Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Nat Immunol ; 24(9): 1473-1486, 2023 09.
Article in English | MEDLINE | ID: mdl-37580603

ABSTRACT

Omnivorous animals, including mice and humans, tend to prefer energy-dense nutrients rich in fat over plant-based diets, especially for short periods of time, but the health consequences of this short-term consumption of energy-dense nutrients are unclear. Here, we show that short-term reiterative switching to 'feast diets', mimicking our social eating behavior, breaches the potential buffering effect of the intestinal microbiota and reorganizes the immunological architecture of mucosa-associated lymphoid tissues. The first dietary switch was sufficient to induce transient mucosal immune depression and suppress systemic immunity, leading to higher susceptibility to Salmonella enterica serovar Typhimurium and Listeria monocytogenes infections. The ability to respond to antigenic challenges with a model antigen was also impaired. These observations could be explained by a reduction of CD4+ T cell metabolic fitness and cytokine production due to impaired mTOR activity in response to reduced microbial provision of fiber metabolites. Reintroducing dietary fiber rewired T cell metabolism and restored mucosal and systemic CD4+ T cell functions and immunity. Finally, dietary intervention with human volunteers confirmed the effect of short-term dietary switches on human CD4+ T cell functionality. Therefore, short-term nutritional changes cause a transient depression of mucosal and systemic immunity, creating a window of opportunity for pathogenic infection.


Subject(s)
Mucous Membrane , Salmonella typhimurium , Humans , Mice , Animals , T-Lymphocytes , Immunity, Mucosal
2.
bioRxiv ; 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37425769

ABSTRACT

Extraintestinal autoimmune diseases are multifactorial with translocating gut pathobionts implicated as instigators and perpetuators in mice. However, the microbial contributions to autoimmunity in humans remain largely unclear, including whether specific pathological human adaptive immune responses are triggered by such pathobionts. We show here that the translocating pathobiont Enterococcus gallinarum induces human IFNγ + Th17 differentiation and IgG3 subclass switch of anti- E. gallinarum RNA and correlating anti-human RNA autoantibody responses in patients with systemic lupus erythematosus and autoimmune hepatitis. Human Th17 induction by E. gallinarum is cell-contact dependent and involves TLR8-mediated human monocyte activation. In murine gnotobiotic lupus models, E. gallinarum translocation triggers IgG3 anti-RNA autoantibody titers that correlate with renal autoimmune pathophysiology and with disease activity in patients. Overall, we define cellular mechanisms of how a translocating pathobiont induces human T- and B-cell-dependent autoimmune responses, providing a framework for developing host- and microbiota-derived biomarkers and targeted therapies in extraintestinal autoimmune diseases. One Sentence Summary: Translocating pathobiont Enterococcus gallinarum promotes human Th17 and IgG3 autoantibody responses linked to disease activity in autoimmune patients.

3.
Cell Rep ; 33(6): 108371, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33176151

ABSTRACT

Transcription factor EB (TFEB) activates lysosomal biogenesis genes in response to environmental cues. Given implications of impaired TFEB signaling and lysosomal dysfunction in metabolic, neurological, and infectious diseases, we aim to systematically identify TFEB-directed circuits by examining transcriptional responses to TFEB subcellular localization and stimulation. We reveal that steady-state nuclear TFEB is sufficient to activate transcription of lysosomal, autophagy, and innate immunity genes, whereas other targets require higher thresholds of stimulation. Furthermore, we identify shared and distinct transcriptional signatures between mTOR inhibition and bacterial autophagy. Using a genome-wide CRISPR library, we find TFEB targets that protect cells from or sensitize cells to lysosomal cell death. BHLHE40 and BHLHE41, genes responsive to high, sustained levels of nuclear TFEB, act in opposition to TFEB upon lysosomal cell death induction. Further investigation identifies genes counter-regulated by TFEB and BHLHE40/41, adding this negative feedback to the current understanding of TFEB regulatory mechanisms.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Gene Knockout Techniques , HeLa Cells , Homeodomain Proteins/genetics , Humans , Lysosomes/metabolism , Mitochondria/metabolism , Transcription, Genetic
4.
J Immunol ; 203(7): 1820-1829, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31451676

ABSTRACT

The clear role of autophagy in human inflammatory diseases such as Crohn disease was first identified by genome-wide association studies and subsequently dissected in multiple mechanistic studies. ATG16L1 has been particularly well studied in knockout and hypomorph settings as well as models recapitulating the Crohn disease-associated T300A polymorphism. Interestingly, ATG16L1 has a single homolog, ATG16L2, which is independently implicated in diseases, including Crohn disease and systemic lupus erythematosus. However, the contribution of ATG16L2 to canonical autophagy pathways and other cellular functions is poorly understood. To better understand its role, we generated and analyzed the first, to our knowledge, ATG16L2 knockout mouse. Our results show that ATG16L1 and ATG16L2 contribute very distinctly to autophagy and cellular ontogeny in myeloid, lymphoid, and epithelial lineages. Dysregulation of any of these lineages could contribute to complex diseases like Crohn disease and systemic lupus erythematosus, highlighting the value of examining cell-specific effects. We also identify a novel genetic interaction between ATG16L2 and epithelial ATG16L1. These findings are discussed in the context of how these genes may contribute distinctly to human disease.


Subject(s)
Autophagic Cell Death , Autophagy-Related Proteins , Carrier Proteins , Crohn Disease , Lupus Erythematosus, Systemic , Animals , Autophagic Cell Death/genetics , Autophagic Cell Death/immunology , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/immunology , Carrier Proteins/genetics , Carrier Proteins/immunology , Crohn Disease/genetics , Crohn Disease/immunology , Disease Models, Animal , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Knockout , Organ Specificity/genetics , Organ Specificity/immunology
5.
PLoS Genet ; 15(5): e1008084, 2019 05.
Article in English | MEDLINE | ID: mdl-31059511

ABSTRACT

The transition from mitotic to meiotic cell cycles is essential for haploid gamete formation and fertility. Stimulated by retinoic acid gene 8 (Stra8) is an essential gatekeeper of meiotic initiation in vertebrates; yet, the molecular role of STRA8 remains principally unknown. Here we demonstrate that STRA8 functions as a suppressor of autophagy during spermatogenesis in mice. Stra8-deficient germ cells fail to enter meiosis and present aberrant upregulation of autophagy-lysosome genes, commensurate with autophagy activation. Biochemical assays show that ectopic expression of STRA8 alone is sufficient to inhibit both autophagy induction and maturation. Studies also revealed that, Nr1d1, a nuclear hormone receptor gene, is upregulated in Stra8-deficient testes and that STRA8 binds to the Nr1d1 promoter, indicating that Nr1d1 is a direct target of STRA8 transcriptional repression. In addition, it was found that NR1D1 binds to the promoter of Ulk1, a gene essential for autophagy initiation, and that Nr1d1 is required for the upregulated Ulk1 expression in Stra8-deficient testes. Furthermore, both genetic deletion of Nr1d1 and pharmacologic inhibition of NR1D1 by its synthetic antagonist SR8278 exhibit rescuing effects on the meiotic initiation defects observed in Stra8-deficient male germ cells. Together, the data suggest a novel link between STRA8-mediated autophagy suppression and meiotic initiation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autophagy-Related Protein-1 Homolog/genetics , Fertility/genetics , Meiosis , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Spermatogenesis/genetics , Spermatozoa/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Animals , Autophagy/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Base Sequence , Gene Expression Regulation, Developmental , Isoquinolines/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Promoter Regions, Genetic , Protein Binding , Spermatozoa/cytology , Testis/cytology , Testis/growth & development , Testis/metabolism , Thiophenes/pharmacology
6.
J Immunol ; 202(11): 3256-3266, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31010852

ABSTRACT

Tetraspanins are a family of proteins possessing four transmembrane domains that help in lateral organization of plasma membrane proteins. These proteins interact with each other as well as other receptors and signaling proteins, resulting in functional complexes called "tetraspanin microdomains." Tetraspanins, including CD82, play an essential role in the pathogenesis of fungal infections. Dectin-1, a receptor for the fungal cell wall carbohydrate ß-1,3-glucan, is vital to host defense against fungal infections. The current study identifies a novel association between tetraspanin CD82 and Dectin-1 on the plasma membrane of Candida albicans-containing phagosomes independent of phagocytic ability. Deletion of CD82 in mice resulted in diminished fungicidal activity, increased C. albicans viability within macrophages, and decreased cytokine production (TNF-α, IL-1ß) at both mRNA and protein level in macrophages. Additionally, CD82 organized Dectin-1 clustering in the phagocytic cup. Deletion of CD82 modulates Dectin-1 signaling, resulting in a reduction of Src and Syk phosphorylation and reactive oxygen species production. CD82 knockout mice were more susceptible to C. albicans as compared with wild-type mice. Furthermore, patient C. albicans-induced cytokine production was influenced by two human CD82 single nucleotide polymorphisms, whereas an additional CD82 single nucleotide polymorphism increased the risk for candidemia independent of cytokine production. Together, these data demonstrate that CD82 organizes the proper assembly of Dectin-1 signaling machinery in response to C. albicans.


Subject(s)
Candida albicans/physiology , Candidiasis/metabolism , Cell Membrane/metabolism , Kangai-1 Protein/metabolism , Lectins, C-Type/metabolism , Macrophages/immunology , Phagosomes/metabolism , Animals , Candidiasis/immunology , Cell Line , Genetic Predisposition to Disease , Humans , Immunity, Cellular , Interleukin-1beta/metabolism , Kangai-1 Protein/genetics , Lectins, C-Type/genetics , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymorphism, Single Nucleotide , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
7.
Elife ; 82019 01 22.
Article in English | MEDLINE | ID: mdl-30666959

ABSTRACT

Inflammatory bowel disease (IBD) is driven by dysfunction between host genetics, the microbiota, and immune system. Knowledge gaps remain regarding how IBD genetic risk loci drive gut microbiota changes. The Crohn's disease risk allele ATG16L1 T300A results in abnormal Paneth cells due to decreased selective autophagy, increased cytokine release, and decreased intracellular bacterial clearance. To unravel the effects of ATG16L1 T300A on the microbiota and immune system, we employed a gnotobiotic model using human fecal transfers into ATG16L1 T300A knock-in mice. We observed increases in Bacteroides ovatus and Th1 and Th17 cells in ATG16L1 T300A mice. Association of altered Schaedler flora mice with B. ovatus specifically increased Th17 cells selectively in ATG16L1 T300A knock-in mice. Changes occur before disease onset, suggesting that ATG16L1 T300A contributes to dysbiosis and immune infiltration prior to disease symptoms. Our work provides insight for future studies on IBD subtypes, IBD patient treatment and diagnostics.


Subject(s)
Autophagy-Related Proteins/genetics , Crohn Disease/genetics , Crohn Disease/microbiology , Gastrointestinal Microbiome , Th1 Cells/cytology , Th17 Cells/cytology , Alleles , Animals , Bacteroides , Dysbiosis/genetics , Dysbiosis/microbiology , Fecal Microbiota Transplantation , Feces/microbiology , Gene Knock-In Techniques , Genotype , Humans , Immune System , Mice , Polymorphism, Genetic , Risk , Th1 Cells/microbiology , Th17 Cells/microbiology
8.
Science ; 359(6380): 1161-1166, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29420262

ABSTRACT

Polymorphisms in C1orf106 are associated with increased risk of inflammatory bowel disease (IBD). However, the function of C1orf106 and the consequences of disease-associated polymorphisms are unknown. Here we demonstrate that C1orf106 regulates adherens junction stability by regulating the degradation of cytohesin-1, a guanine nucleotide exchange factor that controls activation of ARF6. By limiting cytohesin-1-dependent ARF6 activation, C1orf106 stabilizes adherens junctions. Consistent with this model, C1orf106-/- mice exhibit defects in the intestinal epithelial cell barrier, a phenotype observed in IBD patients that confers increased susceptibility to intestinal pathogens. Furthermore, the IBD risk variant increases C1orf106 ubiquitination and turnover with consequent functional impairments. These findings delineate a mechanism by which a genetic polymorphism fine-tunes intestinal epithelial barrier integrity and elucidate a fundamental mechanism of cellular junctional control.


Subject(s)
Adherens Junctions/genetics , Guanine Nucleotide Exchange Factors/metabolism , Inflammatory Bowel Diseases/genetics , Phosphoproteins/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Animals , Caco-2 Cells , HEK293 Cells , Humans , Immunoprecipitation , Intestinal Mucosa/pathology , Mice , Mice, Mutant Strains , Phosphoproteins/genetics , Polymorphism, Genetic , Proteolysis , Risk , Ubiquitination/genetics
9.
Autophagy ; 14(2): 216-220, 2018.
Article in English | MEDLINE | ID: mdl-29130415

ABSTRACT

The discovery of numerous genetic variants in the human genome that are associated with inflammatory bowel disease (IBD) has revealed critical pathways that play important roles in intestinal homeostasis. These genetic studies have identified a critical role for macroautophagy/autophagy and more recently, lysosomal function, in maintaining the intestinal barrier and mucosal homeostasis. This review highlights recent work on the functional characterization of IBD-associated human genetic variants in cell type-specific functions for autophagy.


Subject(s)
Autophagy/immunology , Inflammatory Bowel Diseases/immunology , Lysosomes/metabolism , Animals , Autophagy/genetics , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Crohn Disease/genetics , Crohn Disease/immunology , Cytokines/metabolism , Genetic Predisposition to Disease , Humans , Inflammatory Bowel Diseases/genetics , Intestinal Mucosa/immunology , Mice , Reactive Oxygen Species/metabolism
10.
Sci Rep ; 7(1): 13938, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29066772

ABSTRACT

Understanding the transcription factors that modulate epithelial resistance to injury is necessary for understanding intestinal homeostasis and injury repair processes. Recently, transcription factor EB (TFEB) was implicated in expression of autophagy and host defense genes in nematodes and mammalian cells. However, the in vivo roles of TFEB in the mammalian intestinal epithelium were not known. Here, we used mice with a conditional deletion of Tfeb in the intestinal epithelium (Tfeb ΔIEC) to examine its importance in defense against injury. Unperturbed Tfeb ΔIEC mice exhibited grossly normal intestinal epithelia, except for a defect in Paneth cell granules. Tfeb ΔIEC mice exhibited lower levels of lipoprotein ApoA1 expression, which is downregulated in Crohn's disease patients and causally linked to colitis susceptibility. Upon environmental epithelial injury using dextran sodium sulfate (DSS), Tfeb ΔIEC mice exhibited exaggerated colitis. Thus, our study reveals that TFEB is critical for resistance to intestinal epithelial cell injury, potentially mediated by APOA1.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Intestinal Mucosa/pathology , Animals , Apolipoprotein A-I/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/deficiency , Female , Gene Expression Regulation , Homeostasis , Intestinal Mucosa/metabolism , Male , Mice , Paneth Cells/metabolism
11.
Cell Rep ; 17(9): 2183-2194, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27880896

ABSTRACT

Xenophagy is a form of selective autophagy that involves the targeting and elimination of intracellular pathogens through several recognition, recruitment, and ubiquitination events. E3 ubiquitin ligases control substrate selectivity in the ubiquitination cascade; however, systematic approaches to map the role of E3 ligases in antibacterial autophagy have been lacking. We screened more than 600 putative human E3 ligases, identifying E3 ligases that are required for adaptor protein recruitment and LC3-bacteria colocalization, critical steps in antibacterial autophagy. An unbiased informatics approach pinpointed RNF166 as a key gene that interacts with the autophagy network and controls the recruitment of ubiquitin as well as the autophagy adaptors p62 and NDP52 to bacteria. Mechanistic studies demonstrated that RNF166 catalyzes K29- and K33-linked polyubiquitination of p62 at residues K91 and K189. Thus, our study expands the catalog of E3 ligases that mediate antibacterial autophagy and identifies a critical role for RNF166 in this process.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Anti-Bacterial Agents/pharmacology , Autophagy/drug effects , Ubiquitin-Protein Ligases/metabolism , HEK293 Cells , HeLa Cells , Humans , Listeria/drug effects , Listeria/growth & development , Lysine/metabolism , Protein Binding , RNA, Small Interfering/metabolism , Salmonella typhimurium/drug effects , Salmonella typhimurium/growth & development , Salmonella typhimurium/metabolism , Sequestosome-1 Protein/metabolism , Ubiquitination
12.
Immunity ; 44(6): 1392-405, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27287411

ABSTRACT

Although numerous polymorphisms have been associated with inflammatory bowel disease (IBD), identifying the function of these genetic factors has proved challenging. Here we identified a role for nine genes in IBD susceptibility loci in antibacterial autophagy and characterized a role for one of these genes, GPR65, in maintaining lysosome function. Mice lacking Gpr65, a proton-sensing G protein-coupled receptor, showed increased susceptibly to bacteria-induced colitis. Epithelial cells and macrophages lacking GPR65 exhibited impaired clearance of intracellular bacteria and accumulation of aberrant lysosomes. Similarly, IBD patient cells and epithelial cells expressing an IBD-associated missense variant, GPR65 I231L, displayed aberrant lysosomal pH resulting in lysosomal dysfunction, impaired bacterial restriction, and altered lipid droplet formation. The GPR65 I231L polymorphism was sufficient to confer decreased GPR65 signaling. Collectively, these data establish a role for GPR65 in IBD susceptibility and identify lysosomal dysfunction as a potentially causative element in IBD pathogenesis with effects on cellular homeostasis and defense.


Subject(s)
Colitis/immunology , Epithelial Cells/immunology , Inflammatory Bowel Diseases/genetics , Lysosomes/physiology , Receptors, G-Protein-Coupled/metabolism , Salmonella Infections/immunology , Salmonella enterica/immunology , Salmonella typhimurium/immunology , Animals , Genetic Predisposition to Disease , HeLa Cells , Humans , Inflammatory Bowel Diseases/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagosomes/physiology , Polymorphism, Genetic , Receptors, G-Protein-Coupled/genetics , Risk
13.
mBio ; 6(5): e01157-15, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26350966

ABSTRACT

UNLABELLED: A core set of autophagy proteins is required for gamma interferon (IFN-γ)-mediated clearance of Toxoplasma gondii in the mouse because of their control of several downstream effectors, including immunity-related GTPases (IRGs) and guanylate-binding proteins (GBPs). However, these effectors are absent (i.e., IRGs) from or nonessential (i.e., GBPs) in IFN-γ-activated human cells, raising the question of how these cells control parasite replication. Here, we define a novel role for ubiquitination and recruitment of autophagy adaptors in the strain-specific control of T. gondii replication in IFN-γ-activated human cells. Vacuoles containing susceptible strains of T. gondii became ubiquitinated, recruited the adaptors p62 and NDP52, and were decorated with LC3. Parasites within LC3-positive vacuoles became enclosed in multiple layers of host membranes, resulting in stunting of parasite replication. However, LC3-positive T. gondii-containing vacuoles did not fuse with endosomes and lysosomes, indicating that this process is fundamentally different from xenophagy, a form of autophagy involved in the control of intracellular bacterial pathogens. Genetic knockout of ATG16L or ATG7 reverted the membrane encapsulation and restored parasite replication, indicating that core autophagy proteins involved in LC3 conjugation are important in the control of parasite growth. Despite a role for the core autophagy machinery in this process, upstream activation through Beclin 1 was not sufficient to enhance the ubiquitination of T. gondii-containing vacuoles, suggesting a lack of reliance on canonical autophagy. These findings demonstrate a new mechanism for IFN-γ-dependent control of T. gondii in human cells that depends on ubiquitination and core autophagy proteins that mediate membrane engulfment and restricted growth. IMPORTANCE: Autophagy is a process of cellular remodeling that allows the cell to recycle senescent organelles and recapture nutrients. During innate immune responses in the mouse, autophagy is recruited to help target intracellular pathogens and thus eliminate them. However, the antimicrobial mediators that depend on autophagy in the mouse are not conserved in humans, raising the issue of how human cells control intracellular pathogens. Our study defines a new pathway for the control of the ubiquitous intracellular parasite T. gondii in human cells activated by IFN-γ. Recruitment of autophagy adaptors resulted in engulfment of the parasite in multiple membranes and growth impairment. Although susceptible type 2 and 3 stains of T. gondii were captured by this autophagy-dependent pathway, type 1 strains were able to avoid entrapment.


Subject(s)
Autophagy , Epithelial Cells/immunology , Epithelial Cells/parasitology , Interferon-gamma/metabolism , Toxoplasma/immunology , HeLa Cells , Humans , Ubiquitination , Vacuoles/parasitology
14.
Proc Natl Acad Sci U S A ; 112(31): E4281-7, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26195741

ABSTRACT

Studies of human genetics and pathophysiology have implicated the regulation of autophagy in inflammation, neurodegeneration, infection, and autoimmunity. These findings have motivated the use of small-molecule probes to study how modulation of autophagy affects disease-associated phenotypes. Here, we describe the discovery of the small-molecule probe BRD5631 that is derived from diversity-oriented synthesis and enhances autophagy through an mTOR-independent pathway. We demonstrate that BRD5631 affects several cellular disease phenotypes previously linked to autophagy, including protein aggregation, cell survival, bacterial replication, and inflammatory cytokine production. BRD5631 can serve as a valuable tool for studying the role of autophagy in the context of cellular homeostasis and disease.


Subject(s)
Autophagy/drug effects , Genetics, Medical , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/pathology , Small Molecule Libraries/pharmacology , Bacteria/drug effects , Carrier Proteins/metabolism , Cell Aggregation/drug effects , Green Fluorescent Proteins/metabolism , HeLa Cells , High-Throughput Screening Assays , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Models, Biological , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/metabolism , Peptides/metabolism , Phenotype , Small Molecule Libraries/chemistry
15.
Cell Rep ; 11(12): 1905-18, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26095365

ABSTRACT

The polymorphism ATG16L1 T300A, associated with increased risk of Crohn's disease, impairs pathogen defense mechanisms including selective autophagy, but specific pathway interactions altered by the risk allele remain unknown. Here, we use perturbational profiling of human peripheral blood cells to reveal that CLEC12A is regulated in an ATG16L1-T300A-dependent manner. Antibacterial autophagy is impaired in CLEC12A-deficient cells, and this effect is exacerbated in the presence of the ATG16L1(∗)300A risk allele. Clec12a(-/-) mice are more susceptible to Salmonella infection, supporting a role for CLEC12A in antibacterial defense pathways in vivo. CLEC12A is recruited to sites of bacterial entry, bacteria-autophagosome complexes, and sites of sterile membrane damage. Integrated genomics identified a functional interaction between CLEC12A and an E3-ubiquitin ligase complex that functions in antibacterial autophagy. These data identify CLEC12A as early adaptor molecule for antibacterial autophagy and highlight perturbational profiling as a method to elucidate defense pathways in complex genetic disease.


Subject(s)
Carrier Proteins/genetics , Crohn Disease/genetics , Lectins, C-Type/genetics , Receptors, Mitogen/genetics , Salmonella Infections/genetics , Alleles , Animals , Autophagy/genetics , Autophagy-Related Proteins , Crohn Disease/microbiology , Crohn Disease/pathology , Genetic Predisposition to Disease , Genomics , Humans , Lectins, C-Type/biosynthesis , Mice , Receptors, Mitogen/biosynthesis , Risk Factors , Salmonella/pathogenicity , Salmonella Infections/microbiology
16.
PLoS One ; 9(9): e108777, 2014.
Article in English | MEDLINE | ID: mdl-25268627

ABSTRACT

The pathogenesis of complex diseases, such as type 1 diabetes (T1D), derives from interactions between host genetics and environmental factors. Previous studies have suggested that viral infection plays a significant role in initiation of T1D in genetically predisposed individuals. T1D susceptibility loci may therefore be enriched in previously uncharacterized genes functioning in antiviral defense pathways. To identify genes involved in antiviral immunity, we performed an image-based high-throughput genetic screen using short hairpin RNAs (shRNAs) against 161 genes within T1D susceptibility loci. RAW 264.7 cells transduced with shRNAs were infected with GFP-expressing herpes simplex virus type 1 (HSV-1) and fluorescent microscopy was performed to assess the viral infectivity by fluorescence reporter activity. Of the 14 candidates identified with high confidence, two candidates were selected for further investigation, Il27 and Tagap. Administration of recombinant IL-27 during viral infection was found to act synergistically with interferon gamma (IFN-γ) to activate expression of type I IFNs and proinflammatory cytokines, and to enhance the activities of interferon regulatory factor 3 (IRF3). Consistent with a role in antiviral immunity, Tagap-deficient macrophages demonstrated increased viral replication, reduced expression of proinflammatory chemokines and cytokines, and decreased production of IFN-ß. Taken together, our unbiased loss-of-function genetic screen identifies genes that play a role in host antiviral immunity and delineates roles for IL-27 and Tagap in the production of antiviral cytokines.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Immunity, Cellular , Animals , Cell Line , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Diabetes Mellitus, Type 1/pathology , Disease Susceptibility , GTPase-Activating Proteins/deficiency , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Genetic Loci , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , High-Throughput Screening Assays , Immunity, Cellular/drug effects , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Interferon-gamma/pharmacology , Interleukin-27/genetics , Interleukin-27/metabolism , Interleukin-27/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Microscopy, Fluorescence , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Virus Replication
17.
Autophagy ; 10(10): 1858-60, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25136803

ABSTRACT

Individuals who harbor a common coding polymorphism (Thr300Ala) within a structurally unclassified region of ATG16L1 are at increased risk for the development of Crohn disease. Recently, we reported on the generation and characterization of knockin mice carrying the ATG16L1 T300A variant. We demonstrate that multiple cell types from T300A knock-in mice exhibit reduced selective autophagy, and we mechanistically link this phenotype with an increased susceptibility of ATG16L1 T300A to CASP3- and CASP7-mediated cleavage. These findings demonstrate how a single polymorphism can result in cell type- and pathway-specific disruptions of selective autophagy and alterations in the inflammatory milieu that can contribute to disease.


Subject(s)
Carrier Proteins/metabolism , Crohn Disease/metabolism , Animals , Carrier Proteins/genetics , Crohn Disease/pathology , Humans , Mice , Models, Biological , Polymorphism, Single Nucleotide/genetics , Protein Stability , Signal Transduction
18.
Proc Natl Acad Sci U S A ; 111(21): 7741-6, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24821797

ABSTRACT

A coding polymorphism (Thr300Ala) in the essential autophagy gene, autophagy related 16-like 1 (ATG16L1), confers increased risk for the development of Crohn disease, although the mechanisms by which single disease-associated polymorphisms contribute to pathogenesis have been difficult to dissect given that environmental factors likely influence disease initiation in these patients. Here we introduce a knock-in mouse model expressing the Atg16L1 T300A variant. Consistent with the human polymorphism, T300A knock-in mice do not develop spontaneous intestinal inflammation, but exhibit morphological defects in Paneth and goblet cells. Selective autophagy is reduced in multiple cell types from T300A knock-in mice compared with WT mice. The T300A polymorphism significantly increases caspase 3- and caspase 7-mediated cleavage of Atg16L1, resulting in lower levels of full-length Atg16Ll T300A protein. Moreover, Atg16L1 T300A is associated with decreased antibacterial autophagy and increased IL-1ß production in primary cells and in vivo. Quantitative proteomics for protein interactors of ATG16L1 identified previously unknown nonoverlapping sets of proteins involved in ATG16L1-dependent antibacterial autophagy or IL-1ß production. These findings demonstrate how the T300A polymorphism leads to cell type- and pathway-specific disruptions of selective autophagy and suggest a mechanism by which this polymorphism contributes to disease.


Subject(s)
Carrier Proteins/genetics , Crohn Disease/immunology , Paneth Cells/pathology , Polymorphism, Single Nucleotide/genetics , Salmonella Infections/immunology , Animals , Autophagy/genetics , Autophagy-Related Proteins , Blotting, Western , Chromatography, Liquid , Crohn Disease/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Knock-In Techniques , Goblet Cells/pathology , Mice , Proteomics , Real-Time Polymerase Chain Reaction , Tandem Mass Spectrometry
19.
PLoS One ; 8(10): e77749, 2013.
Article in English | MEDLINE | ID: mdl-24204950

ABSTRACT

Attempts to eradicate HIV have been thwarted by the persistence of a small pool of quiescent memory CD4 T cells that harbor a transcriptionally silent, integrated form of the virus that can produce infectious virions following an anamnestic immune response. Transcription factors downstream of T-cell receptor activation, such as NF-κB/Rel and nuclear factor of activated T cells (NFAT) transcription members, are considered important regulators of HIV transcription during acute HIV infection. We now report studies exploring their precise role as antagonists of HIV latency using cell and primary CD4 T cell models of HIV-1 latency. Surprisingly, RNA interference studies performed in J-Lat CD4 T cells suggested that none of the NFATs, including NFATc1, NFATc2, NFATc3, and NFAT5, played a key role in the reactivation of latent HIV. However, cyclosporin A markedly inhibited the reactivation response. These results were reconciled when calcium signaling through calcineurin was shown to potentiate prostratin induced activation of NF-κB that in turn stimulated the latent HIV long terminal repeat (LTR). Similar effects of calcineurin were confirmed in a primary CD4 T cell model of HIV latency. These findings highlight an important role for calcineurin in NF-κB-dependent induction of latent HIV transcription. Innovative approaches exploiting the synergistic actions of calcineurin and prostratin in the absence of generalized T-cell activation merit exploration as a means to attack the latent viral reservoir.


Subject(s)
Calcineurin/metabolism , Calcium/metabolism , HIV Infections/immunology , HIV-1/immunology , NF-kappa B/metabolism , Phorbol Esters/pharmacology , Virus Latency/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Calcineurin/genetics , Calcineurin/immunology , Calcium/immunology , Calcium Signaling/genetics , Calcium Signaling/immunology , Cell Line, Tumor , HIV Infections/genetics , HIV Infections/metabolism , HIV Long Terminal Repeat/genetics , HIV Long Terminal Repeat/immunology , HIV-1/genetics , HIV-1/metabolism , Humans , Jurkat Cells , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , NF-kappa B/genetics , NF-kappa B/immunology , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , NFATC Transcription Factors/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism , Transcription, Genetic/genetics , Transcription, Genetic/immunology , Virus Latency/genetics
20.
ACS Chem Biol ; 8(12): 2724-2733, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-24168452

ABSTRACT

Autophagy is an evolutionarily conserved catabolic process that directs cytoplasmic proteins, organelles and microbes to lysosomes for degradation. Autophagy acts at the intersection of pathways involved in cellular stress, host defense, and modulation of inflammatory and immune responses; however, the details of how the autophagy network intersects with these processes remain largely undefined. Given the role of autophagy in several human diseases, it is important to determine the extent to which modulators of autophagy also modify inflammatory or immune pathways and whether it is possible to modulate a subset of these pathways selectively. Here, we identify small-molecule inducers of basal autophagy (including several FDA-approved drugs) and characterize their effects on IL-1ß production, autophagic engulfment and killing of intracellular bacteria, and development of Treg, TH17, and TH1 subsets from naïve T cells. Autophagy inducers with distinct, selective activity profiles were identified that reveal the functional architecture of connections between autophagy, and innate and adaptive immunity. In macrophages from mice bearing a conditional deletion of the essential autophagy gene Atg16L1, the small molecules inhibit IL-1ß production to varying degrees suggesting that individual compounds may possess both autophagy-dependent and autophagy-independent activity on immune pathways. The small molecule autophagy inducers constitute useful probes to test the contributions of autophagy-related pathways in diseases marked by impaired autophagy or elevated IL-1ß and to test novel therapeutic hypotheses.


Subject(s)
Adaptive Immunity/drug effects , Autophagy/drug effects , Immunity, Innate/drug effects , Immunologic Factors/pharmacology , Small Molecule Libraries/pharmacology , Animals , Autophagy/immunology , Autophagy-Related Proteins , Carrier Proteins/genetics , Carrier Proteins/immunology , Gene Expression Regulation , HeLa Cells , High-Throughput Screening Assays , Humans , Immunologic Factors/chemistry , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Signal Transduction , Small Molecule Libraries/chemistry , Structure-Activity Relationship , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells/cytology , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/drug effects , Th17 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...