Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
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.
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
3.
Cell ; 143(5): 789-801, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21111238

ABSTRACT

The mechanism by which CD4 T cells are depleted in HIV-infected hosts remains poorly understood. In ex vivo cultures of human tonsil tissue, CD4 T cells undergo a pronounced cytopathic response following HIV infection. Strikingly, >95% of these dying cells are not productively infected but instead correspond to bystander cells. We now show that the death of these "bystander" cells involves abortive HIV infection. Inhibitors blocking HIV entry or early steps of reverse transcription prevent CD4 T cell death while inhibition of later events in the viral life cycle does not. We demonstrate that the nonpermissive state exhibited by the majority of resting CD4 tonsil T cells leads to accumulation of incomplete reverse transcripts. These cytoplasmic nucleic acids activate a host defense program that elicits a coordinated proapoptotic and proinflammatory response involving caspase-3 and caspase-1 activation. While this response likely evolved to protect the host, it centrally contributes to the immunopathogenic effects of HIV.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/immunology , HIV-1/immunology , Lymphoid Tissue/virology , Apoptosis , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , DNA Replication , DNA, Viral/metabolism , HIV Reverse Transcriptase/metabolism , Humans , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology
4.
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
5.
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
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.
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
8.
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
9.
Gut ; 62(10): 1505-10, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24037875

ABSTRACT

Studies of the genetics underlying inflammatory bowel diseases have increased our understanding of the pathways involved in both ulcerative colitis and Crohn's disease and focused attention on the role of the microbiome in these diseases. Full understanding of pathogenesis will require a comprehensive grasp of the delicate homeostasis between gut bacteria and the human host. In this review, we present current evidence of microbiome-gene interactions in the context of other known risk factors and mechanisms, and describe the next steps necessary to pair genetic variant and microbiome sequencing data from patient cohorts. We discuss the concept of dysbiosis, proposing that the functional composition of the gut microbiome may provide a more consistent definition of dysbiosis and may more readily provide evidence of genome-microbiome interactions in future exploratory studies.


Subject(s)
Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/microbiology , Microbiota , Bacteria/immunology , Genetic Predisposition to Disease , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Inflammatory Bowel Diseases/immunology , Intestines/microbiology , Phenotype , Signal Transduction/genetics
10.
PLoS Pathog ; 7(8): e1002184, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21876670

ABSTRACT

The essential transactivator function of the HIV Tat protein is regulated by multiple posttranslational modifications. Although individual modifications are well characterized, their crosstalk and dynamics of occurrence during the HIV transcription cycle remain unclear.We examine interactions between two critical modifications within the RNA-binding domain of Tat: monomethylation of lysine 51 (K51) mediated by Set7/9/KMT7, an early event in the Tat transactivation cycle that strengthens the interaction of Tat with TAR RNA, and acetylation of lysine 50 (K50) mediated by p300/KAT3B, a later process that dissociates the complex formed by Tat, TAR RNA and the cyclin T1 subunit of the positive transcription elongation factor b (P-TEFb). We find K51 monomethylation inhibited in synthetic Tat peptides carrying an acetyl group at K50 while acetylation can occur in methylated peptides, albeit at a reduced rate. To examine whether Tat is subject to sequential monomethylation and acetylation in cells, we performed mass spectrometry on immunoprecipitated Tat proteins and generated new modification-specific Tat antibodies against monomethylated/acetylated Tat. No bimodified Tat protein was detected in cells pointing to a demethylation step during the Tat transactivation cycle. We identify lysine-specific demethylase 1 (LSD1/KDM1) as a Tat K51-specific demethylase, which is required for the activation of HIV transcription in latently infected T cells. LSD1/KDM1 and its cofactor CoREST associates with the HIV promoter in vivo and activate Tat transcriptional activity in a K51-dependent manner. In addition, small hairpin RNAs directed against LSD1/KDM1 or inhibition of its activity with the monoamine oxidase inhibitor phenelzine suppresses the activation of HIV transcription in latently infected T cells.Our data support the model that a LSD1/KDM1/CoREST complex, normally known as a transcriptional suppressor, acts as a novel activator of HIV transcription through demethylation of K51 in Tat. Small molecule inhibitors of LSD1/KDM1 show therapeutic promise by enforcing HIV latency in infected T cells.


Subject(s)
Histone Demethylases/metabolism , Transcription, Genetic/drug effects , tat Gene Products, Human Immunodeficiency Virus/metabolism , Acetylation , Animals , Epigenesis, Genetic/physiology , Genes, Viral/drug effects , Histone Demethylases/antagonists & inhibitors , Methylation , Phenelzine/pharmacology , Positive Transcriptional Elongation Factor B/metabolism , Rabbits , tat Gene Products, Human Immunodeficiency Virus/immunology
11.
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.

12.
J Biol Chem ; 285(38): 29326-35, 2010 Sep 17.
Article in English | MEDLINE | ID: mdl-20624919

ABSTRACT

Approximately half of all human genes undergo alternative mRNA splicing. This process often yields homologous gene products exhibiting diverse functions. Alternative splicing of APOBEC3G (A3G) and APOBEC3F (A3F), the major host resistance factors targeted by the HIV-1 protein Vif, has not been explored. We investigated the effects of alternative splicing on A3G/A3F gene expression and antiviral activity. Three alternatively spliced A3G mRNAs and two alternatively spliced A3F mRNAs were detected in peripheral blood mononuclear cells in each of 10 uninfected, healthy donors. Expression of these splice variants was altered in different cell subsets and in response to cellular stimulation. Alternatively spliced A3G variants were insensitive to degradation by Vif but displayed no antiviral activity against HIV-1. Conversely, alternative splicing of A3F produced a 37-kDa variant lacking exon 2 (A3FΔ2) that was prominently expressed in macrophages and monocytes and was resistant to Vif-mediated degradation. Alternative splicing also produced a 24-kDa variant of A3F lacking exons 2-4 (A3FΔ2-4) that was highly sensitive to Vif. Both A3FΔ2 and A3FΔ2-4 displayed reduced cytidine deaminase activity and moderate antiviral activity. These alternatively spliced A3F gene products, particularly A3FΔ2, were incorporated into HIV virions, albeit at levels less than wild-type A3F. Thus, alternative splicing of A3F mRNA generates truncated antiviral proteins that differ sharply in their sensitivity to Vif.


Subject(s)
Cytosine Deaminase/metabolism , HIV-1/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , Alternative Splicing/genetics , Alternative Splicing/physiology , Blotting, Western , Cell Line , Cytosine Deaminase/genetics , Humans , Protein Binding/genetics , Protein Binding/physiology , Reverse Transcriptase Polymerase Chain Reaction , vif Gene Products, Human Immunodeficiency Virus/genetics
13.
PLoS Pathog ; 5(4): e1000377, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19360131

ABSTRACT

Elite suppressors (ES) are a rare subset of HIV-1-infected individuals who are able to maintain HIV-1 viral loads below the limit of detection by ultra-sensitive clinical assays in the absence of antiretroviral therapy. Mechanism(s) responsible for this elite control are poorly understood but likely involve both host and viral factors. This study assesses ES plasma-derived envelope glycoprotein (env) fitness as a function of entry efficiency as a possible contributor to viral suppression. Fitness of virus entry was first evaluated using a novel inducible cell line with controlled surface expression levels of CD4 (receptor) and CCR5 (co-receptor). In the context of physiologic CCR5 and CD4 surface densities, ES envs exhibited significantly decreased entry efficiency relative to chronically infected viremic progressors. ES envs also demonstrated slow entry kinetics indicating the presence of virus with reduced entry fitness. Overall, ES env clones were less efficient at mediating entry than chronic progressor envs. Interestingly, acute infection envs exhibited an intermediate phenotypic pattern not distinctly different from ES or chronic progressor envs. These results imply that lower env fitness may be established early and may directly contribute to viral suppression in ES individuals.


Subject(s)
Gene Products, env/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/pathogenicity , Virus Internalization , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Flow Cytometry , Humans , Receptors, CCR5/immunology , Reverse Transcriptase Polymerase Chain Reaction , Viral Load
14.
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
15.
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
16.
PLoS Pathog ; 2(7): e68, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16839202

ABSTRACT

HIV-1 latency in resting CD4+ T cells represents a major barrier to virus eradication in patients on highly active antiretroviral therapy (HAART). We describe here a novel post-transcriptional block in HIV-1 gene expression in resting CD4+ T cells from patients on HAART. This block involves the aberrant localization of multiply spliced (MS) HIV-1 RNAs encoding the critical positive regulators Tat and Rev. Although these RNAs had no previously described export defect, we show that they exhibit strict nuclear localization in resting CD4+ T cells from patients on HAART. Overexpression of the transcriptional activator Tat from non-HIV vectors allowed virus production in these cells. Thus, the nuclear retention of MS HIV-1 RNA interrupts a positive feedback loop and contributes to the non-productive nature of infection of resting CD4+ T cells. To define the mechanism of nuclear retention, proteomic analysis was used to identify proteins that bind MS HIV-1 RNA. Polypyrimidine tract binding protein (PTB) was identified as an HIV-1 RNA-binding protein differentially expressed in resting and activated CD4+ T cells. Overexpression of PTB in resting CD4+ T cells from patients on HAART allowed cytoplasmic accumulation of HIV-1 RNAs. PTB overexpression also induced virus production by resting CD4+ T cells. Virus culture experiments showed that overexpression of PTB in resting CD4+ T cells from patients on HAART allowed release of replication-competent virus, while preserving a resting cellular phenotype. Whether through effects on RNA export or another mechanism, the ability of PTB to reverse latency without inducing cellular activation is a result with therapeutic implications.


Subject(s)
CD4-Positive T-Lymphocytes/chemistry , Cell Nucleus/chemistry , Gene Expression Regulation, Viral , HIV-1/genetics , RNA, Viral/analysis , Acquired Immunodeficiency Syndrome/genetics , Acquired Immunodeficiency Syndrome/physiopathology , Antiretroviral Therapy, Highly Active , CD4-Positive T-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/virology , Cell Nucleus/physiology , Cell Nucleus/virology , Gene Products, rev/analysis , Gene Products, rev/genetics , Gene Products, rev/physiology , Gene Products, tat/analysis , Gene Products, tat/genetics , Gene Products, tat/physiology , HIV-1/pathogenicity , HIV-1/physiology , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/physiology , Polypyrimidine Tract-Binding Protein/analysis , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/physiology , RNA Splicing , RNA, Viral/genetics , Virus Latency/physiology , Virus Replication/genetics , Virus Replication/physiology , rev Gene Products, Human Immunodeficiency Virus , tat Gene Products, Human Immunodeficiency Virus
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL