151
|
Lee IT, Lin CC, Wu YC, Yang CM. TNF-alpha induces matrix metalloproteinase-9 expression in A549 cells: role of TNFR1/TRAF2/PKCalpha-dependent signaling pathways. J Cell Physiol 2010; 224:454-64. [PMID: 20333651 DOI: 10.1002/jcp.22142] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Matrix metalloproteinases (MMPs), in particular MMP-9, have been shown to be induced by cytokines, including TNF-alpha and contributes to airway inflammation. However, the mechanisms underlying TNF-alpha-induced MMP-9 expression in human A549 cells remain unclear. Here, we report that TNF-alpha-induced MMP-9 gene expression was mediated through the TNFR1/TRAF2/PKCalpha-dependent signaling pathways in A549 cells, determined by zymographic, RT-PCR, and Western blotting analyses. TNF-alpha-induced MMP-9 expression was reduced by pretreatment with a TNFR Ab. Furthermore, TNF-alpha-induced TNFR1 and TRAF2 complex formation was revealed by immunoprecipitation using an anti-TNFR1 Ab followed by Western blot analysis against an anti-TRAF2 or anti-TNFR1 Ab. In addition, TNF-alpha-induced MMP-9 expression was also reduced by pretreatment with the inhibitor of PKCalpha (Gö6983), c-Src (PP1), EGFR (AG1478), or PI3K (LY294002) or transfection with siRNAs of PKCalpha, Src, EGFR, Akt, p65, p300, and c-Jun. On the other hand, TNF-alpha stimulated the phosphorylation of c-Src, EGFR, Akt, JNK1/2, and c-Jun, which were inhibited by pretreatment with Gö6983. We also showed that TNF-alpha induced Akt translocation and the formation of an Akt/p65/p300 complex. Pretreatment with the inhibitor of JNK1/2 (SP600125) but not the inhibitor of MEK1/2 (U0126), p38 MAPK (SB202190), or PI3K (LY294002), markedly inhibited TNF-alpha-induced c-Jun mRNA levels. Taken together, these data suggest that in A549 cells, TNF-alpha induces MMP-9 expression via the TNFR1/TRAF2/PKCalpha-dependent JNK1/2/c-Jun and c-Src/EGFR/PI3K/Akt pathways.
Collapse
Affiliation(s)
- I-Ta Lee
- Department of Physiology and Pharmacology, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | | | | | | |
Collapse
|
152
|
Lodolce JP, Kolodziej LE, Rhee L, Kariuki SN, Franek BS, McGreal NM, Logsdon MF, Bartulis SJ, Perera MA, Ellis NA, Adams EJ, Hanauer SB, Jolly M, Niewold TB, Boone DL. African-derived genetic polymorphisms in TNFAIP3 mediate risk for autoimmunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:7001-9. [PMID: 20483768 PMCID: PMC3307531 DOI: 10.4049/jimmunol.1000324] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The TNF alpha-induced protein 3 (TNFAIP3) is an ubiquitin-modifying enzyme and an essential negative regulator of inflammation. Genome-wide association studies have implicated the TNFAIP3 locus in susceptibility to autoimmune disorders in European cohorts, including rheumatoid arthritis, coronary artery disease, psoriasis, celiac disease, type 1 diabetes, inflammatory bowel disease, and systemic lupus erythematosus (SLE). There are two nonsynonymous coding polymorphisms in the deubiquitinating (DUB) domain of TNFAIP3: F127C, which is in high-linkage disequilibrium with reported SLE-risk variants, and A125V, which has not been previously studied. We conducted a case-control study in African-American SLE patients using these coding variants, along with tagging polymorphisms in TNFAIP3, and identified a novel African-derived risk haplotype that is distinct from previously reported risk variants (odds ratio=1.6, p=0.006). In addition, a rare protective haplotype was defined by A125V (odds ratio=0.31, p=0.027). Although A125V was associated with protection from SLE, surprisingly the same allele was associated with increased risk of inflammatory bowel disease. We tested the functional activity of nonsynonymous coding polymorphisms within TNFAIP3, and found that the A125V coding-change variant alters the DUB activity of the protein. Finally, we used computer modeling to depict how the A125V amino acid change in TNFAIP3 may affect the three-dimensional structure of the DUB domain to a greater extent than F127C. This is the first report of an association between TNFAIP3 polymorphisms and autoimmunity in African-Americans.
Collapse
Affiliation(s)
| | | | - Lesley Rhee
- Department of Medicine, University of Chicago
| | | | | | | | | | | | | | | | - Erin J. Adams
- Department of Biochemistry and Molecular Biophysics, University of Chicago
| | | | | | | | | |
Collapse
|
153
|
Zinc decreases C-reactive protein, lipid peroxidation, and inflammatory cytokines in elderly subjects: a potential implication of zinc as an atheroprotective agent. Am J Clin Nutr 2010; 91:1634-41. [PMID: 20427734 PMCID: PMC2869512 DOI: 10.3945/ajcn.2009.28836] [Citation(s) in RCA: 270] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Chronic inflammation and oxidative stress are common risk factors for atherosclerosis. Zinc is an essential micronutrient that can function as an antiinflammatory and antioxidative agent, and as such, it may have atheroprotective properties. OBJECTIVE We hypothesized that zinc down-regulates the production of atherosclerosis-related cytokines/molecules in humans. DESIGN To examine these effects, we conducted a randomized, double-blinded, placebo trial of zinc supplementation in elderly subjects. We recruited 40 healthy elderly subjects (aged 56-83 y) and randomly assigned them to 2 groups. One group was given an oral dose of 45 mg zinc/d as a gluconate for 6 mo. The other group was given a placebo. Cell culture models were conducted to study the mechanism of zinc as an atheroprotective agent. RESULTS After 6 mo of supplementation, the intake of zinc, compared with intake of placebo, increased the concentrations of plasma zinc and decreased the concentrations of plasma high-sensitivity C-reactive protein (hsCRP), interleukin (IL)-6, macrophage chemoattractant protein 1 (MCP-1), vascular cell adhesion molecule 1 (VCAM-1), secretory phospholipase A2, and malondialdehyde and hydroxyalkenals (MDA+HAE) in elderly subjects. Regression analysis showed that changes in concentrations of plasma zinc were inversely associated with changes in concentrations of plasma hsCRP, MCP-1, VCAM-1, and MDA+HAE after 6 mo of supplementation. In cell culture studies, we showed that zinc decreased the generation of tumor necrosis factor-alpha, IL-1beta, VCAM-1, and MDA+HAE and the activation of nuclear transcription factor kappaB and increased antiinflammatory proteins A20 and peroxisome proliferator-activated receptor-alpha in human monocytic leukemia THP-1 cells and human aortic endothelial cells compared with zinc-deficient cells. CONCLUSION These findings suggest that zinc may have a protective effect in atherosclerosis because of its antiinflammatory and antioxidant functions.
Collapse
|
154
|
Dichotomous Actions of NF-κB Signaling Pathways in Heart. J Cardiovasc Transl Res 2010; 3:344-54. [DOI: 10.1007/s12265-010-9195-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Accepted: 05/04/2010] [Indexed: 12/28/2022]
|
155
|
Missiou A, Köstlin N, Varo N, Rudolf P, Aichele P, Ernst S, Münkel C, Walter C, Stachon P, Sommer B, Pfeifer D, Zirlik K, MacFarlane L, Wolf D, Tsitsikov E, Bode C, Libby P, Zirlik A. Tumor necrosis factor receptor-associated factor 1 (TRAF1) deficiency attenuates atherosclerosis in mice by impairing monocyte recruitment to the vessel wall. Circulation 2010; 121:2033-44. [PMID: 20421522 PMCID: PMC2995263 DOI: 10.1161/circulationaha.109.895037] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Members of the tumor necrosis factor superfamily, such as tumor necrosis factor-alpha, potently promote atherogenesis in mice and humans. Tumor necrosis factor receptor-associated factors (TRAFs) are cytoplasmic adaptor proteins for this group of cytokines. METHODS AND RESULTS This study tested the hypothesis that TRAF1 modulates atherogenesis in vivo. TRAF1(-/-)/LDLR(-/-) mice that consumed a high-cholesterol diet for 18 weeks developed significantly smaller atherosclerotic lesions than LDLR(-/-) (LDL receptor-deficient) control animals. As the most prominent change in histological composition, plaques of TRAF1-deficient animals contained significantly fewer macrophages. Bone marrow transplantations revealed that TRAF1 deficiency in both hematopoietic and vascular resident cells contributed to the reduction in atherogenesis observed. Mechanistic studies showed that deficiency of TRAF1 in endothelial cells and monocytes reduced adhesion of inflammatory cells to the endothelium in static and dynamic assays. Impaired adhesion coincided with reduced cell spreading, actin polymerization, and CD29 expression in macrophages, as well as decreased expression of the adhesion molecules intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 in endothelial cells. Small interfering RNA studies in human cells verified these findings. Furthermore, TRAF1 messenger RNA levels were significantly elevated in the blood of patients with acute coronary syndrome. CONCLUSIONS TRAF1 deficiency attenuates atherogenesis in mice, most likely owing to impaired monocyte recruitment to the vessel wall. These data identify TRAF1 as a potential treatment target for atherosclerosis.
Collapse
Affiliation(s)
- Anna Missiou
- Department of Cardiology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Germany
- Faculty of Biology, University of Freiburg, Germany
| | - Natascha Köstlin
- Department of Cardiology, University of Freiburg, Freiburg, Germany
| | - Nerea Varo
- Department of Clinical Chemistry, University of Navarra, Pamplona, Spain
| | - Philipp Rudolf
- Department of Cardiology, University of Freiburg, Freiburg, Germany
| | - Peter Aichele
- Institute for Medical Microbiology and Hygiene, Department of Immunology, University of Freiburg, Germany
| | - Sandra Ernst
- Department of Cardiology, University of Freiburg, Freiburg, Germany
| | - Christian Münkel
- Department of Cardiology, University of Freiburg, Freiburg, Germany
| | - Carina Walter
- Department of Cardiology, University of Freiburg, Freiburg, Germany
| | - Peter Stachon
- Department of Cardiology, University of Freiburg, Freiburg, Germany
| | - Benjamin Sommer
- Department of Cardiology, University of Freiburg, Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Hematology and Oncology, University of Freiburg, Freiburg, Germany
| | - Katja Zirlik
- Department of Hematology and Oncology, University of Freiburg, Freiburg, Germany
| | - Lindsey MacFarlane
- Donald W. Reynolds Cardiovascular Research Center, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Dennis Wolf
- Department of Cardiology, University of Freiburg, Freiburg, Germany
| | | | - Christoph Bode
- Department of Cardiology, University of Freiburg, Freiburg, Germany
| | - Peter Libby
- Donald W. Reynolds Cardiovascular Research Center, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Andreas Zirlik
- Department of Cardiology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Germany
| |
Collapse
|
156
|
Abstract
Clinicians have suspected for hundreds of years that chronic activation of the immune system contributes to the development of cancer. However, the molecular mechanisms that mediate this precarious interplay are only now being elucidated. Recent reports have identified A20 as a crucial tumour suppressor in various lymphomas. A20 is a ubiquitin-editing enzyme that attenuates the activity of proximal signalling complexes at pro-inflammatory receptors. In this Review we summarize the evidence linking chronic inflammation with tumorigenesis and consider how A20 modulates inflammatory signalling cascades, thereby providing a mechanism to explain how deregulation of ubiquitylation can promote tumorigenesis.
Collapse
Affiliation(s)
- Sarah G Hymowitz
- Department of Structural Biology, Genentech, Inc.1 DNA Way, M/S 40, South San Francisco, CA 94080, USA
| | | |
Collapse
|
157
|
Abstract
The stimulation of TLR4 by LPS activates two distinct signaling pathways leading to the expression of diverse inflammatory genes. Intensive studies over the past decade have revealed the components involved in these signaling pathways, however, more recently the focus has shifted somewhat towards the components that regulate these pathways. Several regulatory mechanisms, including localisation of components, splice variants and inhibitory molecules will be discussed in this review.
Collapse
Affiliation(s)
- Anne F McGettrick
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland.
| | | |
Collapse
|
158
|
Ha H, Han D, Choi Y. TRAF-mediated TNFR-family signaling. CURRENT PROTOCOLS IN IMMUNOLOGY 2009; Chapter 11:11.9D.1-11.9D.19. [PMID: 19918944 DOI: 10.1002/0471142735.im1109ds87] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The tumor necrosis factor (TNF) superfamily consists of a wide variety of cell-bound and secreted proteins that regulate numerous cellular processes. In particular, TNF-family proteins regulate the proliferation and death of tumor cells, as well as activated immune cells. This overview discusses the mammalian TNF receptor-associated factors (TRAFs), of which TRAF1, 2, 3, 5, and 6 have been shown to interact directly or indirectly with members of the TNF receptor superfamily. Structural features of TRAF proteins are described along with a discussion of TRAF-interacting proteins and the signaling pathways activated by the TRAF proteins. Finally, we examine the phenotypes observed in TRAF-knockout mice.
Collapse
Affiliation(s)
- Hyunil Ha
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Daehee Han
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Yongwon Choi
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
159
|
Barton A, Worthington J. Genetic susceptibility to rheumatoid arthritis: An emerging picture. ACTA ACUST UNITED AC 2009; 61:1441-6. [DOI: 10.1002/art.24672] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
160
|
Schumacher MA, Schmitz R, Brune V, Tiacci E, Döring C, Hansmann ML, Siebert R, Küppers R. Mutations in the genes coding for the NF-κB regulating factors IκBα and A20 are uncommon in nodular lymphocyte-predominant Hodgkin's lymphoma. Haematologica 2009; 95:153-7. [PMID: 19648161 DOI: 10.3324/haematol.2009.010157] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Nodular lymphocyte-predominant Hodgkin's lymphoma (NLPHL) shows constitutive NF-kappaB activity in the malignant lymphocyte-predominant (LP) cells. Constitutive NF-kappaB activity also plays a central pathogenetic role in classical Hodgkin's lymphoma (cHL), where inactivating mutations in the NFKBIA and TNFAIP3 genes, coding for the negative NF-kappaB regulators IkappaBalpha and A20, respectively, contribute to NF-kappaB activation. To determine whether mutations in NFKBIA and TNFAIP3 are also involved in the pathogenesis of NLPHL these genes were sequenced from microdissected LP cells of 10 primary NLPHL. We also studied DEV, the only cell line proposedly derived from LP cells, after we had confirmed its derivation from NLPHL by gene expression analysis. A heterozygous somatic missense mutation in the NFKBIA gene was found in one NLPHL, and a heterozygous, possibly subclonal, two base pair insertion in TNFAIP3 in another case. The low mutation frequency and the absence of biallelic destructive mutations propose a minor contribution of NFKBIA and TNFAIP3 mutations to the NF-kappaB activity of NLPHL, suggesting different mechanisms of NF-kappaB activation in NLPHL and cHL.
Collapse
Affiliation(s)
- Martin A Schumacher
- Institute of Cell Biology (Cancer Research), Medical School, University of Duisburg-Essen, Essen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
161
|
Folco EJ, Rocha VZ, López-Ilasaca M, Libby P. Adiponectin inhibits pro-inflammatory signaling in human macrophages independent of interleukin-10. J Biol Chem 2009; 284:25569-75. [PMID: 19617629 DOI: 10.1074/jbc.m109.019786] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Macrophages participate pivotally in the pathogenesis of many chronic inflammatory diseases including atherosclerosis. Adiponectin, a vasculoprotective molecule with insulin-sensitizing and anti-atherogenic properties, suppresses pro-inflammatory gene expression in macrophages by mechanisms that remain incompletely understood. This study investigated the effects of adiponectin on major pro-inflammatory signaling pathways in human macrophages. We demonstrate that pretreatment of these cells with adiponectin inhibits phosphorylation of nuclear factor kappaB inhibitor (IkappaB), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (MAPK), induced by either lipopolysaccharide (LPS) or tumor necrosis factor (TNF) alpha, as well as STAT3 phosphorylation induced by interleukin-6 (IL6). Antagonism of IL10 by either neutralizing antibodies or siRNA-mediated silencing did not abrogate the anti-inflammatory actions of adiponectin, indicating that the ability of adiponectin to render human macrophages tolerant to various pro-inflammatory stimuli does not require this cytokine. A systematic search for adiponectin-inducible genes with established anti-inflammatory properties revealed that adiponectin augmented the expression of A20, suppressor of cytokine signaling (SOCS) 3, B-cell CLL/lymphoma (BCL) 3, TNF receptor-associated factor (TRAF) 1, and TNFAIP3-interacting protein (TNIP) 3. These results suggest that adiponectin triggers a multifaceted response in human macrophages by inducing the expression of various anti-inflammatory proteins that act at different levels in concert to suppress macrophage activation.
Collapse
Affiliation(s)
- Eduardo J Folco
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
162
|
Wang J, Ouyang Y, Guner Y, Ford HR, Grishin AV. Ubiquitin-editing enzyme A20 promotes tolerance to lipopolysaccharide in enterocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 183:1384-92. [PMID: 19570823 PMCID: PMC2739219 DOI: 10.4049/jimmunol.0803987] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Although enterocytes are capable of innate immune responses, the intestinal epithelium is normally tolerant to commensal bacteria. To elucidate the mechanisms of tolerance, we examined the effect of preexposure to LPS on activation of p38, c-Jun, and NF-kappaB in enterocytes by several inflammatory and stress stimuli. Shortly after the initial LPS challenge, enterocytes become tolerant to restimulation with LPS or CpG DNA, but not with IL-17 or UV. The state of tolerance, which lasts 20-26 h, temporally coincides with LPS-induced expression of the anti-inflammatory ubiquitin-editing enzyme A20. Small interfering RNA silencing of A20 prevents tolerance, whereas ectopic expression of A20 blocks responses to LPS and CpG DNA, but not to IL-17 or UV. A20 levels in the epithelium of the small intestine are low at birth and following gut decontamination with antibiotics, but high under conditions of bacterial colonization. In the small intestine of adult rodents, A20 prominently localizes to the luminal interface of villus enterocytes. Lower parts of the crypts display relatively low levels of A20, but relatively high levels of phospho-p38. Gut decontamination with antibiotics reduces the levels of both A20 and phospho-p38. Along with the fact that A20-deficient mice develop severe intestinal inflammation, our results indicate that induction of A20 plays a key role in the tolerance of the intestinal epithelium to TLR ligands and bacteria.
Collapse
Affiliation(s)
- Jin Wang
- Division of Pediatric Surgery, Childrens Hospital Los Angeles, CA90027, USA
| | - Yannan Ouyang
- Division of Pathology, Childrens Hospital Los Angeles, CA90027, USA
| | - Yigit Guner
- Division of Pediatric Surgery, Childrens Hospital Los Angeles, CA90027, USA
- Department of Surgery, University of Southern California, 4661 Sunset Boulevard, Los Angeles, CA90027, USA
| | - Henri R. Ford
- Division of Pediatric Surgery, Childrens Hospital Los Angeles, CA90027, USA
- Department of Surgery, University of Southern California, 4661 Sunset Boulevard, Los Angeles, CA90027, USA
| | - Anatoly V. Grishin
- Division of Pediatric Surgery, Childrens Hospital Los Angeles, CA90027, USA
- Department of Surgery, University of Southern California, 4661 Sunset Boulevard, Los Angeles, CA90027, USA
| |
Collapse
|
163
|
Kang NI, Yoon HY, Lee YR, Won M, Chung MJ, Park JW, Hur GM, Lee HK, Park BH. A20 attenuates allergic airway inflammation in mice. THE JOURNAL OF IMMUNOLOGY 2009; 183:1488-95. [PMID: 19553520 DOI: 10.4049/jimmunol.0900163] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
TNF receptor 1 can activate signaling pathways leading to the activation of NF-kappaB. A20, an NF-kappaB-inducible protein, negatively regulates these signaling pathways and acts as an anti-inflammatory mediator. Therefore, A20 is viewed as a potential therapeutic target for inflammatory disease. In this study, we examined the effect of A20 on an OVA-induced allergic airway inflammation model in mice. We used an adenovirus containing A20 cDNA (Ad-A20) that was delivered intratracheally before OVA challenge. Single administration of Ad-A20 reduced airway inflammatory cell recruitment and peribronchiolar inflammation and suppressed the production of various cytokines in bronchoalveolar fluid. In addition, Ad-A20 suppressed mucus production and prevented the development of airway hyperresponsiveness. The protective effect of Ad-A20 was mediated by the inhibition of the NF-kappaB signaling pathway. Taken together, our results suggest that the development of an immunoregulatory strategy based on A20 may have therapeutic potential for the treatment of allergic asthma.
Collapse
Affiliation(s)
- Nam-In Kang
- Department of Immunology, Medical School and Diabetes Research Center, Chonbuk National University, Jeonju, Jeonbuk, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Gao Z, Yin J, Zhang J, He Q, McGuinness OP, Ye J. Inactivation of NF-kappaB p50 leads to insulin sensitization in liver through post-translational inhibition of p70S6K. J Biol Chem 2009; 284:18368-76. [PMID: 19433583 DOI: 10.1074/jbc.m109.007260] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
In this study, we investigated the metabolic phenotype of the NF-kappaB p50 knock-out (p50-KO) mice. Compared with wild type mice, the p50-KO mice had an increase in food intake, but a decrease in body fat content. On chow diet, their blood glucose dropped much more than the wild type (WT) mice in the insulin tolerance test. Their glucose infusion rate was 30% higher than that of the WT mice in the hyperinsulinemic-euglycemic clamp. Their hepatic glucose production was suppressed more actively by insulin, and their insulin-induced glucose uptake was not altered in skeletal muscle or adipose tissue. In the liver, their p70S6K (S6K1) protein was significantly lower, and tumor necrosis factor-alpha (TNF-alpha) expression was much higher. Their S6K1 protein was reduced by TNF-alpha treatment in the primary culture of hepatocytes. S6K1 reduction was blocked by the proteasome inhibitor MG132. In their livers, IKK2 (IKKbeta) activity was reduced together with IKKgamma. Their S6K1 degradation was dependent on IKK2 deficiency. Reconstitution of the S6K1 protein in their liver blocked the increase in insulin sensitivity. S6K1 degradation was not observed in hepatocytes of the WT mice. The data suggest that inactivation of NF-kappaB p50 leads to suppression of IKK2 activity in the liver. IKK2 deficiency leads to S6K1 inhibition through TNF-induced protein degradation. The S6K1 reduction may contribute to insulin sensitivity in p50-KO mice. This study suggests that hepatic S6K1 may be a drug target in the treatment of insulin resistance.
Collapse
Affiliation(s)
- Zhanguo Gao
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808, USA
| | | | | | | | | | | |
Collapse
|
165
|
Frequent inactivation of A20 in B-cell lymphomas. Nature 2009; 459:712-6. [PMID: 19412163 DOI: 10.1038/nature07969] [Citation(s) in RCA: 448] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Accepted: 03/03/2009] [Indexed: 12/24/2022]
Abstract
A20 is a negative regulator of the NF-kappaB pathway and was initially identified as being rapidly induced after tumour-necrosis factor-alpha stimulation. It has a pivotal role in regulation of the immune response and prevents excessive activation of NF-kappaB in response to a variety of external stimuli; recent genetic studies have disclosed putative associations of polymorphic A20 (also called TNFAIP3) alleles with autoimmune disease risk. However, the involvement of A20 in the development of human cancers is unknown. Here we show, using a genome-wide analysis of genetic lesions in 238 B-cell lymphomas, that A20 is a common genetic target in B-lineage lymphomas. A20 is frequently inactivated by somatic mutations and/or deletions in mucosa-associated tissue lymphoma (18 out of 87; 21.8%) and Hodgkin's lymphoma of nodular sclerosis histology (5 out of 15; 33.3%), and, to a lesser extent, in other B-lineage lymphomas. When re-expressed in a lymphoma-derived cell line with no functional A20 alleles, wild-type A20, but not mutant A20, resulted in suppression of cell growth and induction of apoptosis, accompanied by downregulation of NF-kappaB activation. The A20-deficient cells stably generated tumours in immunodeficient mice, whereas the tumorigenicity was effectively suppressed by re-expression of A20. In A20-deficient cells, suppression of both cell growth and NF-kappaB activity due to re-expression of A20 depended, at least partly, on cell-surface-receptor signalling, including the tumour-necrosis factor receptor. Considering the physiological function of A20 in the negative modulation of NF-kappaB activation induced by multiple upstream stimuli, our findings indicate that uncontrolled signalling of NF-kappaB caused by loss of A20 function is involved in the pathogenesis of subsets of B-lineage lymphomas.
Collapse
|
166
|
Malynn BA, Ma A. A20 takes on tumors: tumor suppression by an ubiquitin-editing enzyme. ACTA ACUST UNITED AC 2009; 206:977-80. [PMID: 19380636 PMCID: PMC2715039 DOI: 10.1084/jem.20090765] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Many B cell cancers are characterized in part by the dysregulation of the NF-kappaB signaling pathway. A new study identifies somatic mutations in TNFAIP3, the gene encoding the NF-kappaB inhibitor A20, in Hodgkin lymphomas and primary mediastinal lymphomas. These data reveal the role of A20 as a tumor suppressor protein.
Collapse
Affiliation(s)
- Barbara A Malynn
- Program in Biomedical Sciences, Program in Biological Sciences, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | | |
Collapse
|
167
|
Schmitz R, Hansmann ML, Bohle V, Martin-Subero JI, Hartmann S, Mechtersheimer G, Klapper W, Vater I, Giefing M, Gesk S, Stanelle J, Siebert R, Küppers R. TNFAIP3 (A20) is a tumor suppressor gene in Hodgkin lymphoma and primary mediastinal B cell lymphoma. ACTA ACUST UNITED AC 2009; 206:981-9. [PMID: 19380639 PMCID: PMC2715030 DOI: 10.1084/jem.20090528] [Citation(s) in RCA: 373] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Proliferation and survival of Hodgkin and Reed/Sternberg (HRS) cells, the malignant cells of classical Hodgkin lymphoma (cHL), are dependent on constitutive activation of nuclear factor kappaB (NF-kappaB). NF-kappaB activation through various stimuli is negatively regulated by the zinc finger protein A20. To determine whether A20 contributes to the pathogenesis of cHL, we sequenced TNFAIP3, encoding A20, in HL cell lines and laser-microdissected HRS cells from cHL biopsies. We detected somatic mutations in 16 out of 36 cHLs (44%), including missense mutations in 2 out of 16 Epstein-Barr virus-positive (EBV(+)) cHLs and a missense mutation, nonsense mutations, and frameshift-causing insertions or deletions in 14 out of 20 EBV(-) cHLs. In most mutated cases, both TNFAIP3 alleles were inactivated, including frequent chromosomal deletions of TNFAIP3. Reconstitution of wild-type TNFAIP3 in A20-deficient cHL cell lines revealed a significant decrease in transcripts of selected NF-kappaB target genes and caused cytotoxicity. Extending the mutation analysis to primary mediastinal B cell lymphoma (PMBL), another lymphoma with constitutive NF-kappaB activity, revealed destructive mutations in 5 out of 14 PMBLs (36%). This report identifies TNFAIP3 (A20), a key regulator of NF-kappaB activity, as a novel tumor suppressor gene in cHL and PMBL. The significantly higher frequency of TNFAIP3 mutations in EBV(-) than EBV(+) cHL suggests complementing functions of TNFAIP3 inactivation and EBV infection in cHL pathogenesis.
Collapse
Affiliation(s)
- Roland Schmitz
- Institute of Cell Biology (Cancer Research), Medical School, University of Duisburg-Essen, 45122 Essen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Abstract
Inappropriate functioning of the immune system is linked to immune deficiency, autoimmune disease, and cancer. It is therefore not surprising that intracellular immune signaling pathways are tightly controlled. One of the best studied transcription factors in immune signaling is NF-kappaB, which is activated by multiple receptors and regulates the expression of a wide variety of proteins that control innate and adaptive immunity. A20 is an early NF-kappaB-responsive gene that encodes a ubiquitin-editing protein that is involved in the negative feedback regulation of NF-kappaB signaling. Here, we discuss the mechanism of action of A20 and its role in the regulation of inflammation and immunity.
Collapse
|
169
|
Nedelcu AM. Comparative Genomics of Phylogenetically Diverse Unicellular Eukaryotes Provide New Insights into the Genetic Basis for the Evolution of the Programmed Cell Death Machinery. J Mol Evol 2009; 68:256-68. [DOI: 10.1007/s00239-009-9201-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Revised: 12/21/2008] [Accepted: 01/12/2009] [Indexed: 11/30/2022]
|
170
|
Li L, Soetandyo N, Wang Q, Ye Y. The zinc finger protein A20 targets TRAF2 to the lysosomes for degradation. BIOCHIMICA ET BIOPHYSICA ACTA 2009; 1793:346-53. [PMID: 18952128 PMCID: PMC2657479 DOI: 10.1016/j.bbamcr.2008.09.013] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 09/09/2008] [Accepted: 09/17/2008] [Indexed: 11/22/2022]
Abstract
The zinc finger-containing protein A20 is a negative regulator of TNF-induced JNK (c-Jun-N-terminal kinase) and NFkappaB (nuclear factor kappaB) signaling. A20 is an unusual enzyme that contains both ubiquitinating and deubiquitinating activities. Although A20 is mostly localized in the cytosol, our recent studies reveal that a fraction of A20 can associate with a lysosome-interacting compartment in a manner that requires its carboxy terminal zinc fingers, but independent of its ubiquitin modifying activities. Whether the lysosome-associated A20 has a function in cellular signaling is unclear. Here, we demonstrate that A20 is capable of targeting an associated signaling molecule such as TRAF2 to the lysosomes for degradation. This process is dependent on the membrane tethering zinc finger domains of A20, but does not require A20 ubiquitin modifying activity. Our findings suggest a novel mode of A20 action that involves lysosomal targeting of signal molecules bound to A20.
Collapse
Affiliation(s)
- Lianyun Li
- Building 5, Room 433, Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health 5, center drive, Bethesda, MD 20892, USA
| | - Nia Soetandyo
- Building 5, Room 433, Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health 5, center drive, Bethesda, MD 20892, USA
| | - Qiuyan Wang
- Building 5, Room 433, Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health 5, center drive, Bethesda, MD 20892, USA
| | - Yihong Ye
- Building 5, Room 433, Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health 5, center drive, Bethesda, MD 20892, USA
| |
Collapse
|
171
|
Raychaudhuri S, Remmers EF, Lee AT, Hackett R, Guiducci C, Burtt NP, Gianniny L, Korman BD, Padyukov L, Kurreeman FAS, Chang M, Catanese JJ, Ding B, Wong S, van der Helm-van Mil AHM, Neale BM, Coblyn J, Cui J, Tak PP, Wolbink GJ, Crusius JBA, van der Horst-Bruinsma IE, Criswell LA, Amos CI, Seldin MF, Kastner DL, Ardlie KG, Alfredsson L, Costenbader KH, Altshuler D, Huizinga TWJ, Shadick NA, Weinblatt ME, de Vries N, Worthington J, Seielstad M, Toes REM, Karlson EW, Begovich AB, Klareskog L, Gregersen PK, Daly MJ, Plenge RM. Common variants at CD40 and other loci confer risk of rheumatoid arthritis. Nat Genet 2008; 40:1216-23. [PMID: 18794853 PMCID: PMC2757650 DOI: 10.1038/ng.233] [Citation(s) in RCA: 422] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2008] [Accepted: 07/10/2008] [Indexed: 01/15/2023]
Abstract
To identify rheumatoid arthritis risk loci in European populations, we conducted a meta-analysis of two published genome-wide association (GWA) studies totaling 3,393 cases and 12,462 controls. We genotyped 31 top-ranked SNPs not previously associated with rheumatoid arthritis in an independent replication of 3,929 autoantibody-positive rheumatoid arthritis cases and 5,807 matched controls from eight separate collections. We identified a common variant at the CD40 gene locus (rs4810485, P = 0.0032 replication, P = 8.2 x 10(-9) overall, OR = 0.87). Along with other associations near TRAF1 (refs. 2,3) and TNFAIP3 (refs. 4,5), this implies a central role for the CD40 signaling pathway in rheumatoid arthritis pathogenesis. We also identified association at the CCL21 gene locus (rs2812378, P = 0.00097 replication, P = 2.8 x 10(-7) overall), a gene involved in lymphocyte trafficking. Finally, we identified evidence of association at four additional gene loci: MMEL1-TNFRSF14 (rs3890745, P = 0.0035 replication, P = 1.1 x 10(-7) overall), CDK6 (rs42041, P = 0.010 replication, P = 4.0 x 10(-6) overall), PRKCQ (rs4750316, P = 0.0078 replication, P = 4.4 x 10(-6) overall), and KIF5A-PIP4K2C (rs1678542, P = 0.0026 replication, P = 8.8 x 10(-8) overall).
Collapse
Affiliation(s)
- Soumya Raychaudhuri
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Abstract
Protein modification by ubiquitin and ubiquitin-like molecules is a critical regulatory process. Like most regulated protein modifications, ubiquitination is reversible. Deubiquitination, the reversal of ubiquitination, is quickly being recognized as an important regulatory strategy. Nearly one hundred human DUBs (deubiquitinating enzymes) in five different gene families oppose the action of several hundred ubiquitin ligases, suggesting that both ubiquitination and its reversal are highly regulated and specific processes. It has long been recognized that ubiquitin ligases are modular enzyme systems that often depend on scaffolds and adaptors to deliver substrates to the catalytically active macromolecular complex. Although many DUBs bind ubiquitin with reasonable affinities (in the nM to microM range), a larger number have little affinity but exhibit robust catalytic capability. Thus it is apparent that these DUBs must acquire their substrates by binding the target protein in a conjugate or by associating with other macromolecular complexes. We would then expect that a study of protein partners of DUBs would reveal a variety of substrates, scaffolds, adaptors and ubiquitin receptors. In the present review we suggest that, like ligases, much of the regulation and specificity of deubiquitination arises from the association of DUBs with these protein partners.
Collapse
Affiliation(s)
- Karen H. Ventii
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
| | - Keith D. Wilkinson
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
173
|
Beyond tumor necrosis factor receptor: TRADD signaling in toll-like receptors. Proc Natl Acad Sci U S A 2008; 105:12429-34. [PMID: 18719121 DOI: 10.1073/pnas.0806585105] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Tumor necrosis factor receptor 1-associated death domain protein (TRADD) is the core adaptor recruited to TNF receptor 1 (TNFR1) upon TNFalpha stimulation. In cells from TRADD-deficient mice, TNFalpha-mediated apoptosis and TNFalpha-stimulated NF-kappaB, JNK, and ERK activation are defective. TRADD is also important for germinal center formation, DR3-mediated costimulation of T cells, and TNFalpha-mediated inflammatory responses in vivo. TRADD deficiency does not enhance IFNgamma-induced signaling. Importantly, TRADD has a novel role in TLR3 and TLR4 signaling. TRADD participates in the TLR4 complex formed upon LPS stimulation, and TRADD-deficient macrophages show impaired cytokine production in response to TLR ligands in vitro. Thus, TRADD is a multifunctional protein crucial both for TNFR1 signaling and other signaling pathways relevant to immune responses.
Collapse
|
174
|
Prasad AS. Zinc in human health: effect of zinc on immune cells. Mol Med 2008; 14:353-7. [PMID: 18385818 DOI: 10.2119/2008-00033.prasad] [Citation(s) in RCA: 492] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Accepted: 03/24/2008] [Indexed: 11/06/2022] Open
Abstract
Although the essentiality of zinc for plants and animals has been known for many decades, the essentiality of zinc for humans was recognized only 40 years ago in the Middle East. The zinc-deficient patients had severe immune dysfunctions, inasmuch as they died of intercurrent infections by the time they were 25 years of age. In our studies in an experimental human model of zinc deficiency, we documented decreased serum testosterone level, oligospermia, severe immune dysfunctions mainly affecting T helper cells, hyperammonemia, neurosensory disorders, and decreased lean body mass. It appears that zinc deficiency is prevalent in the developing world and as many as two billion subjects may be growth retarded due to zinc deficiency. Besides growth retardation and immune dysfunctions, cognitive impairment due to zinc deficiency also has been reported recently. Our studies in the cell culture models showed that the activation of many zinc-dependent enzymes and transcription factors were adversely affected due to zinc deficiency. In HUT-78 (T helper 0 [Th(0)] cell line), we showed that a decrease in gene expression of interleukin-2 (IL-2) and IL-2 receptor alpha(IL-2Ralpha) were due to decreased activation of nuclear factor-kappaB (NF-kappaB) in zinc deficient cells. Decreased NF-kappaB activation in HUT-78 due to zinc deficiency was due to decreased binding of NF-kappaB to DNA, decreased level of NF-kappaB p105 (the precursor of NF-kappaB p50) mRNA, decreased kappaB inhibitory protein (IkappaB) phosphorylation, and decreased Ikappa kappa. These effects of zinc were cell specific. Zinc also is an antioxidant and has anti-inflammatory actions. The therapeutic roles of zinc in acute infantile diarrhea, acrodermatitis enteropathica, prevention of blindness in patients with age-related macular degeneration, and treatment of common cold with zinc have been reported. In HL-60 cells (promyelocytic leukemia cell line), zinc enhances the up-regulation of A20 mRNA, which, via TRAF pathway, decreases NF-kappaB activation, leading to decreased gene expression and generation of tumor necrosis factor-alpha (TNF-alpha), IL-1beta, and IL-8. We have reported recently that in both young adults and elderly subjects, zinc supplementation decreased oxidative stress markers and generation of inflammatory cytokines.
Collapse
Affiliation(s)
- Ananda S Prasad
- Wayne State University School of Medicine, Detroit, Michigan 48201, United States of America.
| |
Collapse
|
175
|
Prasad AS. Zinc in human health: effect of zinc on immune cells. MOLECULAR MEDICINE (CAMBRIDGE, MASS.) 2008. [PMID: 18385818 DOI: 10.2119/2008-00033] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although the essentiality of zinc for plants and animals has been known for many decades, the essentiality of zinc for humans was recognized only 40 years ago in the Middle East. The zinc-deficient patients had severe immune dysfunctions, inasmuch as they died of intercurrent infections by the time they were 25 years of age. In our studies in an experimental human model of zinc deficiency, we documented decreased serum testosterone level, oligospermia, severe immune dysfunctions mainly affecting T helper cells, hyperammonemia, neurosensory disorders, and decreased lean body mass. It appears that zinc deficiency is prevalent in the developing world and as many as two billion subjects may be growth retarded due to zinc deficiency. Besides growth retardation and immune dysfunctions, cognitive impairment due to zinc deficiency also has been reported recently. Our studies in the cell culture models showed that the activation of many zinc-dependent enzymes and transcription factors were adversely affected due to zinc deficiency. In HUT-78 (T helper 0 [Th(0)] cell line), we showed that a decrease in gene expression of interleukin-2 (IL-2) and IL-2 receptor alpha(IL-2Ralpha) were due to decreased activation of nuclear factor-kappaB (NF-kappaB) in zinc deficient cells. Decreased NF-kappaB activation in HUT-78 due to zinc deficiency was due to decreased binding of NF-kappaB to DNA, decreased level of NF-kappaB p105 (the precursor of NF-kappaB p50) mRNA, decreased kappaB inhibitory protein (IkappaB) phosphorylation, and decreased Ikappa kappa. These effects of zinc were cell specific. Zinc also is an antioxidant and has anti-inflammatory actions. The therapeutic roles of zinc in acute infantile diarrhea, acrodermatitis enteropathica, prevention of blindness in patients with age-related macular degeneration, and treatment of common cold with zinc have been reported. In HL-60 cells (promyelocytic leukemia cell line), zinc enhances the up-regulation of A20 mRNA, which, via TRAF pathway, decreases NF-kappaB activation, leading to decreased gene expression and generation of tumor necrosis factor-alpha (TNF-alpha), IL-1beta, and IL-8. We have reported recently that in both young adults and elderly subjects, zinc supplementation decreased oxidative stress markers and generation of inflammatory cytokines.
Collapse
Affiliation(s)
- Ananda S Prasad
- Wayne State University School of Medicine, Detroit, Michigan 48201, United States of America.
| |
Collapse
|
176
|
Werner SL, Kearns JD, Zadorozhnaya V, Lynch C, O’Dea E, Boldin MP, Ma A, Baltimore D, Hoffmann A. Encoding NF-kappaB temporal control in response to TNF: distinct roles for the negative regulators IkappaBalpha and A20. Genes Dev 2008; 22:2093-101. [PMID: 18676814 PMCID: PMC2492747 DOI: 10.1101/gad.1680708] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Accepted: 06/16/2008] [Indexed: 11/25/2022]
Abstract
TNF-induced NF-kappaB activity shows complex temporal regulation whose different phases lead to distinct gene expression programs. Combining experimental studies and mathematical modeling, we identify two temporal amplification steps-one determined by the obligate negative feedback regulator IkappaBalpha-that define the duration of the first phase of NF-kappaB activity. The second phase is defined by A20, whose inducible expression provides for a rheostat function by which other inflammatory stimuli can regulate TNF responses. Our results delineate the nonredundant functions implied by the knockout phenotypes of ikappabalpha and a20, and identify the latter as a signaling cross-talk mediator controlling inflammatory and developmental responses.
Collapse
Affiliation(s)
- Shannon L. Werner
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, University of California at San Diego, La Jolla, California 92093, USA
| | - Jeffrey D. Kearns
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, University of California at San Diego, La Jolla, California 92093, USA
| | - Victoria Zadorozhnaya
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, University of California at San Diego, La Jolla, California 92093, USA
| | - Candace Lynch
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, University of California at San Diego, La Jolla, California 92093, USA
| | - Ellen O’Dea
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, University of California at San Diego, La Jolla, California 92093, USA
| | - Mark P. Boldin
- Division of Biology, California Institute of Technology, Pasadena, California 91125, USA
| | - Averil Ma
- Biomedical Sciences Division, University of California at San Francisco, San Francisco, California 94143, USA
| | - David Baltimore
- Division of Biology, California Institute of Technology, Pasadena, California 91125, USA
| | - Alexander Hoffmann
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, University of California at San Diego, La Jolla, California 92093, USA
| |
Collapse
|
177
|
Barton A, Thomson W, Ke X, Eyre S, Hinks A, Bowes J, Gibbons L, Plant D, Wellcome Trust Case Control Consortium, Wilson AG, Marinou I, Morgan A, Emery P, YEAR consortium, Steer S, Hocking L, Reid DM, Wordsworth P, Harrison P, Worthington J. Re-evaluation of putative rheumatoid arthritis susceptibility genes in the post-genome wide association study era and hypothesis of a key pathway underlying susceptibility. Hum Mol Genet 2008; 17:2274-9. [PMID: 18434327 PMCID: PMC2465799 DOI: 10.1093/hmg/ddn128] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2008] [Accepted: 04/15/2008] [Indexed: 12/11/2022] Open
Abstract
Rheumatoid arthritis (RA) is an archetypal, common, complex autoimmune disease with both genetic and environmental contributions to disease aetiology. Two novel RA susceptibility loci have been reported from recent genome-wide and candidate gene association studies. We, therefore, investigated the evidence for association of the STAT4 and TRAF1/C5 loci with RA using imputed data from the Wellcome Trust Case Control Consortium (WTCCC). No evidence for association of variants mapping to the TRAF1/C5 gene was detected in the 1860 RA cases and 2930 control samples tested in that study. Variants mapping to the STAT4 gene did show evidence for association (rs7574865, P = 0.04). Given the association of the TRAF1/C5 locus in two previous large case-control series from populations of European descent and the evidence for association of the STAT4 locus in the WTCCC study, single nucleotide polymorphisms mapping to these loci were tested for association with RA in an independent UK series comprising DNA from >3000 cases with disease and >3000 controls and a combined analysis including the WTCCC data was undertaken. We confirm association of the STAT4 and the TRAF1/C5 loci with RA bringing to 5 the number of confirmed susceptibility loci. The effect sizes are less than those reported previously but are likely to be a more accurate reflection of the true effect size given the larger size of the cohort investigated in the current study.
Collapse
Affiliation(s)
- Anne Barton
- Arc-Epidemiology Unit, Stopford Building, The University of Manchester, Manchester, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
Collaborators
Paul R Burton, David G Clayton, Lon R Cardon, Nick Craddock, Panos Deloukas, Audrey Duncanson, Dominic P Kwiatkowski, Mark I McCarthy, Willem H Ouwehand, Nilesh J Samani, John A Todd, Peter Donnelly, Jeffrey C Barrett, Paul R Burton, Dan Davison, Peter Donnelly, Doug Easton, David Evans, Hin-Tak Leung, Jonathan L Marchini, Andrew P Morris, Chris C A Spencer, Martin D Tobin, Lon R Cardon, David G Clayton, Antony P Attwood, James P Boorman, Barbara Cant, Ursula Everson, Judith M Hussey, Jennifer D Jolley, Alexandra S Knight, Kerstin Koch, Elizabeth Meech, Sarah Nutland, Christopher V Prowse, Helen E Stevens, Niall C Taylor, Graham R Walters, Neil M Walker, Nicholas A Watkins, Thilo Winzer, John A Todd, Willem H Ouwehand, Richard W Jones, Wendy L McArdle, Susan M Ring, David P Strachan, Marcus Pembrey, Gerome Breen, David St Clair, Sian Caesar, Katherine Gordon-Smith, Lisa Jones, Christine Fraser, Elaine K Green, Detelina Groze, Marian L Hamshere, Peter A Holmans, Ian R Jones, George Kirov, Valentina Moskvina, Ivan Nikolov, Michael C O'Donovan, Michel J Owen, Nick Craddock, David A Collier, Amanda Elkin, Anne Farmer, Richard Williamson, Peter McGuffin, Allan H Young, I Nicol Ferrier, Stephen G Ball, Anthony J Balmforth, Jennifer H Barrett, D Timothy Bishop, Mark M Iles, Azhar Maqbool, Nadira Yuldasheva, Alistair S Hall, Peter S Braund, Paul R Burton, Richard J Dixon, Massimo Mangino, Suzanne Stevens, Martin D Tobin, John R Thompson, Nilesh J Samani, Francesca Bredin, Mark Tremelling, Miles Parkes, Hazel Drummond, Charles W Lees, Elaine R Nimmo, Jack Satsangi, Sheila A Fisher, Alastair Forbes, Cathryn Lewis, Clive M Onnie, Natalie J Prescott, Jeremy Sanderson, Christopher G Mathew, Jamie Barbour, M Khalid Mohiuddin, Catherine E Todhunter, John C Mansfield, Tariq Ahmad, Fraser R Cummings, Derek P Jewell, John Webster, Morris J Brown, David G Clayton, G Mark Lathrop, John Connell, Anna Dominiczak, Nilesh J Samani, Carolina A Braga Marcano, Beverley Burke, Richard Dobson, Johannie Gungadoo, Kate L Lee, Patricia B Munroe, Stephen J Newhouse, Abiodun Onipinla, Chris Wallace, Mingzhan Xue, Mark Caulfield, Martin Farrall, Anne Barton, The Biologics in RA Genetics and Genomics Study Syndicate BRAGGS, Ian N Bruce, Hannah Donovan, Steve Eyre, Paul D Gilbert, Samantha L Hider, Anne M Hinks, Sally L John, Catherine Potter, Alan J Silman, Deborah P M Symmons, Wendy Thomson, Jane Worthington, David G Clayton, David B Dunger, Sarah Nutland, Helen E Stevens, Neil M Walker, Barry Widmer, John A Todd, Timothy M Frayling, Rachel M Freathy, Hana Lango, John R B Perry, Beverley M Shields, Michael N Weedon, Andrew T Hattersley, Graham A Hitman, Mark Walker, Kate S Elliott, Christopher J Groves, Cecilia M Lindgren, Nigel W Rayner, Nicholas J Timpson, Eleftheria Zeggini, Mark I McCarthy, Melanie Newport, Giorgio Sirugo, Emily Lyons, Fredrik Vannberg, Adrian V S Hill, Linda A Bradbury, Claire Farrar, Jennifer J Pointon, Paul Wordsworth, Matthew A Brown, Jayne A Franklyn, Joanne M Heward, Matthew J Simmonds, Stephen C L Gough, Sheila Seal, Breast Cancer Susceptibility Collaboration, Michael R Stratton, Nazneen Rahman, Maria Ban, An Goris, Stephen J Sawcer, Alastair Compston, David Conway, Miminatou Jallow, Melanie Newport, Giorgio Sirugo, Kirk A Rockett, Dominic P Kwiatkowski, Suzannah J Bumpstead, Amy Chaney, Kate Downes, Mohammed J R Ghori, Rhian Gwilliam, Sarah E Hunt, Michael Inouye, Andrew Keniry, Emma King, Ralph McGinnis, Simon Potters, Rathi Ravindrarajah, Pamela Whittaker, Claire Widden, David Withers, Panos Deloukas, Hin-Tak Leung, Sarah Nutland, Helen E Stevens, Neil M Walker, John A Todd, Doug Easton, David G Clayton, Paul R Burton, Martin D Tobin, Jeffrey C Barrett, David Evans, Andrew P Morris, Lon R Cardon, Niall J Cardin, Dan Davison, Teresa Ferreira, Joanne Pereira-Gale, Ingileif B Hallgrimsdóttir, Bryan N Howie, Jonathan L Marchini, Chris C A Spencer, Zhan Su, Yik Ying Teo, Damjan Vukcevic, Peter Donnelly, David Bentley, Matthew A Brown, Lon R Cardon, Mark Caulfield, David G Clayton, Alistair Compston, Nick Craddock, Panos Deloukas, Peter Donnelly, Martin Farrall, Stephen C L Gough, Alistair S Hall, Andrew T Hattersley, Adrian V S Hill, Dominic P Kwiatkowski, Christopher G Mathew, Mark I McCarthy, Willem H Ouwehand, Miles Parkes, Marcus Pembrey, Nazneen Rahman, Nilesh J Samani, Michael R Stratton, John A Todd, Jane Worthington, Paul Emery, Philip Conaghan, Mark Quinn, Ann Morgan, Anne-Maree Keenan, Elizabeth Hensor, Julie Kitcheman, Andrew Gough, Michael Green, Richard Reece, Lesley Hordon, Philip Helliwell, Richard Melsom, Sheelagh Doherty, Ade Adebajo, Andrew Harvey, Steve Jarret, Gareth Huson, Amanda Isdale, Mike Martin, Zinaid Karim, Dennis McGonagle, Colin Pease, Sally Cox, Victoria Bejarano, Jackie Nam, Claire Brown, Christine Thomas, David Pickles, Alison Hammond, Beverley Neville, Alan Fairclough, Caroline Nunns, Anne Gill, Julie Green, Belinda Rhys-Evans, Barbara Padwell, Julie Madden, Lynda Taylor, Sally Smith, Heather King, Jill Firth, Jayne Heard, Linda Sigsworth, Diane Corscadden, Karen Henshaw, Lubna-Haroon Rashid, Stephen G Martin, James I Robinson,
Collapse
|
178
|
Li L, Hailey DW, Soetandyo N, Li W, Lippincott-Schwartz J, Shu HB, Ye Y. Localization of A20 to a lysosome-associated compartment and its role in NFkappaB signaling. BIOCHIMICA ET BIOPHYSICA ACTA 2008; 1783:1140-9. [PMID: 18329387 PMCID: PMC2587335 DOI: 10.1016/j.bbamcr.2008.01.029] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/12/2007] [Revised: 01/08/2008] [Accepted: 01/22/2008] [Indexed: 11/26/2022]
Abstract
A20 is a tumor necrosis factor (TNF)-inducible zinc finger protein that contains both ubiquitinating and deubiquitinating activities. A20 negatively regulates NFkappaB (nuclear factor kappaB) signaling induced by TNF receptor family and Toll-like receptors, but the mechanism of A20 action is poorly defined. Here we show that a fraction of endogenous and ectopically expressed A20 is localized to an endocytic membrane compartment that is in association with the lysosome. The lysosomal association of A20 requires its carboxy terminal zinc finger domains, but is independent of its ubiquitin-modifying activities. Interestingly, A20 mutants defective in membrane association also contain reduced NFkappaB inhibitory activity. These findings suggest the involvement of a lysosome-associated mechanism in A20-dependent termination of NFkappaB signaling.
Collapse
Affiliation(s)
- Lianyun Li
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dale W. Hailey
- Cell Biology and Metabolism Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nia Soetandyo
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Li
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jennifer Lippincott-Schwartz
- Cell Biology and Metabolism Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hong-bing Shu
- College of Life Sciences, Wuhan University, Wuhan 430072, P.R. China
| | - Yihong Ye
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
179
|
Li H, Lin X. Positive and negative signaling components involved in TNFalpha-induced NF-kappaB activation. Cytokine 2008; 41:1-8. [PMID: 18068998 DOI: 10.1016/j.cyto.2007.09.016] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2007] [Accepted: 09/28/2007] [Indexed: 01/01/2023]
Abstract
Tumor Necrosis Factor alpha (TNFalpha) is a pro-inflammatory cytokine that plays important roles in different biological processes, including the induction of other cytokines. One of the most important downstream signaling targets activated by TNFalpha is the NF-kappaB transcription factor, which has been identified to be involved in inflammatory, anti-apoptotic, and immune responses. Stimulation of cells with TNFalpha triggers activation of NF-kappaB through various signaling molecules, including TRAF2, RIP, MAP3K, and the IKK complex. Recently, numerous studies have been performed to explore the detailed mechanism by which NF-kappaB is activated upon TNFalpha stimulation. Current understanding of this pathway has been focused on the identification of signaling components, the role of post-translational modification and the sub-cellular translocation of those components. Additionally, more negative regulators in the TNF-IKK pathway are emerging.
Collapse
Affiliation(s)
- Hongxiu Li
- Department of Molecular and Cellular Oncology, University of Texas, M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 108, Houston, TX 77030, USA
| | | |
Collapse
|
180
|
Shembade N, Harhaj NS, Parvatiyar K, Copeland NG, Jenkins NA, Matesic LE, Harhaj EW. The E3 ligase Itch negatively regulates inflammatory signaling pathways by controlling the function of the ubiquitin-editing enzyme A20. Nat Immunol 2008; 9:254-62. [PMID: 18246070 DOI: 10.1038/ni1563] [Citation(s) in RCA: 236] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2007] [Accepted: 01/10/2008] [Indexed: 12/18/2022]
Abstract
The ubiquitin-editing enzyme A20 is a critical negative regulator of inflammation and cytokine-mediated activation of the transcription factor NF-kappaB; however, little is known about the mechanisms of A20-mediated inactivation of signaling intermediates such as RIP1. Here we demonstrate that the regulatory molecule TAX1BP1 recruited the E3 ligase Itch to A20 via two 'PPXY' motifs. Itch was essential for the termination of tumor necrosis factor receptor signaling by controlling A20-mediated recruitment and inactivation of RIP1. Furthermore, the Tax oncoprotein of human T cell leukemia virus type I targeted this complex for inactivation by disrupting the interaction among TAX1BP1, A20 and Itch. Thus, our studies show a previously unappreciated complexity of A20 substrate recognition and inactivation whereby TAX1BP1 and Itch function as essential subunits of an A20 ubiquitin-editing complex.
Collapse
Affiliation(s)
- Noula Shembade
- Department of Microbiology and Immunology, Sylvester Comprehensive Cancer Center, The University of Miami, Miller School of Medicine, Miami, Florida 33136, USA
| | | | | | | | | | | | | |
Collapse
|
181
|
Turer EE, Tavares RM, Mortier E, Hitotsumatsu O, Advincula R, Lee B, Shifrin N, Malynn BA, Ma A. Homeostatic MyD88-dependent signals cause lethal inflamMation in the absence of A20. ACTA ACUST UNITED AC 2008; 205:451-64. [PMID: 18268035 PMCID: PMC2271029 DOI: 10.1084/jem.20071108] [Citation(s) in RCA: 232] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Toll-like receptors (TLRs) on host cells are chronically engaged by microbial ligands during homeostatic conditions. These signals do not cause inflammatory immune responses in unperturbed mice, even though they drive innate and adaptive immune responses when combating microbial infections. A20 is a ubiquitin-modifying enzyme that restricts exogenous TLR-induced signals. We show that MyD88-dependent TLR signals drive the spontaneous T cell and myeloid cell activation, cachexia, and premature lethality seen in A20-deficient mice. We have used broad spectrum antibiotics to demonstrate that these constitutive TLR signals are driven by commensal intestinal flora. A20 restricts TLR signals by restricting ubiquitylation of the E3 ligase tumor necrosis factor receptor–associated factor 6. These results reveal both the severe proinflammatory pathophysiology that can arise from homeostatic TLR signals as well as the critical role of A20 in restricting these signals in vivo. In addition, A20 restricts MyD88-independent TLR signals by inhibiting Toll/interleukin 1 receptor domain–containing adaptor inducing interferon (IFN) β–dependent nuclear factor κB signals but not IFN response factor 3 signaling. These findings provide novel insights into how physiological TLR signals are regulated.
Collapse
Affiliation(s)
- Emre E Turer
- Gastrointestinal Division, Department of Medicine, Biomedical Sciences Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Chen RAJ, Ryzhakov G, Cooray S, Randow F, Smith GL. Inhibition of IkappaB kinase by vaccinia virus virulence factor B14. PLoS Pathog 2008; 4:e22. [PMID: 18266467 PMCID: PMC2233672 DOI: 10.1371/journal.ppat.0040022] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2007] [Accepted: 12/20/2007] [Indexed: 01/08/2023] Open
Abstract
The IkappaB kinase (IKK) complex is a key regulator of signal transduction pathways leading to the induction of NF-kappaB-dependent gene expression and production of pro-inflammatory cytokines. It therefore represents a major target for the development of anti-inflammatory therapeutic drugs and may be targeted by pathogens seeking to diminish the host response to infection. Previously, the vaccinia virus (VACV) strain Western Reserve B14 protein was characterised as an intracellular virulence factor that alters the inflammatory response to infection by an unknown mechanism. Here we demonstrate that ectopic expression of B14 inhibited NF-kappaB activation in response to TNFalpha, IL-1beta, poly(I:C), and PMA. In cells infected with VACV lacking gene B14R (vDeltaB14) there was a higher level of phosphorylated IkappaBalpha but a similar level of IkappaBalpha compared to cells infected with control viruses expressing B14, suggesting B14 affects IKK activity. Direct evidence for this was obtained by showing that B14 co-purified and co-precipitated with the endogenous IKK complex from human and mouse cells and inhibited IKK complex enzymatic activity. Notably, the interaction between B14 and the IKK complex required IKKbeta but not IKKalpha, suggesting the interaction occurs via IKKbeta. B14 inhibited NF-kappaB activation induced by overexpression of IKKalpha, IKKbeta, and a constitutively active mutant of IKKalpha, S176/180E, but did not inhibit a comparable mutant of IKKbeta, S177/181E. This suggested that phosphorylation of these serine residues in the activation loop of IKKbeta is targeted by B14, and this was confirmed using Ab specific for phospho-IKKbeta.
Collapse
Affiliation(s)
- Ron A.-J Chen
- Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Grigory Ryzhakov
- Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Samantha Cooray
- Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Felix Randow
- Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Geoffrey L Smith
- Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
183
|
Enesa K, Zakkar M, Chaudhury H, Luong LA, Rawlinson L, Mason JC, Haskard DO, Dean JLE, Evans PC. NF-kappaB suppression by the deubiquitinating enzyme Cezanne: a novel negative feedback loop in pro-inflammatory signaling. J Biol Chem 2008; 283:7036-45. [PMID: 18178551 DOI: 10.1074/jbc.m708690200] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Transcription factors belonging to the NF-kappaB family regulate inflammation by inducing pro-inflammatory molecules (e.g. interleukin (IL)-8) in response to cytokines (e.g. tumor necrosis factor (TNF) alpha, IL-1) or other stimuli. Several negative regulators of NF-kappaB, including the ubiquitin-editing enzyme A20, participate in the resolution of inflammatory responses. We report that Cezanne, a member of the A20 family of the deubiquitinating cysteine proteases, can be induced by TNFalpha in cultured cells. Silencing of endogenous Cezanne using small interfering RNA led to elevated NF-kappaB luciferase reporter gene activity and enhanced expression of IL-8 transcripts in TNFalpha-treated cells. Thus we conclude that endogenous Cezanne can attenuate NF-kappaB activation and the induction of pro-inflammatory transcripts in response to TNF receptor (TNFR) signaling. Overexpression studies revealed that Cezanne suppressed NF-kappaB nuclear translocation and transcriptional activity by targeting the TNFR signaling pathway at the level of the IkappaB kinase complex or upstream from it. These effects were not observed in a form of Cezanne that was mutated at the catalytic cysteine residue (Cys209), indicating that the deubiquitinating activity of Cezanne is essential for NF-kappaB regulation. Finally, we demonstrate that Cezanne can be recruited to activated TNFRs where it suppresses the build-up of polyubiquitinated RIP1 signal adapter proteins. Thus we conclude that Cezanne forms a novel negative feedback loop in pro-inflammatory signaling and that it suppresses NF-kappaB activation by targeting RIP1 signaling intermediaries for deubiquitination.
Collapse
Affiliation(s)
- Karine Enesa
- British Heart Foundation Cardiovascular Sciences Unit, National Heart and Lung Institute, Imperial College London, London W12 ONN, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Hirano T, Murakami M, Fukada T, Nishida K, Yamasaki S, Suzuki T. Roles of zinc and zinc signaling in immunity: zinc as an intracellular signaling molecule. Adv Immunol 2008; 97:149-76. [PMID: 18501770 DOI: 10.1016/s0065-2776(08)00003-5] [Citation(s) in RCA: 177] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Zinc (Zn) is an essential nutrient required for cell growth, differentiation, and survival, and its deficiency causes growth retardation, immunodeficiency, and other health problems. Therefore, Zn homeostasis must be tightly controlled in individual cells. Zn is known to be important in the immune system, although its precise roles and mechanisms have not yet been resolved. Zn has been suggested to act as a kind of neurotransmitter. In addition, Zn has been shown to bind and affect the activity of several signaling molecules, such as protein tyrosine phosphatases (PTPs). However, it has not been known whether Zn itself might act as an intracellular signaling molecule, that is, a molecule whose intracellular status is altered in response to an extracellular stimulus, and that is capable of transducing the extracellular stimulus into an intracellular signaling event. Here we propose that Zn acts as a signaling molecule and that there are at least two kinds of Zn signaling: "late Zn signaling," which is dependent on a change in the expression profile of Zn transporters, and "early Zn signaling," which involves a "Zn wave" and is directly induced by an extracellular stimulus. We also review recent progress in uncovering the roles of Zn in the immune system.
Collapse
Affiliation(s)
- Toshio Hirano
- Laboratory of Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
185
|
Prasad AS. Clinical, immunological, anti-inflammatory and antioxidant roles of zinc. Exp Gerontol 2007; 43:370-7. [PMID: 18054190 DOI: 10.1016/j.exger.2007.10.013] [Citation(s) in RCA: 269] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2007] [Revised: 10/18/2007] [Accepted: 10/23/2007] [Indexed: 11/29/2022]
Abstract
The essentiality of zinc for humans was recognized only 40 years ago. Zinc deficiency was suspected to occur in Iranian patients with growth retardation, hypogonadism in males, hepato-splenomegaly, rough and dry skin, geophagia and severe iron deficiency anemia. Later we documented zinc deficiency in similar patients in Egypt. The diet of these patients consisted of mainly cereal proteins which contained high phytate and this led to decreased availability of iron and zinc. These patients had severe immune dysfunctions, inasmuch as they died of intercurrent infections by the time they were 25 years of age. In our studies in experimental human model of zinc deficiency, we documented decreased serum testosterone level, oligospermia, severe immune dysfunctions mainly affecting T helper cells, decreased serum thymulin activity hyperammonemia, neuro-sensory disorders and decreased lean body mass. The basic mechanisms of zinc action on immune cells have been reviewed in this paper. Our studies showed that the activation of many zinc dependent enzymes and transcription factors were affected adversely due to zinc deficiency. The gene expression and production of Th1 cytokines were affected adversely due to zinc deficiency. Zinc is also an antioxidant and has anti-inflammatory actions. We have reported decreased plasma zinc, increased plasma oxidative stress markers and increased generation of inflammatory cytokines in the elderly subjects which were corrected by zinc supplementation. In cell culture studies, we have observed that zinc induces A20 which inhibits NF-kappaB activation resulting in decreased generation of inflammatory cytokines.
Collapse
Affiliation(s)
- Ananda S Prasad
- Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
186
|
Stewart R, Wei W, Challa A, Armitage RJ, Arrand JR, Rowe M, Young LS, Eliopoulos A, Gordon J. CD154 tone sets the signaling pathways and transcriptome generated in model CD40-pluricompetent L3055 Burkitt's lymphoma cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 179:2705-12. [PMID: 17709483 DOI: 10.4049/jimmunol.179.5.2705] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Activated B cells reacting to small amounts of CD40L (CD154) maintain homeostasis by suppressing default apoptosis. Additional outcomes, particularly differentiation, demand higher CD40 occupancy. Here, focusing on survival, we compared changes in the transcriptome of pleiotropically competent, early passage L3055 Burkitt's lymphoma cells confronted with low (picomolar) and high (nanomolar) concentrations of CD154 to gain insight into how a single receptor sets these distinct phenotypes. Of 267 genes altering transcriptional activity in response to strong CD154 tone, only 25 changed coordinately on low receptor occupancy. Seven of the top nine common up-regulated genes were targets of NF-kappaB. Direct measurement and functional inhibition of the NF-kappaB pathway revealed it to be central to a CD40-dependent survival signature. Although the canonical NF-kappaB axis was engaged by both signaling strengths equally, robust alternative pathway activation was a feature selective to a strong CD40 signal. Discriminatory exploitation of the two separate arms of NF-kappaB activation may indicate a principle whereby a cell senses and reacts differentially to shifting ligand availability. Identifying components selectively coupling CD40 to each axis could indicate targets for disruption in B cell pathologies underpinned by ectopic and/or hyper-CD154 activity such as neoplasia and some autoimmunities.
Collapse
Affiliation(s)
- Ross Stewart
- MRC Centre for Immune Regulation, University of Birmingham Medical School, Birmingham, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Hutti JE, Turk BE, Asara JM, Ma A, Cantley LC, Abbott DW. IkappaB kinase beta phosphorylates the K63 deubiquitinase A20 to cause feedback inhibition of the NF-kappaB pathway. Mol Cell Biol 2007; 27:7451-61. [PMID: 17709380 PMCID: PMC2169042 DOI: 10.1128/mcb.01101-07] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Misregulation of NF-kappaB signaling leads to infectious, inflammatory, or autoimmune disorders. IkappaB kinase beta (IKKbeta) is an essential activator of NF-kappaB and is known to phosphorylate the NF-kappaB inhibitor, IkappaBalpha, allowing it to undergo ubiquitin-mediated proteasomal degradation. However, beyond IkappaBalpha, few additional IKKbeta substrates have been identified. Here we utilize a peptide library and bioinformatic approach to predict likely substrates of IKKbeta. This approach predicted Ser381 of the K63 deubiquitinase A20 as a likely site of IKKbeta phosphorylation. While A20 is a known negative regulator of innate immune signaling pathways, the mechanisms regulating the activity of A20 are poorly understood. We show that IKKbeta phosphorylates A20 in vitro and in vivo at serine 381, and we further show that this phosphorylation event increases the ability of A20 to inhibit the NF-kappaB signaling pathway. Phosphorylation of A20 by IKKbeta thus represents part of a novel feedback loop that regulates the duration of NF-kappaB signaling following activation of innate immune signaling pathways.
Collapse
Affiliation(s)
- Jessica E Hutti
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
188
|
Au PYB, Yeh WC. Physiological roles and mechanisms of signaling by TRAF2 and TRAF5. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 597:32-47. [PMID: 17633015 DOI: 10.1007/978-0-387-70630-6_3] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
RAF2 and TRAF5 are closely related members of the TRAF family of proteins. They are important signal transducers for a wide range of TNF receptor superfamily members, including TNFR1, TNFR2, CD40 and other lymphocyte costimulatory receptors, RANK/TRANCE-R, EDAR, LTbetaR, LMP-1 and IRE1. TRAF2 andTRAF5 therefore regulate diverse physiological roles, ranging from T and B cell signaling and inflammatory responses to organogenesis and cell survival. The major pathways mediated by TRAF2 and TRAF5 are the classical and alternative pathways of NF-kappaB activation, and MAPK and JNK activation. TRAF2 is heavily regulated by ubiquitin signals, and many of the signaling functions of TRAF2 are mediated through its RING domain and likely its own role as an E3 ubiquitin ligase.
Collapse
Affiliation(s)
- Ping-Yee Billie Au
- Campbell Family for Breast Cancer Research, University Health Network and Department of Medical Biophysics, University of Toronto, Toranto, Ontario, Canada
| | | |
Collapse
|
189
|
Prasad AS, Beck FWJ, Bao B, Fitzgerald JT, Snell DC, Steinberg JD, Cardozo LJ. Zinc supplementation decreases incidence of infections in the elderly: effect of zinc on generation of cytokines and oxidative stress. Am J Clin Nutr 2007; 85:837-44. [PMID: 17344507 DOI: 10.1093/ajcn/85.3.837] [Citation(s) in RCA: 321] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Zinc deficiency, cell-mediated immune dysfunction, susceptibility to infections, and increased oxidative stress have been observed in elderly subjects (ie, those >55 y old). Zinc is an effective antiinflammatory and antioxidant agent. OBJECTIVES The primary objective was to determine the effect of zinc on the incidence of total infections in healthy elderly subjects. The secondary objective was to determine the effect of zinc on cytokines and oxidative stress markers. DESIGN A randomized, double-blind, placebo-controlled trial of zinc supplementation was conducted in elderly subjects. Fifty healthy subjects of both sexes aged 55-87 y and inclusive of all ethnic groups were recruited for this study from a senior center. The zinc-supplemented group received zinc gluconate (45 mg elemental Zn/d) orally for 12 mo. Incidence of infections during the supplementation period was documented. The generation of inflammatory cytokines, T helper 1 and T helper 2 cytokines, and oxidative stress markers and the plasma concentrations of zinc were measured at baseline and after supplementation. RESULTS Compared with a group of younger adults, at baseline the older subjects had significantly lower plasma zinc, higher ex vivo generation of inflammatory cytokines and interleukin 10, and higher plasma oxidative stress markers and endothelial cell adhesion molecules. The incidence of infections and ex vivo generation of tumor necrosis factor alpha and plasma oxidative stress markers were significantly lower in the zinc-supplemented than in the placebo group. Plasma zinc and phytohemagglutin-induced interleukin 2 mRNA in isolated mononuclear cells were significantly higher in the zinc-supplemented than in the placebo group. CONCLUSIONS After zinc supplementation, the incidence of infections was significantly lower, plasma zinc was significantly higher, and generation of tumor necrosis factor alpha and oxidative stress markers was significantly lower in the zinc-supplemented than in the placebo group.
Collapse
Affiliation(s)
- Ananda S Prasad
- Wayne State University School of Medicine, Detroit, MI, and the University of Michigan, Ann Arbor, MI 48201, USA.
| | | | | | | | | | | | | |
Collapse
|
190
|
Abstract
Zinc (Zn) deficiency, a frequent condition in human population especially in aged persons, induces oxidative stress and subsequently activates/inhibits oxidant-sensitive transcription factors that can affect cell function, proliferation and survival leading to disease. Zn deficiency-triggered oxidative stress could affect cell signalling, including transcription factors containing Zn finger motifs and other oxidant-sensitive transcription factors such as nuclear factor kappa B (NF-kappaB) and activator protein-1 (AP-1). AP-1 can be activated in Zn deficiency that can occur secondary to an increase in cellular H(2)O(2), followed by activation of MAPKs p38 and JNK. Similarly, the cytosolic steps of the NF-kappaB cascade are activated by oxidants in Zn deficiency. However, an impaired nuclear transport of the active transcription factor leads to a low expression of NF-kappaB-dependent genes that could be involved in multiple steps of Zn deficiency associated pathology. We present here evidence that, following experimental depletion of Zn, both NF-kappaB and AP-1 signallings are altered in primary T cells isolated from young and elderly healthy individuals under CD3/CD28 costimulation. A supplementation of Zn restored both NF-kappaB and AP-1 activation in CD3/CD28 costimulated T cells from young, but not from elderly, healthy individuals, indicating that the Zn deficiency is only one component of a more complex mechanism involved in immunosenescence. In this review we summarize our present knowledge on NF-kappaB and AP-1 activation and underline the role of Zn in this process, especially in the context of Zn deficiency observed in aged persons leading to immunosenescence.
Collapse
Affiliation(s)
- Georges Herbein
- Department of Virology, IFR 133, EA 3186, Franche-Comte University, F-25030, Besancon, France.
| | | | | |
Collapse
|
191
|
Chng HW, Camplejohn RS, Stone MG, Hart IR, Nicholson LJ. A new role for the anti-apoptotic gene A20 in angiogenesis. Exp Cell Res 2006; 312:2897-907. [PMID: 16824518 DOI: 10.1016/j.yexcr.2006.05.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2006] [Revised: 05/19/2006] [Accepted: 05/28/2006] [Indexed: 11/26/2022]
Abstract
A20 is a negative regulator of NF-kappaB activation and thus a potential therapeutic tool for the treatment of diseases where apoptosis and/or inflammatory responses are part of the pathogenic process. Thus, A20 has been shown to improve the long-term outcome of organ transplantation, particularly, the transplantation of islets of Langerhans which may aid the cure of type I diabetes. We now report a new role for A20 in regulating neovascularisation. We used RNA interference to inhibit A20 expression in primary human umbilical vein endothelial cells (HUVECs) and investigated the effect on tubule formation in two in vitro angiogenesis assays, Matrigel and a co-culture assay. Tubule area and tubule length were both reduced following inhibition of A20 expression in HUVECs. These inhibitory effects were particularly evident in the co-culture assay, which incorporates the critical steps of the angiogenic process and ultimately results in the formation of an intricate network of anastomosing tubules that resemble the formed capillary bed: a partial down-regulation of A20 protein (50-60%) resulted in a 28% reduction in tubule area (P < 0.05) and a 26% reduction in tubule length (P < 0.05). A20 may offer a new target in the treatment of human conditions, including cancer, which are characterised by neovascularisation.
Collapse
Affiliation(s)
- Hsiao W Chng
- Department of Oncology, King's College London School of Medicine, The Rayne Institute, St Thomas' Hospital, London SE1 7EH, UK
| | | | | | | | | |
Collapse
|
192
|
Riachy R, Vandewalle B, Moerman E, Belaich S, Lukowiak B, Gmyr V, Muharram G, Kerr Conte J, Pattou F. 1,25-Dihydroxyvitamin D3 protects human pancreatic islets against cytokine-induced apoptosis via down-regulation of the Fas receptor. Apoptosis 2006; 11:151-9. [PMID: 16502254 DOI: 10.1007/s10495-006-3558-z] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Beta cell loss occurs at the onset of type 1 diabetes and after islet graft. It results from the dysfunction and destruction of beta cells mainly achieved by apoptosis. One of the mediators believed to be involved in beta cell apoptosis is Fas, a transmembrane cell surface receptor transducing an apoptotic death signal and contributing to the pathogenesis of several autoimmune diseases. Fas expression is particularly induced in beta cells by inflammatory cytokines secreted by islet-infiltrating mononuclear cells and makes cells susceptible to apoptosis by interaction with Fas-ligand expressing cells. We have previously demonstrated that 1,25(OH)2D3, the active metabolite of vitamin D, known to exhibit immunomodulatory properties and prevent the development of type 1 diabetes in NOD mice, is efficient against apoptosis induced by cytokines in human pancreatic islets in vitro. The effects were mainly mediated by the inactivation of NF-kappa-B. In this study we demonstrated that 1,25(OH)2D3 was also able to counteract cytokine-induced Fas expression in human islets both at the mRNA and protein levels. These results were reinforced by our microarray analysis highlighting the beneficial effects of 1,25(OH)2D3 on death signals induced by Fas activation. Our results provides additional evidence that 1,25(OH)2D3 may be an interesting tool to help prevent the onset of type 1 diabetes and improve islet graft survival.
Collapse
Affiliation(s)
- R Riachy
- Faculté de Médecine, Cellular Therapy of Diabetes, Institut National de la Santé et de la Recherche Médicale, ERIT-M 0106, 59045, Lille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Onose A, Hashimoto S, Hayashi S, Maruoka S, Kumasawa F, Mizumura K, Jibiki I, Matsumoto K, Gon Y, Kobayashi T, Takahashi N, Shibata Y, Abiko Y, Shibata T, Shimizu K, Horie T. An inhibitory effect of A20 on NF-kappaB activation in airway epithelium upon influenza virus infection. Eur J Pharmacol 2006; 541:198-204. [PMID: 16765340 DOI: 10.1016/j.ejphar.2006.03.073] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2005] [Revised: 03/15/2006] [Accepted: 03/27/2006] [Indexed: 11/19/2022]
Abstract
Influenza is a major disease in humans. The reemergence of avian influenza A viruses has indicated that hyperinflammatory responses are closely related to the severity of disease. Influenza virus infection induces nuclear transcription factor kappaB (NF-kappaB) activation. NF-kappaB and NF-kappaB-dependent gene products promote lung inflammation and injury. Therefore, it is important to investigate the means to attenuate NF-kappaB activation. A20 is a cytoplasmic zinc finger protein that inhibits NF-kappaB activity, However, little is known about the role of A20 in influenza virus infection. Here, we have examined the role of A20 in influenza virus infection-induced NF-kappaB promoter activation in human bronchial epithelial cells. The results showed that (1) A20 protein and mRNA are inducible and expressed in the lung from mice and human bronchial epithelial cells upon influenza virus infection; (2) NF-kappaB promoter activation was induced in bronchial epithelial cells upon influenza virus infection; and (3) overexpression by transient transfection of A20 attenuated NF-kappaB promoter activation in bronchial epithelial cells. These results indicate that A20 may function as a negative regulator of NF-kappaB-mediated lung inflammation and injury upon influenza virus infection, thereby protecting the host against inflammatory response to influenza virus infection.
Collapse
Affiliation(s)
- Akira Onose
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Xie P, Hostager BS, Munroe ME, Moore CR, Bishop GA. Cooperation between TNF Receptor-Associated Factors 1 and 2 in CD40 Signaling. THE JOURNAL OF IMMUNOLOGY 2006; 176:5388-400. [PMID: 16622006 DOI: 10.4049/jimmunol.176.9.5388] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
TNFR-associated factor 1 (TRAF1) is unique among the TRAF family, lacking most zinc-binding features, and showing marked up-regulation following activation signals. However, the biological roles that TRAF1 plays in immune cell signaling have been elusive, with many reports assigning contradictory roles to TRAF1. The overlapping binding site for TRAFs 1, 2, and 3 on many TNFR superfamily molecules, together with the early lethality of mice deficient in TRAFs 2 and 3, has complicated the quest for a clear understanding of the functions of TRAF1. Using a new method for gene targeting by homologous recombination in somatic cells, we produced and studied signaling by CD40 and its viral oncogenic mimic, latent membrane protein 1 (LMP1) in mouse B cell lines lacking TRAF1, TRAF2, or both TRAFs. Results indicate that TRAFs 1 and 2 cooperate in CD40-mediated activation of the B cell lines, with a dual deficiency leading to a markedly greater loss of function than that of either TRAF alone. In the absence of TRAF1, an increased amount of TRAF2 was recruited to lipid rafts, and subsequently, more robust degradation of TRAF2 and TRAF3 was induced in response to CD40 signaling. In contrast, LMP1 did not require either TRAFs 1 or 2 to induce activation. Taken together, our findings indicate that TRAF1 and TRAF2 cooperate in CD40 but not LMP1 signaling and suggest that cellular levels of TRAF1 may play an important role in modulating the degradation of TRAF2 and TRAF3 in response to signals from the TNFR superfamily.
Collapse
Affiliation(s)
- Ping Xie
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | |
Collapse
|
195
|
Tian B, Nowak DE, Brasier AR. A TNF-induced gene expression program under oscillatory NF-kappaB control. BMC Genomics 2005; 6:137. [PMID: 16191192 PMCID: PMC1262712 DOI: 10.1186/1471-2164-6-137] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2005] [Accepted: 09/28/2005] [Indexed: 11/10/2022] Open
Abstract
Background The cytokine tumor necrosis factor (TNF) initiates tissue inflammation, a process mediated by the NF-κB transcription factor. In response to TNF, latent cytoplasmic NF-κB is activated, enters the nucleus, and induces expression of inflammatory and anti-apoptotic gene expression programs. Recently it has been shown that NF-κB displays two distinct activation modes, monophasic and oscillatory, depending on stimulus duration. Characterization of temporal expression patterns for the NF-κB network and determination of those genes under monophasic- or oscillatory control has not been experimentally addressed. Results To identify the kinetics of NF-κB-dependent gene expression and determine whether these two types of NF-κB translocation modes control distinct gene programs, a detailed kinetic analysis of a validated microarray data set was performed on 74 unique NF-κB-dependent genes in response to TNF. Hierarchical clustering identified distinct expression profiles termed the "Early", "Middle", "Late" response groups, peaking 1, 3, and 6 h after stimulation, respectively. These expression patterns were validated by Quantitative Real Time PCR (Q-RT-PCR) and NF-κB binding was demonstrated by chromatin immunoprecipitation (ChIP) assays. Each response group was mapped to its molecular function; this analysis indicated that the Early group encodes cytokines or negative regulators of the IKK-NF-κB pathway, and the Late group encodes cell surface receptors, adhesion molecules and signal adapters. That similar coordinated sequential cascades of gene expression were also seen in response to stimulation by the cytokine IL-1, and expression patterns observed in MRC-5 fibroblasts indicated that the epithelial NF-κB program is relatively stimulus- and cell type-independent. Bioinformatic analysis of the Early and Late gene promoters indicates that although both groups contain similar patterns of NF-κB-binding sites, only the Early gene promoters contain NF-κB-binding sites located in phylogenetically conserved domains. Stimulation protocols designed to produce either monophasic or oscillatory NF-κB activation modes showed that the oscillatory mode is required only for expression of the Late genes. Conclusion This analysis provides important insights into the TNF-regulated genetic response program in epithelial cells, where NF-κB controls sequential expression patterns of functionally distinct genes that depend on its oscillatory activation mode.
Collapse
Affiliation(s)
- Bing Tian
- Department of Medicine, The University of Texas Medical Branch, 301 University Blvd., Galveston, Texas 77555-1060, USA
| | - David E Nowak
- Department of Medicine, The University of Texas Medical Branch, 301 University Blvd., Galveston, Texas 77555-1060, USA
| | - Allan R Brasier
- Department of Medicine, The University of Texas Medical Branch, 301 University Blvd., Galveston, Texas 77555-1060, USA
- Sealy Center for Molecular Sciences, The University of Texas Medical Branch, 301 University Blvd., Galveston, Texas 77555-1060, USA
| |
Collapse
|
196
|
Anderle P, Rumbo M, Sierro F, Mansourian R, Michetti P, Roberts MA, Kraehenbuhl JP. Novel markers of the human follicle-associated epithelium identified by genomic profiling and microdissection. Gastroenterology 2005; 129:321-7. [PMID: 16012957 DOI: 10.1053/j.gastro.2005.03.044] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Regulation of gene expression in the follicle-associated epithelium (FAE) over Peyer's patches is largely unknown. CCL20, a chemokine that recruits immature dendritic cells, is one of the few FAE-specific markers described so far. Lymphotoxin beta (LTalpha1beta2) expressed on the membrane of immune cells triggers CCL20 expression in enterocytes. In this study, we measured expression profiles of LTalpha1beta2-treated intestinal epithelial cells and selected CCL20 -coregulated genes to identify new FAE markers. METHODS Genomic profiles of T84 and Caco-2 cell lines treated with either LTalpha1beta2, flagellin, or tumor necrosis factor alpha were measured using the Affymetrix GeneChip U133A. Clustering analysis was used to select CCL20 -coregulated genes, and laser dissection microscopy and real-time polymerase chain reaction on human biopsy specimens was used to assess the expression of the selected markers. RESULTS Applying a 2-way analysis of variance, we identified regulated genes upon the different treatments. A subset of genes involved in inflammation and related to the nuclear factor kappaB pathway was coregulated with CCL20 . Among these genes, the antiapoptotic factor TNFAIP3 was highly expressed in the FAE. CCL23 , which was not coregulated in vitro with CCL20 , was also specifically expressed in the FAE. CONCLUSIONS We have identified 2 novel human FAE specifically expressed genes. Most of the CCL20 -coregulated genes did not show FAE-specific expression, suggesting that other signaling pathways are critical to modulate FAE-specific gene expression.
Collapse
Affiliation(s)
- Pascale Anderle
- Swiss Institute for Experimental Cancer Research and Institute of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | | | | | | | | | | | | |
Collapse
|
197
|
Esparza EM, Arch RH. Glucocorticoid-Induced TNF Receptor, a Costimulatory Receptor on Naive and Activated T Cells, Uses TNF Receptor-Associated Factor 2 in a Novel Fashion as an Inhibitor of NF-κB Activation. THE JOURNAL OF IMMUNOLOGY 2005; 174:7875-82. [PMID: 15944293 DOI: 10.4049/jimmunol.174.12.7875] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Glucocorticoid-induced TNFR (GITR) has been implicated as an essential regulator of immune responses to self tissues and pathogens. We have recently shown that GITR-induced cellular events promote survival of naive T cells, but are insufficient to protect against activation-induced cell death. However, the molecular mechanisms of GITR-induced signal transduction that influence physiologic and pathologic immune responses are not well understood. TNFR-associated factors (TRAFs) are pivotal adapter proteins involved in signal transduction pathways of TNFR-related proteins. Yeast two-hybrid assays and studies in HEK293 cells and primary lymphocytes indicated interactions between TRAF2 and GITR mediated by acidic residues in the cytoplasmic domain of the receptor. GITR-induced activation of NF-kappaB is blocked by A20, an NF-kappaB-inducible protein that interacts with TRAFs and functions in a negative feedback mechanism downstream of other TNFRs. Interestingly, in contrast with its effects on signaling triggered by other TNFRs, our functional studies revealed that TRAF2 plays a novel inhibitory role in GITR-triggered NF-kappaB activation.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Carrier Proteins/physiology
- Cell Line
- Conserved Sequence
- Cysteine Endopeptidases
- Cytoplasm/chemistry
- Cytoplasm/metabolism
- DNA-Binding Proteins
- Down-Regulation/genetics
- Down-Regulation/immunology
- Glucocorticoid-Induced TNFR-Related Protein
- Humans
- Hydrogen-Ion Concentration
- Intracellular Signaling Peptides and Proteins
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Mice
- Mice, Transgenic
- Molecular Sequence Data
- NF-kappa B/antagonists & inhibitors
- NF-kappa B/metabolism
- Nuclear Proteins
- Protein Structure, Tertiary/genetics
- Protein Transport/genetics
- Protein Transport/immunology
- Proteins/pharmacology
- Receptors, Nerve Growth Factor/antagonists & inhibitors
- Receptors, Nerve Growth Factor/metabolism
- Receptors, Nerve Growth Factor/physiology
- Receptors, Tumor Necrosis Factor/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor/physiology
- Resting Phase, Cell Cycle/genetics
- Resting Phase, Cell Cycle/immunology
- TNF Receptor-Associated Factor 2/metabolism
- TNF Receptor-Associated Factor 2/physiology
- Transfection
- Tumor Necrosis Factor alpha-Induced Protein 3
- Tumor Necrosis Factors
Collapse
Affiliation(s)
- Edward M Esparza
- Department of Medicine, School of Medicine, Washington University, St. Louis, MO 63110, USA
| | | |
Collapse
|
198
|
Chang MX, Nie P, Sun BJ, Yao WJ. Molecular cloning of TRAF2 binding protein gene and its promoter region from the grass carp Ctenopharyngodon idellus. Vet Immunol Immunopathol 2005; 105:105-13. [PMID: 15797480 DOI: 10.1016/j.vetimm.2004.12.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2004] [Revised: 11/26/2004] [Accepted: 12/21/2004] [Indexed: 11/22/2022]
Abstract
A tumor necrosis factor receptor-associated factor 2 binding protein (T2BP) gene was isolated from the grass carp (Ctenopharyngodon idellus) by utilizing suppression subtractive hybridization (SSH) and rapid amplification of cDNA ends (RACE). The grass carp T2BP (GT2BP) gene contains an open reading frame of 579 nucleotide(s) (nt), encoding 193 amino acids, with 23 nt 5'-untranslated region and a long 3'-untranslated region of 434 nt including poly (A), 1 AUUUA motif and 4 AUUUUA motifs. No signal peptide has been detected in the predicted GT2BP, but a characteristic forkhead associated domain is present. The GT2BP mRNA shares 83% identity with the zebrafish DNA sequence, and they both have no introns in the genomic DNA. The putative transcription factor binding sites of GT2BP include two C/EBP alpha binding sites, and one c-Jun binding, one AP-1 binding, and one nuclear factor kappaB (NF kappaB) binding sites. Southern blot analysis revealed that the GT2BP was a single-copy gene. Individual difference was observed in GT2BP expression in examined organs of healthy grass carp. However, the expression of GT2BP in all examined organs in a fish with the highest copepod infection level and the significantly higher expression level in spleen and liver in infected fish may indicate its up-regulation with the parasite infection.
Collapse
Affiliation(s)
- M X Chang
- State Key Laboratory of Freshwater Ecology and Biotechnology, and Laboratory of Fish Diseases, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province 430072, China
| | | | | | | |
Collapse
|
199
|
Hsu LJ, Schultz L, Mattison J, Lin YS, Chang NS. Cloning and characterization of a small-size peptide Zfra that regulates the cytotoxic function of tumor necrosis factor by interacting with JNK1. Biochem Biophys Res Commun 2005; 327:415-23. [PMID: 15629131 DOI: 10.1016/j.bbrc.2004.12.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2004] [Indexed: 11/19/2022]
Abstract
By cDNA library screening, here we isolated an unusual gene transcript encoding a 31-amino-acid zinc finger-like peptide that regulates apoptosis (named Zfra). Northern blotting and RT/PCR showed the transcript is abundant in spleen but absent in several prostate and breast cancer cells. When stably expressed in L929 fibroblasts, Zfra conferred resistance to the cytotoxic effects of TNF and FasL. In contrast, transiently expressed Zfra could enhance or inhibit the cytotoxicity of overexpressed death domain proteins TRADD, FADD, and RIP of the TNF signaling pathway. By GST pull-down assay and co-immunoprecipitation, TNF and UV light were shown to induce Zfra to rapidly self-associate and bind JNK1. While JNK1 is a downstream effector of the TNF signaling, Zfra regulation of the TNF cytotoxic function is likely due to its interaction, in part, with JNK1.
Collapse
Affiliation(s)
- Li-Jin Hsu
- Guthrie Research Institute, Laboratory of Molecular Immunology, 1 Guthrie Square, Sayre, PA 18840, USA
| | | | | | | | | |
Collapse
|
200
|
|