagnostic imaging , Arthritis/pathology , Base Sequence , Bone Marrow Transplantation , Chemokines/blood , Chronic Disease , Disease Models, Animal , Fibrosis , Heart Valve Diseases/blood , Heart Valve Diseases/diagnostic imaging , Heart Valve Diseases/pathology , Humans , Inflammation/pathology , Joints/pathology , Mice, Mutant Strains , Mitral Valve/pathology , Molecular Sequence Data , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Short Interspersed Nucleotide Elements/genetics , Tumor Necrosis Factor-alpha/metabolism , Ultrasonography , X-Ray Microtomography , Zinc Fingers/genetics
15.
Reproduction ; 151(5): 553-62, 2016 May.
Article in English | MEDLINE | ID: mdl-26917450

ABSTRACT

Apoptosis plays a prominent role during ovarian development by eliminating large numbers of germ cells from the female germ line. However, the precise mechanisms and regulatory proteins involved in germ cell death are yet to be determined. In this study, we characterised the role of the pro-apoptotic BH3-only protein, BCL2-modifying factor (BMF), in germ cell apoptosis in embryonic and neonatal mouse ovaries. BMF protein was immunohistochemically localised to germ cells at embryonic days 15.5 (E15.5) and E17.5 and postnatal day 1 (PN1), coincident with entry into the meiotic prophase, but was undetectable at E13.5, and only present at low levels at PN3 and PN5. Consistent with this expression pattern, loss of BMF in female mice was associated with a decrease in apoptosis at E15.5 and E17.5. Furthermore, increased numbers of germ cells were found in ovaries from Bmf(-/-) mice compared with WT animals at E15.5 and PN1. However, germ cell numbers were comparable between Bmf(-/-) and WT ovaries at PN3, PN5 and PN10. Collectively, these data indicate that BMF mediates foetal oocyte loss and its action limits the maximal number of germ cells attained in the developing ovary, but does not influence the number of primordial follicles initially established in ovarian reserve.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Apoptosis , Embryo, Mammalian/pathology , Germ Cells/pathology , Oocytes/pathology , Oogenesis/physiology , Ovary/pathology , Animals , Blotting, Western , Cells, Cultured , Embryo, Mammalian/metabolism , Female , Fluorescent Antibody Technique , Germ Cells/metabolism , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/metabolism , Ovary/metabolism , Proto-Oncogene Proteins c-bcl-2
16.
EMBO Rep ; 15(9): 982-90, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24990442

ABSTRACT

A current paradigm proposes that mitochondrial damage is a critical determinant of NLRP3 inflammasome activation. Here, we genetically assess whether mitochondrial signalling represents a unified mechanism to explain how NLRP3 is activated by divergent stimuli. Neither co-deletion of the essential executioners of mitochondrial apoptosis BAK and BAX, nor removal of the mitochondrial permeability transition pore component cyclophilin D, nor loss of the mitophagy regulator Parkin, nor deficiency in MAVS affects NLRP3 inflammasome function. In contrast, caspase-8, a caspase essential for death-receptor-mediated apoptosis, is required for efficient Toll-like-receptor-induced inflammasome priming and cytokine production. Collectively, these results demonstrate that mitochondrial apoptosis is not required for NLRP3 activation, and highlight an important non-apoptotic role for caspase-8 in regulating inflammasome activation and pro-inflammatory cytokine levels.


Subject(s)
Carrier Proteins/biosynthesis , Caspase 8/biosynthesis , Inflammasomes/metabolism , Mitochondria/metabolism , Apoptosis/genetics , Autophagy/genetics , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Carrier Proteins/genetics , Caspase 8/genetics , Cells, Cultured , Peptidyl-Prolyl Isomerase F , Cyclophilins/antagonists & inhibitors , Cyclophilins/genetics , Humans , Interleukin-1beta/biosynthesis , Mitochondria/pathology , Mitophagy/genetics , NLR Family, Pyrin Domain-Containing 3 Protein , Toll-Like Receptors/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
17.
J Pathol ; 236(3): 326-36, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25727407

ABSTRACT

NF-κB signalling is an important factor in the development of inflammation-associated cancers. Mouse models of Helicobacter-induced gastric cancer and colitis-associated colorectal cancer have demonstrated that classical NF-κB signalling is an important regulator of these processes. In the stomach, it has also been demonstrated that signalling involving specific NF-κB proteins, including NF-κB1/p50, NF-κB2/p52, and c-Rel, differentially regulate the development of gastric pre-neoplasia. To investigate the effect of NF-κB subunit loss on colitis-associated carcinogenesis, we administered azoxymethane followed by pulsed dextran sodium sulphate to C57BL/6, Nfkb1(-/-), Nfkb2(-/-), and c-Rel(-/-) mice. Animals lacking the c-Rel subunit were more susceptible to colitis-associated cancer than wild-type mice, developing 3.5 times more colonic polyps per animal than wild-type mice. Nfkb2(-/-) mice were resistant to colitis-associated cancer, developing fewer polyps per colon than wild-type mice (median 1 compared to 4). To investigate the mechanisms underlying these trends, azoxymethane and dextran sodium sulphate were administered separately to mice of each genotype. Nfkb2(-/-) mice developed fewer clinical signs of colitis and exhibited less severe colitis and an attenuated cytokine response compared with all other groups following DSS administration. Azoxymethane administration did not fully suppress colonic epithelial mitosis in c-Rel(-/-) mice and less colonic epithelial apoptosis was also observed in this genotype compared to wild-type counterparts. These observations demonstrate different functions of specific NF-κB subunits in this model of colitis-associated carcinogenesis. NF-κB2/p52 is necessary for the development of colitis, whilst c-Rel-mediated signalling regulates colonic epithelial cell turnover following DNA damage.


Subject(s)
Adenoma/metabolism , Colitis/complications , Colonic Neoplasms/metabolism , NF-kappa B p50 Subunit/metabolism , NF-kappa B p52 Subunit/metabolism , Proto-Oncogene Proteins c-rel/metabolism , Adenoma/chemically induced , Adenoma/etiology , Animals , Azoxymethane/toxicity , Cell Transformation, Neoplastic/metabolism , Colitis/chemically induced , Colonic Neoplasms/chemically induced , Colonic Neoplasms/etiology , Cytokines/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , Disease Susceptibility , Epithelial Cells/metabolism , Female , Inflammation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
19.
Proc Natl Acad Sci U S A ; 110(7): 2599-604, 2013 Feb 12.
Article in English | MEDLINE | ID: mdl-23349374

ABSTRACT

Dysregulation of the "intrinsic" apoptotic pathway is associated with the development of cancer and autoimmune disease. Bak and Bax are two proapoptotic members of the Bcl-2 protein family with overlapping, essential roles in the intrinsic apoptotic pathway. Their activity is critical for the control of cell survival during lymphocyte development and homeostasis, best demonstrated by defects in thymic T-cell differentiation and peripheral lymphoid homeostasis caused by their combined loss. Because most bak(-/-)bax(-/-) mice die perinatally, the roles of Bax and Bak in immunological tolerance and prevention of autoimmune disease remain unclear. We show that mice reconstituted with a Bak/Bax doubly deficient hematopoietic compartment develop a fatal systemic lupus erythematosus-like autoimmune disease characterized by hypergammaglobulinemia, autoantibodies, lymphadenopathy, glomerulonephritis, and vasculitis. Importantly, these mice also develop a multiorgan autoimmune disease with autoantibodies against most solid glandular structures and evidence of glandular atrophy and necrotizing vasculitis. Interestingly, similar albeit less severe pathology was observed in mice containing a hematopoietic compartment deficient for only Bak, a phenotype reminiscent of the disease seen in patients with point mutations in BAK. These studies demonstrate a critical role for Bak and an ancillary role for Bax in safeguarding immunological tolerance and prevention of autoimmune disease. This suggests that direct activators of the intrinsic apoptotic pathway, such as BH3 mimetics, may be useful for treatment of diverse autoimmune diseases.


Subject(s)
Apoptosis/immunology , Autoimmune Diseases/immunology , bcl-2 Homologous Antagonist-Killer Protein/immunology , bcl-2-Associated X Protein/immunology , Animals , Autoantibodies/immunology , Autoimmune Diseases/pathology , Blotting, Western , Chemokines/blood , Crosses, Genetic , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Histological Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , bcl-2 Homologous Antagonist-Killer Protein/deficiency , bcl-2-Associated X Protein/deficiency
20.
Nature ; 461(7264): 659-63, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19794494

ABSTRACT

Fas ligand (FasL), an apoptosis-inducing member of the TNF cytokine family, and its receptor Fas are critical for the shutdown of chronic immune responses and prevention of autoimmunity. Accordingly, mutations in their genes cause severe lymphadenopathy and autoimmune disease in mice and humans. FasL function is regulated by deposition in the plasma membrane and metalloprotease-mediated shedding. Here we generated gene-targeted mice that selectively lack either secreted FasL (sFasL) or membrane-bound FasL (mFasL) to resolve which of these forms is required for cell killing and to explore their hypothesized non-apoptotic activities. Mice lacking sFasL (FasL(Deltas/Deltas)) appeared normal and their T cells readily killed target cells, whereas T cells lacking mFasL (FasL(Deltam/Deltam)) could not kill cells through Fas activation. FasL(Deltam/Deltam) mice developed lymphadenopathy and hyper-gammaglobulinaemia, similar to FasL(gld/gld) mice, which express a mutant form of FasL that cannot bind Fas, but surprisingly, FasL(Deltam/Deltam) mice (on a C57BL/6 background) succumbed to systemic lupus erythematosus (SLE)-like autoimmune kidney destruction and histiocytic sarcoma, diseases that occur only rarely and much later in FasL(gld/gld) mice. These results demonstrate that mFasL is essential for cytotoxic activity and constitutes the guardian against lymphadenopathy, autoimmunity and cancer, whereas excess sFasL appears to promote autoimmunity and tumorigenesis through non-apoptotic activities.


Subject(s)
Apoptosis , Cell Membrane/metabolism , Fas Ligand Protein/metabolism , fas Receptor/metabolism , Animals , Antibodies, Antinuclear/immunology , Cytidine Deaminase/metabolism , Cytotoxicity, Immunologic , Fas Ligand Protein/deficiency , Fas Ligand Protein/genetics , Glomerulonephritis/metabolism , Histiocytic Sarcoma/metabolism , Hypergammaglobulinemia/metabolism , Lupus Erythematosus, Systemic/metabolism , Lymphatic Diseases/metabolism , Mice , Mice, Inbred C57BL , Mutation , Splenomegaly/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL