Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 31: 743-91, 2013.
Article in English | MEDLINE | ID: mdl-23330953

ABSTRACT

Dendritic cells (DCs) are specialized sentinels responsible for coordinating adaptive immunity. This function is dependent upon coupled sensitivity to environmental signs of inflammation and infection to cellular maturation-the programmed alteration of DC phenotype and function to enhance immune cell activation. Although DCs are thus well equipped to respond to pathogens, maturation triggers are not unique to infection. Given that immune cells are exquisitely sensitive to the biological functions of DCs, we now appreciate that multiple layers of suppression are required to restrict the environmental sensitivity, cellular maturation, and even life span of DCs to prevent aberrant immune activation during the steady state. At the same time, steady-state DCs are not quiescent but rather perform key functions that support homeostasis of numerous cell types. Here we review these functions and molecular mechanisms of suppression that control steady-state DC maturation. Corruption of these steady-state operatives has diverse immunological consequences and pinpoints DCs as potent drivers of autoimmune and inflammatory disease.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Homeostasis/immunology , Signal Transduction/immunology , Animals , Cell Differentiation/genetics , Dendritic Cells/metabolism , Homeostasis/genetics , Humans , Lectins, C-Type/physiology , Membrane Glycoproteins/physiology , Mice , Receptors, Immunologic/physiology , Receptors, Pattern Recognition/physiology , Signal Transduction/genetics , Toll-Like Receptors/physiology
2.
Cell ; 174(2): 271-284.e14, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29887373

ABSTRACT

The small intestinal tuft cell-ILC2 circuit mediates epithelial responses to intestinal helminths and protists by tuft cell chemosensory-like sensing and IL-25-mediated activation of lamina propria ILC2s. Small intestine ILC2s constitutively express the IL-25 receptor, which is negatively regulated by A20 (Tnfaip3). A20 deficiency in ILC2s spontaneously triggers the circuit and, unexpectedly, promotes adaptive small-intestinal lengthening and remodeling. Circuit activation occurs upon weaning and is enabled by dietary polysaccharides that render mice permissive for Tritrichomonas colonization, resulting in luminal accumulation of acetate and succinate, metabolites of the protist hydrogenosome. Tuft cells express GPR91, the succinate receptor, and dietary succinate, but not acetate, activates ILC2s via a tuft-, TRPM5-, and IL-25-dependent pathway. Also induced by parasitic helminths, circuit activation and small intestinal remodeling impairs infestation by new helminths, consistent with the phenomenon of concomitant immunity. We describe a metabolic sensing circuit that may have evolved to facilitate mutualistic responses to luminal pathosymbionts.


Subject(s)
Intestine, Small/physiology , Tritrichomonas/metabolism , Acetates/metabolism , Animals , Dietary Fiber/metabolism , Energy Metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/parasitology , Interleukins/genetics , Interleukins/metabolism , Intestinal Mucosa/cytology , Intestine, Small/microbiology , Intestine, Small/parasitology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microbiota , Plasmids/genetics , Plasmids/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Interleukin/metabolism , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/metabolism , Succinic Acid/metabolism , TRPM Cation Channels/metabolism , Tritrichomonas/growth & development , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism
3.
Nat Immunol ; 21(4): 422-433, 2020 04.
Article in English | MEDLINE | ID: mdl-32205880

ABSTRACT

A20 is an anti-inflammatory protein that is strongly linked to human disease. Here, we find that mice expressing three distinct targeted mutations of A20's zinc finger 7 (ZF7) ubiquitin-binding motif uniformly developed digit arthritis with features common to psoriatic arthritis, while mice expressing point mutations in A20's OTU or ZF4 motifs did not exhibit this phenotype. Arthritis in A20ZF7 mice required T cells and MyD88, was exquisitely sensitive to tumor necrosis factor and interleukin-17A, and persisted in germ-free conditions. A20ZF7 cells exhibited prolonged IκB kinase activity that drove exaggerated transcription of late-phase nuclear factor-κB response genes in vitro and in prediseased mouse paws in vivo. In addition, mice expressing double-mutant A20 proteins in A20's ZF4 and ZF7 motifs died perinatally with multi-organ inflammation. Therefore, A20's ZF4 and ZF7 motifs synergistically prevent inflammatory disease in a non-catalytic manner.


Subject(s)
Arthritis, Psoriatic/metabolism , Inflammation/metabolism , Ubiquitin/metabolism , Animals , Cells, Cultured , Interleukin-17 , Mice , Mice, Inbred C57BL , Mutation/genetics , NF-kappa B/metabolism , Protein Binding/physiology , Signal Transduction/physiology , Transcription, Genetic/physiology , Tumor Necrosis Factor-alpha/metabolism , Ubiquitination/physiology , Zinc Fingers/physiology
4.
Nat Immunol ; 21(5): 535-545, 2020 05.
Article in English | MEDLINE | ID: mdl-32313245

ABSTRACT

Despite evidence of chronic inflammation in myelodysplastic syndrome (MDS) and cell-intrinsic dysregulation of Toll-like receptor (TLR) signaling in MDS hematopoietic stem and progenitor cells (HSPCs), the mechanisms responsible for the competitive advantage of MDS HSPCs in an inflammatory milieu over normal HSPCs remain poorly defined. Here, we found that chronic inflammation was a determinant for the competitive advantage of MDS HSPCs and for disease progression. The cell-intrinsic response of MDS HSPCs, which involves signaling through the noncanonical NF-κB pathway, protected these cells from chronic inflammation as compared to normal HSPCs. In response to inflammation, MDS HSPCs switched from canonical to noncanonical NF-κB signaling, a process that was dependent on TLR-TRAF6-mediated activation of A20. The competitive advantage of TLR-TRAF6-primed HSPCs could be restored by deletion of A20 or inhibition of the noncanonical NF-κB pathway. These findings uncover the mechanistic basis for the clonal dominance of MDS HSPCs and indicate that interfering with noncanonical NF-κB signaling could prevent MDS progression.


Subject(s)
Hematopoietic Stem Cells/physiology , Inflammation/immunology , Myelodysplastic Syndromes/immunology , NF-kappa B/metabolism , TNF Receptor-Associated Factor 6/metabolism , Aged , Animals , Cell Differentiation , Cells, Cultured , Humans , Male , Mice , Mice, Transgenic , Myelopoiesis , NF-kappa B/genetics , Signal Transduction , TNF Receptor-Associated Factor 6/genetics , Toll-Like Receptors/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism
5.
Nat Immunol ; 16(6): 618-27, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25939025

ABSTRACT

A20 is an anti-inflammatory protein linked to multiple human diseases; however, the mechanisms by which A20 prevents inflammatory disease are incompletely defined. We found that A20-deficient T cells and fibroblasts were susceptible to caspase-independent and kinase RIPK3-dependent necroptosis. Global deficiency in RIPK3 significantly restored the survival of A20-deficient mice. A20-deficient cells exhibited exaggerated formation of RIPK1-RIPK3 complexes. RIPK3 underwent physiological ubiquitination at Lys5 (K5), and this ubiquitination event supported the formation of RIPK1-RIPK3 complexes. Both the ubiquitination of RIPK3 and formation of the RIPK1-RIPK3 complex required the catalytic cysteine of A20's deubiquitinating motif. Our studies link A20 and the ubiquitination of RIPK3 to necroptotic cell death and suggest additional mechanisms by which A20 might prevent inflammatory disease.


Subject(s)
Cysteine Endopeptidases/metabolism , Fibroblasts/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/physiology , Animals , Apoptosis/genetics , Catalytic Domain/genetics , Cysteine Endopeptidases/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/genetics , Necrosis/genetics , Protein Binding , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitination/genetics , Ubiquitins/metabolism
6.
Nat Immunol ; 15(3): 239-47, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24487321

ABSTRACT

Here we found that the transcription repressor DREAM bound to the promoter of the gene encoding A20 to repress expression of this deubiquitinase that suppresses inflammatory NF-κB signaling. DREAM-deficient mice displayed persistent and unchecked A20 expression in response to endotoxin. DREAM functioned by transcriptionally repressing A20 through binding to downstream regulatory elements (DREs). In contrast, binding of the transcription factor USF1 to the DRE-associated E-box domain in the gene encoding A20 activated its expression in response to inflammatory stimuli. Our studies define the critical opposing functions of DREAM and USF1 in inhibiting and inducing A20 expression, respectively, and thereby the strength of NF-κB signaling. Targeting of DREAM to induce USF1-mediated A20 expression is therefore a potential anti-inflammatory strategy for the treatment of diseases associated with unconstrained NF-κB activity, such as acute lung injury.


Subject(s)
DNA-Binding Proteins/biosynthesis , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Kv Channel-Interacting Proteins/metabolism , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/biosynthesis , Upstream Stimulatory Factors/metabolism , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , Animals , Chromatin Immunoprecipitation , Cysteine Endopeptidases , DNA-Binding Proteins/genetics , Disease Models, Animal , Gene Expression Regulation/immunology , Immunoblotting , Inflammation/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitin-Protein Ligases/genetics
7.
Immunity ; 46(3): 405-420, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28314591

ABSTRACT

During immune responses, naive T cells transition from small quiescent cells to rapidly cycling cells. We have found that T cells lacking TAX1BP1 exhibit delays in growth of cell size and cell cycling. TAX1BP1-deficient T cells exited G0 but stalled in S phase, due to both bioenergetic and biosynthetic defects. These defects were due to deficiencies in mTOR complex formation and activation. These mTOR defects in turn resulted from defective autophagy induction. TAX1BP1 binding of LC3 and GABARAP via its LC3-interacting region (LIR), but not its ubiquitin-binding domain, supported T cell proliferation. Supplementation of TAX1BP1-deficient T cells with metabolically active L-cysteine rescued mTOR activation and proliferation but not autophagy. These studies reveal that TAX1BP1 drives a specialized form of autophagy, providing critical amino acids that activate mTOR and enable the metabolic transition of activated T cells.


Subject(s)
Autophagosomes/immunology , Intracellular Signaling Peptides and Proteins/immunology , Lymphocyte Activation/immunology , Neoplasm Proteins/immunology , T-Lymphocytes/immunology , Animals , Autophagosomes/metabolism , Autophagy/immunology , Cell Separation , Chromosomes, Artificial, Bacterial , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/metabolism , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism
8.
FASEB J ; 38(13): e23757, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38965999

ABSTRACT

Hepatic stellate cells (HSCs) are responsible for liver fibrosis accompanied by its activation into myofibroblasts and the abundant production of extracellular matrix. However, the HSC contribution to progression of liver inflammation has been less known. We aimed to elucidate the mechanism in HSCs underlying the inflammatory response and the function of tumor necrosis factor α-related protein A20 (TNFAIP3). We established A20 conditional knockout (KO) mice crossing Twist2-Cre and A20 floxed mice. Using these mice, the effect of A20 was analyzed in mouse liver and HSCs. The human HSC line LX-2 was also used to examine the role and underlying molecular mechanism of A20. In this KO model, A20 was deficient in >80% of HSCs. Spontaneous inflammation with mild fibrosis was found in the liver of the mouse model without any exogenous agents, suggesting that A20 in HSCs suppresses chronic hepatitis. Comprehensive RNA sequence analysis revealed that A20-deficient HSCs exhibited an inflammatory phenotype and abnormally expressed chemokines. A20 suppressed JNK pathway activation in HSCs. Loss of A20 function in LX-2 cells also induced excessive chemokine expression, mimicking A20-deficient HSCs. A20 overexpression suppressed chemokine expression in LX-2. In addition, we identified DCLK1 in the genes regulated by A20. DCLK1 activated the JNK pathway and upregulates chemokine expression. DCLK1 inhibition significantly decreased chemokine induction by A20-silencing, suggesting that A20 controlled chemokine expression in HSCs via the DCLK1-JNK pathway. In conclusion, A20 suppresses chemokine induction dependent on the DCLK1-JNK signaling pathway. These findings demonstrate the therapeutic potential of A20 and the DCLK1-JNK pathway for the regulation of inflammation in chronic hepatitis.


Subject(s)
Chemokines , Hepatic Stellate Cells , MAP Kinase Signaling System , Mice, Knockout , Protein Serine-Threonine Kinases , Tumor Necrosis Factor alpha-Induced Protein 3 , Animals , Hepatic Stellate Cells/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Mice , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Chemokines/metabolism , Chemokines/genetics , Hepatitis, Chronic/metabolism , Hepatitis, Chronic/pathology , Hepatitis, Chronic/genetics , Doublecortin-Like Kinases , Mice, Inbred C57BL , Cell Line , Male
9.
Immunity ; 42(1): 55-67, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25607459

ABSTRACT

Inappropriate inflammasome activation contributes to multiple human diseases, but the mechanisms by which inflammasomes are suppressed are poorly understood. The NF-κB inhibitor A20 is a ubiquitin-modifying enzyme that might be critical in preventing human inflammatory diseases. Here, we report that A20-deficient macrophages, unlike normal cells, exhibit spontaneous NLRP3 inflammasome activity to LPS alone. The kinase RIPK3, but not the adaptor MyD88, is required for this response. In normal cells, A20 constitutively associates with caspase-1 and pro-IL-1ß, and NLRP3 activation further promotes A20 recruitment to the inflammasome. Pro-IL-1ß also co-immunoprecipitates with RIPK1, RIPK3, caspase-1, and caspase-8 in a complex that is modified with K63-linked and unanchored polyubiquitin. In A20-deficient macrophages, this pro-IL-1ß-associated ubiquitination is markedly increased in a RIPK3-dependent manner. Mass spectrometric and mutational analyses reveal that K133 of pro-IL-1ß is a physiological ubiquitination site that supports processing. Our study reveals a mechanism by which A20 prevents inflammatory diseases.


Subject(s)
Carrier Proteins/metabolism , Cysteine Endopeptidases/metabolism , Inflammasomes/immunology , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/physiology , Animals , Cell Line , Cysteine Endopeptidases/genetics , DNA Mutational Analysis , Immune Tolerance , Interleukin-1beta/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred Strains , Mice, Knockout , Multiprotein Complexes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitination/genetics
10.
Nat Immunol ; 12(12): 1184-93, 2011 Oct 23.
Article in English | MEDLINE | ID: mdl-22019834

ABSTRACT

Dendritic cells (DCs), which are known to support immune activation during infection, may also regulate immune homeostasis in resting animals. Here we show that mice lacking the ubiquitin-editing molecule A20 specifically in DCs spontaneously showed DC activation and population expansion of activated T cells. Analysis of DC-specific epistasis in compound mice lacking both A20 and the signaling adaptor MyD88 specifically in DCs showed that A20 restricted both MyD88-independent signals, which drive activation of DCs and T cells, and MyD88-dependent signals, which drive population expansion of T cells. In addition, mice lacking A20 specifically in DCs spontaneously developed lymphocyte-dependent colitis, seronegative ankylosing arthritis and enthesitis, conditions stereotypical of human inflammatory bowel disease (IBD). Our findings indicate that DCs need A20 to preserve immune quiescence and suggest that A20-dependent DC functions may underlie IBD and IBD-associated arthritides.


Subject(s)
Colitis/immunology , DNA-Binding Proteins/genetics , Dendritic Cells/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Spondylitis, Ankylosing/immunology , Ubiquitin-Protein Ligases/genetics , Animals , Colitis/pathology , Colitis/prevention & control , Crohn Disease/genetics , Cysteine Endopeptidases , DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Genetic Predisposition to Disease , Homeostasis/immunology , Humans , Lymphatic Diseases/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Differentiation Factor 88/metabolism , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Signal Transduction , Splenomegaly/genetics , Spondylitis, Ankylosing/pathology , Spondylitis, Ankylosing/prevention & control , T-Lymphocytes/immunology , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitin-Protein Ligases/metabolism
11.
Cell ; 134(5): 724-5, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18775305

ABSTRACT

Intestinal epithelial cells interact with both microbes in the gut lumen and host immune cells. In this issue, Kaser et al. (2008) link a key mediator of endoplasmic reticulum stress, the protein XBP1, with survival of intestinal secretory epithelial cells and inflammatory bowel disease.


Subject(s)
DNA-Binding Proteins/immunology , Endoplasmic Reticulum/immunology , Inflammatory Bowel Diseases/genetics , Intestinal Mucosa/immunology , Transcription Factors/immunology , Animals , Epithelial Cells/cytology , Humans , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/cytology , Regulatory Factor X Transcription Factors , X-Box Binding Protein 1
12.
Biochem Biophys Res Commun ; 592: 74-80, 2022 02 12.
Article in English | MEDLINE | ID: mdl-35032835

ABSTRACT

Crohn's disease is an inflammatory disease of the gut caused by a complex interplay among genetic, microbial, and environmental factors. The intestinal tract is constantly exposed to metals and other trace elements ingested as food. Synchrotron radiation-induced X-ray fluorescence spectroscopy and X-ray absorption fine structure analysis revealed the deposition of nickel particles within Crohn's disease tissue specimens. After nickel particle stimulation, THP-1 cells showed filopodia formation and autophagic vacuoles containing lipid bodies. Nickel particles precipitated colitis in mice bearing mutations of the IBD susceptibility protein A20/TNFAIP3. Nickel particles also exacerbated dextran sulfate sodium-induced colitis in mice harboring myeloid cell-specific Atg5 deficiency. These findings illustrate that nickel particle ingestion may worsen Crohn's disease by perturbing autophagic processes in the intestine, providing new insights into environmental factors in Crohn's disease pathogenesis.


Subject(s)
Crohn Disease/pathology , Disease Progression , Inflammation/pathology , Intestines/pathology , Nickel/toxicity , Animals , Autophagy/drug effects , Autophagy-Related Protein 5/metabolism , Dextran Sulfate , Disease Susceptibility , Humans , Macrophages/drug effects , Macrophages/pathology , Macrophages/ultrastructure , Mice, Inbred C57BL , THP-1 Cells , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism
14.
Immunity ; 38(5): 896-905, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23602765

ABSTRACT

A20 is an anti-inflammatory protein linked to multiple human autoimmune diseases and lymphomas. A20 possesses a deubiquitinating motif and a zinc finger, ZF4, that binds ubiquitin and supports its E3 ubiquitin ligase activity. To understand how these activities mediate A20's physiological functions, we generated two lines of gene-targeted mice, abrogating either A20's deubiquitinating activity (Tnfaip3(OTU) mice) or A20's ZF4 (Tnfaip3(ZF4) mice). Both Tnfaip3(OTU) and Tnfaip3(ZF4) mice exhibited increased responses to TNF and sensitivity to colitis. A20's C103 deubiquitinating motif restricted both K48- and K63-linked ubiquitination of receptor interacting protein 1 (RIP1). A20's ZF4 was required for recruiting A20 to ubiquitinated RIP1. A20(OTU) proteins and A20(ZF4) proteins complemented each other to regulate RIP1 ubiquitination and NFκB signaling normally in compound mutant Tnfaip3(OTU/ZF4) cells. This complementation involved homodimerization of A20 proteins, and we have defined an extensive dimerization interface in A20. These studies reveal how A20 proteins collaborate to restrict TNF signaling.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , GTPase-Activating Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cells, Cultured , Colitis/chemically induced , Colitis/genetics , Cysteine Endopeptidases , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Multimerization , Signal Transduction/genetics , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Zinc Fingers/genetics
15.
J Immunol ; 202(7): 2044-2056, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30760622

ABSTRACT

Deregulated immune response to a dysbiotic resident microflora within the oral cavity leads to chronic periodontal disease, local tissue destruction, and various systemic complications. To preserve tissue homeostasis, inflammatory signaling pathways involved in the progression of periodontitis must be tightly regulated. A20 (TNFAIP3), a ubiquitin-editing enzyme, has emerged as one of the key regulators of inflammation. Yet, the function of A20 in the oral mucosa and the biological pathways in which A20 mitigates periodontal inflammation remain elusive. Using a combination of in vivo and ex vivo disease models, we report in this study that A20 regulates inflammatory responses to a keystone oral bacterium, Porphyromonas gingivalis, and restrains periodontal inflammation through its effect on NF-κB signaling and cytokine production. Depletion of A20 using gene editing in human macrophage-like cells (THP-1) significantly increased cytokine secretion, whereas A20 overexpression using lentivirus infection dampened the cytokine production following bacterial challenge through modulating NF-κB activity. Similar to human cells, bone marrow-derived macrophages from A20-deficient mice infected with P. gingivalis displayed increased NF-κB activity and cytokine production compared with the cells isolated from A20-competent mice. Subsequent experiments using a murine ligature-induced periodontitis model showed that even a partial loss of A20 promotes an increased inflammatory phenotype and more severe bone loss, further verifying the critical function of A20 in the oral mucosa. Collectively, to our knowledge, these findings reveal the first systematic evidence of a physiological role for A20 in the maintenance of oral tissue homeostasis as a negative regulator of inflammation.


Subject(s)
Inflammation/immunology , Mouth Mucosa/immunology , NF-kappa B/immunology , Periodontitis/immunology , Tumor Necrosis Factor alpha-Induced Protein 3/immunology , Animals , HEK293 Cells , Humans , Immunity, Mucosal/immunology , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mouth Mucosa/metabolism , NF-kappa B/metabolism , Periodontitis/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism
17.
Dig Dis Sci ; 65(4): 1099-1106, 2020 04.
Article in English | MEDLINE | ID: mdl-31302808

ABSTRACT

AIMS: This pilot study assessed the efficacy, safety, and microbiome dynamics of fecal microbiota transplantation (FMT) for patients with chronic pouchitis. METHODS: A prospective open-label pilot study was performed at an academic center among pouchitis patients undergoing FMT. Patients received a minimum of a single FMT by pouchoscopy from healthy, screened donors. The primary outcome was clinical improvement in pouchitis assessed by patient survey at week 4. Secondary outcomes included decrease in total Pouchitis Disease Activity Index (PDAI) Score ≥ 3 at week 4, bowel movement frequency, ESR, CRP, fecal calprotectin, abdominal pain, and PDAI subscores including endoscopic and histologic changes. Stool samples were collected at baseline and 4 weeks post-FMT to assess bacterial microbiota using V4 16S rRNA sequencing. RESULTS: Nineteen patients were enrolled; however, 1 patient was lost to follow-up. No patients had a major adverse event or escalation of therapy related to FMT. Total PDAI scores, endoscopic scores, and histologic scores did not decrease significantly post-FMT. However, there was a statistically significant improvement in bowel movement (BM) frequency (9.25-7.25 BM/day, p = 0.03) and trend for improvement in abdominal pain to improve post-FMT (p = 0.05). Bacterial microbiota profiling revealed no distinct community-level changes post-FMT, though a small number of specific bacterial taxa significantly differed in relative abundance. CONCLUSIONS: A single FMT has a tolerable short-term safety profile and may be associated with a decrease in bowel movements in patients with chronic pouchitis; however, no robust endoscopic or histologic changes were observed.


Subject(s)
Endoscopy, Gastrointestinal/methods , Fecal Microbiota Transplantation/methods , Gastrointestinal Microbiome/physiology , Pouchitis/diagnosis , Pouchitis/therapy , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Pilot Projects , Pouchitis/microbiology , Prospective Studies , Young Adult
18.
J Biol Chem ; 293(47): 18285-18295, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30282802

ABSTRACT

OTUB1 is a deubiquitinating enzyme that cleaves Lys-48-linked polyubiquitin chains and also regulates ubiquitin signaling through a unique, noncatalytic mechanism. OTUB1 binds to a subset of E2 ubiquitin-conjugating enzymes and inhibits their activity by trapping the E2∼ubiquitin thioester and preventing ubiquitin transfer. The same set of E2s stimulate the deubiquitinating activity of OTUB1 when the E2 is not charged with ubiquitin. Previous studies have shown that, in cells, OTUB1 binds to E2-conjugating enzymes of the UBE2D (UBCH5) and UBE2E families, as well as to UBE2N (UBC13). Cellular roles have been identified for the interaction of OTUB1 with UBE2N and members of the UBE2D family, but not for interactions with UBE2E E2 enzymes. We report here a novel role for OTUB1-E2 interactions in modulating E2 protein ubiquitination. We observe that Otub1-/- knockout mice exhibit late-stage embryonic lethality. We find that OTUB1 depletion dramatically destabilizes the E2-conjugating enzyme UBE2E1 (UBCH6) in both mouse and human OTUB1 knockout cell lines. Of note, this effect is independent of the catalytic activity of OTUB1, but depends on its ability to bind to UBE2E1. We show that OTUB1 suppresses UBE2E1 autoubiquitination in vitro and in cells, thereby preventing UBE2E1 from being targeted to the proteasome for degradation. Taken together, we provide evidence that OTUB1 rescues UBE2E1 from degradation in vivo.


Subject(s)
Cysteine Endopeptidases/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Amino Acid Motifs , Animals , Cysteine Endopeptidases/genetics , Deubiquitinating Enzymes , Mice , Mice, Inbred C57BL , Protein Binding , Protein Stability , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitination
19.
Cell Immunol ; 340: 103914, 2019 06.
Article in English | MEDLINE | ID: mdl-31030956

ABSTRACT

A20, also known as TNFAIP3, is a potent regulator of ubiquitin (Ub) dependent signals. A20 prevents multiple human diseases, indicating that the critical functions of this protein are clinically as well as biologically impactful. As revealed by mouse models, cell specific functions of A20 are linked to its ability to regulate diverse signaling pathways. Aberrant expression or functions of A20 in specific cell types underlie divergent disease outcomes. Discernment of A20's biochemical functions and their phenotypic outcomes will contribute to our understanding of how ubiquitination is regulated, how Ub mediated functions can prevent disease, and will pave the way for future therapeutic interventions.


Subject(s)
Arthritis, Rheumatoid/immunology , Asthma/immunology , Lupus Erythematosus, Systemic/immunology , Protein Processing, Post-Translational , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/prevention & control , Asthma/genetics , Asthma/pathology , Asthma/prevention & control , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Disease Models, Animal , Humans , Immunity, Innate/drug effects , Immunologic Factors/therapeutic use , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Lupus Erythematosus, Systemic/prevention & control , Molecular Targeted Therapy/methods , NF-kappa B/genetics , NF-kappa B/immunology , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Necrosis Factor alpha-Induced Protein 3/immunology , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL