Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Immunol ; 9(91): eabq6541, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38181093

ABSTRACT

Pore-forming toxins (PFTs) are the largest class of bacterial toxins and contribute to virulence by triggering host cell death. Vertebrates also express endogenous pore-forming proteins that induce cell death as part of host defense. To mitigate damage and promote survival, cells mobilize membrane repair mechanisms to neutralize and counteract pores, but how these pathways are activated is poorly understood. Here, we use a transposon-based gene activation screen to discover pathways that counteract the cytotoxicity of the archetypal PFT Staphylococcus aureus α-toxin. We identify the endolysosomal protein LITAF as a mediator of cellular resistance to PFT-induced cell death that is active against both bacterial toxins and the endogenous pore, gasdermin D, a terminal effector of pyroptosis. Activation of the ubiquitin ligase NEDD4 by potassium efflux mobilizes LITAF to recruit the endosomal sorting complexes required for transport (ESCRT) machinery to repair damaged membrane. Cells lacking LITAF, or carrying naturally occurring disease-associated mutations of LITAF, are highly susceptible to pore-induced death. Notably, LITAF-mediated repair occurs at endosomal membranes, resulting in expulsion of damaged membranes as exosomes, rather than through direct excision of pores from the surface plasma membrane. These results identify LITAF as a key effector that links sensing of cellular damage to repair.


Subject(s)
Bacterial Toxins , Pyroptosis , Animals , Cell Death , Cell Membrane , Endosomes
2.
J Clin Invest ; 126(10): 3758-3771, 2016 10 03.
Article in English | MEDLINE | ID: mdl-27571405

ABSTRACT

Studies of the genetic factors associated with human autoimmune disease suggest a multigenic origin of susceptibility; however, how these factors interact and through which tolerance pathways they operate generally remain to be defined. One key checkpoint occurs through the activity of the autoimmune regulator AIRE, which promotes central T cell tolerance. Recent reports have described a variety of dominant-negative AIRE mutations that likely contribute to human autoimmunity to a greater extent than previously thought. In families with these mutations, the penetrance of autoimmunity is incomplete, suggesting that other checkpoints play a role in preventing autoimmunity. Here, we tested whether a defect in LYN, an inhibitory protein tyrosine kinase that is implicated in systemic autoimmunity, could combine with an Aire mutation to provoke organ-specific autoimmunity. Indeed, mice with a dominant-negative allele of Aire and deficiency in LYN spontaneously developed organ-specific autoimmunity in the eye. We further determined that a small pool of retinal protein-specific T cells escaped thymic deletion as a result of the hypomorphic Aire function and that these cells also escaped peripheral tolerance in the presence of LYN-deficient dendritic cells, leading to highly destructive autoimmune attack. These findings demonstrate how 2 distinct tolerance pathways can synergize to unleash autoimmunity and have implications for the genetic susceptibility of autoimmune disease.


Subject(s)
Autoimmunity , Transcription Factors/physiology , src-Family Kinases/physiology , Animals , Antigen Presentation , Autoantibodies/metabolism , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Eye Proteins/immunology , Gastrointestinal Microbiome/immunology , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity , Retinol-Binding Proteins/immunology , Uveitis, Posterior/genetics , Uveitis, Posterior/immunology , AIRE Protein
3.
J Exp Med ; 212(12): 1993-2002, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26527800

ABSTRACT

Thymic central tolerance is essential to preventing autoimmunity. In medullary thymic epithelial cells (mTECs), the Autoimmune regulator (Aire) gene plays an essential role in this process by driving the expression of a diverse set of tissue-specific antigens (TSAs), which are presented and help tolerize self-reactive thymocytes. Interestingly, Aire has a highly tissue-restricted pattern of expression, with only mTECs and peripheral extrathymic Aire-expressing cells (eTACs) known to express detectable levels in adults. Despite this high level of tissue specificity, the cis-regulatory elements that control Aire expression have remained obscure. Here, we identify a highly conserved noncoding DNA element that is essential for Aire expression. This element shows enrichment of enhancer-associated histone marks in mTECs and also has characteristics of being an NF-κB-responsive element. Finally, we find that this element is essential for Aire expression in vivo and necessary to prevent spontaneous autoimmunity, reflecting the importance of this regulatory DNA element in promoting immune tolerance.


Subject(s)
DNA/immunology , Immune Tolerance/immunology , Regulatory Sequences, Nucleic Acid/immunology , Transcription Factors/immunology , Animals , Base Sequence , DNA/genetics , DNA/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , HEK293 Cells , Humans , Immune Tolerance/genetics , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Molecular Sequence Data , NF-kappa B/immunology , NF-kappa B/metabolism , Protein Binding/immunology , Regulatory Sequences, Nucleic Acid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome/genetics , Transcriptome/immunology , AIRE Protein
4.
Sci Transl Med ; 7(292): 292ra101, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-26084804

ABSTRACT

Autoimmune polyendocrine syndrome type 1 (APS1), a monogenic disorder caused by AIRE gene mutations, features multiple autoimmune disease components. Infertility is common in both males and females with APS1. Although female infertility can be explained by autoimmune ovarian failure, the mechanisms underlying male infertility have remained poorly understood. We performed a proteome-wide autoantibody screen in APS1 patient sera to assess the autoimmune response against the male reproductive organs. By screening human protein arrays with male and female patient sera and by selecting for gender-imbalanced autoantibody signals, we identified transglutaminase 4 (TGM4) as a male-specific autoantigen. Notably, TGM4 is a prostatic secretory molecule with critical role in male reproduction. TGM4 autoantibodies were detected in most of the adult male APS1 patients but were absent in all the young males. Consecutive serum samples further revealed that TGM4 autoantibodies first presented during pubertal age and subsequent to prostate maturation. We assessed the animal model for APS1, the Aire-deficient mouse, and found spontaneous development of TGM4 autoantibodies specifically in males. Aire-deficient mice failed to present TGM4 in the thymus, consistent with a defect in central tolerance for TGM4. In the mouse, we further link TGM4 immunity with a destructive prostatitis and compromised secretion of TGM4. Collectively, our findings in APS1 patients and Aire-deficient mice reveal prostate autoimmunity as a major manifestation of APS1 with potential role in male subfertility.


Subject(s)
Autoantigens/metabolism , Infertility, Male/enzymology , Infertility, Male/immunology , Prostate/enzymology , Transglutaminases/metabolism , Animals , Autoantibodies/metabolism , Epithelial Cells/enzymology , Female , Humans , Male , Mice, Inbred C57BL , Polyendocrinopathies, Autoimmune/enzymology , Polyendocrinopathies, Autoimmune/immunology , Prostatitis/pathology , Proteome/metabolism , Proteomics , Puberty , Thymus Gland/metabolism , Transcription Factors/deficiency , Transcription Factors/metabolism , AIRE Protein
5.
J Exp Med ; 211(5): 761-8, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24752296

ABSTRACT

Thymic central tolerance is a critical process that prevents autoimmunity but also presents a challenge to the generation of anti-tumor immune responses. Medullary thymic epithelial cells (mTECs) eliminate self-reactive T cells by displaying a diverse repertoire of tissue-specific antigens (TSAs) that are also shared by tumors. Therefore, while protecting against autoimmunity, mTECs simultaneously limit the generation of tumor-specific effector T cells by expressing tumor self-antigens. This ectopic expression of TSAs largely depends on autoimmune regulator (Aire), which is expressed in mature mTECs. Thus, therapies to deplete Aire-expressing mTECs represent an attractive strategy to increase the pool of tumor-specific effector T cells. Recent work has implicated the TNF family members RANK and RANK-Ligand (RANKL) in the development of Aire-expressing mTECs. We show that in vivo RANKL blockade selectively and transiently depletes Aire and TSA expression in the thymus to create a window of defective negative selection. Furthermore, we demonstrate that RANKL blockade can rescue melanoma-specific T cells from thymic deletion and that persistence of these tumor-specific effector T cells promoted increased host survival in response to tumor challenge. These results indicate that modulating central tolerance through RANKL can alter thymic output and potentially provide therapeutic benefit by enhancing anti-tumor immunity.


Subject(s)
Antigens, Neoplasm/metabolism , Autoimmunity/immunology , Central Tolerance/immunology , Epithelial Cells/metabolism , Neoplasms/immunology , RANK Ligand/metabolism , T-Lymphocytes/immunology , Animals , Central Tolerance/drug effects , Epithelial Cells/immunology , Flow Cytometry , Homeodomain Proteins/genetics , Indoles , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Osteoprotegerin/genetics , RANK Ligand/antagonists & inhibitors , Thymus Gland/cytology , Transcription Factors/metabolism , AIRE Protein
6.
Eur J Immunol ; 44(5): 1313-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24515814

ABSTRACT

Medullary thymic epithelial cells (mTECs) facilitate the deletion of developing self-reactive T cells by displaying a diverse repertoire of tissue-specific antigens, a process which largely depends on the expression of the autoimmune regulator (Aire) gene. Mature microRNAs (miRNAs) that regulate gene expression post-transcriptionally are generated in a multistep process. The microprocessor complex, including DGCR8, cleaves canonical miRNAs, but alternative DGCR8-independent miRNA biogenesis pathways exist as well. In order to study the role of canonical miRNAs in thymic epithelial cells (TECs), we ablated Dgcr8 using a FoxN1-Cre transgene. We report that DGCR8-deficient TECs are unable to maintain proper thymic architecture and exhibit a dramatic loss of thymic cellularity. Importantly, DGCR8-deficient TECs develop a severe loss of Aire(+) mTECs. Using a novel immunization approach to amplify and detect self-reactive T cells within a polyclonal TCR repertoire, we demonstrate a link between the loss of Aire expression in DGCR8-deficient TECs and the breakdown of negative selection in the thymus. Thus, DGCR8 and canonical miRNAs are important in TECs for supporting central tolerance.


Subject(s)
Epithelial Cells/immunology , Gene Expression Regulation/immunology , Immune Tolerance/physiology , MicroRNAs/immunology , Thymus Gland/immunology , Transcription Factors/immunology , Animals , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation/genetics , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , RNA-Binding Proteins/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics , AIRE Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...