1751
|
Winsauer G, Resch U, Hofer-Warbinek R, Schichl YM, de Martin R. XIAP regulates bi-phasic NF-kappaB induction involving physical interaction and ubiquitination of MEKK2. Cell Signal 2008; 20:2107-12. [PMID: 18761086 DOI: 10.1016/j.cellsig.2008.08.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Accepted: 08/04/2008] [Indexed: 10/21/2022]
Abstract
The transcription factor NF-kappaB is transiently activated by a wide variety of stress signals, including pro-inflammatory mediators, and regulates the expression of genes with e.g., immune, inflammatory, and anti-apoptotic functions. The strength and kinetics of its induction, as well as its ultimate down-regulation is subject to multiple levels of regulation. One such regulatory protein is X chromosome-linked inhibitor of apoptosis (XIAP) that, besides its anti-apoptotic properties, has been shown to enhance NF-kappaB activity, however, the underlying molecular mechanism has remained elusive. We show here that following TNFalpha stimulation XIAP regulates a second wave of NF-kappaB activation. XIAP interacts with and ubiquitinates MEKK2, a kinase that has previously been associated with bi-phasic NF-kappaB activation. We conclude that, through interaction with MEKK2, XIAP functions in an ubiquitin ligase dependent manner to evoke a second wave of NF-kappaB activation, resulting in the modulation of NF-kappaB target gene expression.
Collapse
Affiliation(s)
- Gabriele Winsauer
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Austria
| | | | | | | | | |
Collapse
|
1752
|
Xiong Z, Yan Y, Song J, Fang P, Yin Y, Yang Y, Cowan A, Wang H, Yang XF. Expression of TCTP antisense in CD25(high) regulatory T cells aggravates cuff-injured vascular inflammation. Atherosclerosis 2008; 203:401-8. [PMID: 18789801 DOI: 10.1016/j.atherosclerosis.2008.07.041] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Revised: 07/30/2008] [Accepted: 07/30/2008] [Indexed: 12/22/2022]
Abstract
This study examines our hypothesis that translationally controlled tumor protein (TCTP) expression in CD4+ CD25(high) regulatory T cells (Tregs) is critical for the interleukin-2 (IL-2) withdrawal-triggered apoptosis pathway in Tregs, and modulation of Treg apoptosis pathway affects development of vascular inflammation. To test this hypothesis, we established a Tregs-specific TCTP antisense transgenic mouse model. Lower TCTP expression in Tregs than in CD4+ CD25- T cells is associated with the higher susceptibility of Tregs to apoptosis induced by IL-2 withdrawal. Overexpression of TCTP antisense in Tregs leads to decreased positive selection of CD25(high) thymic Tregs and reduced survival of peripheral Tregs, which is correlated to our previous report that TCTP antisense knocks-down TCTP protein expression and promotes apoptosis. In addition, TCTP antisense transgene confers higher susceptibility of Tregs to apoptosis induced by IL-2 withdrawal than wild-type Tregs, which can be suppressed by exogenous supply of IL-2, suggesting that IL-2 promotes Treg survival at least partially due to promoting TCTP expression. Finally, decreased expression of TCTP in Tregs aggravates experimental vascular inflammation, presumably due to increased Treg apoptosis and failure of decreased Tregs in suppressing inflammatory cells and immune cells. These results suggest that the modulation of Tregs apoptosis/survival may be used as a new therapeutic approach for inflammatory cardiovascular diseases.
Collapse
Affiliation(s)
- Zeyu Xiong
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
1753
|
Hamacher R, Schmid RM, Saur D, Schneider G. Apoptotic pathways in pancreatic ductal adenocarcinoma. Mol Cancer 2008; 7:64. [PMID: 18652674 PMCID: PMC2515336 DOI: 10.1186/1476-4598-7-64] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Accepted: 07/24/2008] [Indexed: 02/08/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most common causes of cancer related death. Despite the advances in understanding of the molecular pathogenesis, pancreatic cancer remains a major unsolved health problem. Overall, the 5-year survival rate is less than 5% demonstrating the insufficiency of current therapies. Most cytotoxic therapies induce apoptosis and PDAC cells have evolved a plethora of molecular mechanisms to assure survival. We will present anti-apoptotic strategies working at the level of the death receptors, the mitochondria or involving the caspase inhibitors of the IAP family. Furthermore, the survival function of the phosphotidylinositol-3' kinase (PI3K)/AKT- and NF-kappaB-pathways are illustrated. A detailed molecular knowledge of the anti-apoptotic mechanisms of PDAC cells will help to improve therapies for this dismal disease and therapeutic strategies targeting the programmed cell death machinery are in early preclinical and clinical development.
Collapse
Affiliation(s)
- Rainer Hamacher
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Roland M Schmid
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Dieter Saur
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Günter Schneider
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, München, Germany
| |
Collapse
|
1754
|
Nallagatla SR, Toroney R, Bevilacqua PC. A brilliant disguise for self RNA: 5'-end and internal modifications of primary transcripts suppress elements of innate immunity. RNA Biol 2008; 5:140-4. [PMID: 18769134 DOI: 10.4161/rna.5.3.6839] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Interferon inducible protein kinase PKR is a component of innate immunity and mediates antiviral actions by recognizing pathogen associated molecular patterns (PAMPs). A well-known activator of PKR is long dsRNA, which can be produced during viral replication. Our recent results indicate that PKR can also be activated by short stem-loop RNA in a 5'-triphosphate-dependent fashion. A 5'-triphosphate is present primarily in foreign RNAs such as viral and bacterial transcripts, while a non-activating 5'-cap or 5'-monophosphate is present in most cellular RNAs. Additional studies indicate that internal RNA modifications and non-Watson-Crick motifs also repress PKR activation, and do so in an RNA structure-specific fashion. Interestingly, self-RNAs have more nucleoside modifications than non-self RNAs. Internal and 5'-end RNA modifications have repressive effects on other innate immune sensors as well, including TLR3, TLR7, TLR8, and RIG-I, suggesting that nucleoside modifications suppress innate immunity on a wide scale.
Collapse
Affiliation(s)
- Subba Rao Nallagatla
- Department of Chemistry, Pennsylvania State University, University Park, Pennsylvania, USA.
| | | | | |
Collapse
|
1755
|
Ni CY, Wu ZH, Florence WC, Parekh VV, Arrate MP, Pierce S, Schweitzer B, Van Kaer L, Joyce S, Miyamoto S, Ballard DW, Oltz EM. Cutting edge: K63-linked polyubiquitination of NEMO modulates TLR signaling and inflammation in vivo. THE JOURNAL OF IMMUNOLOGY 2008; 180:7107-11. [PMID: 18490708 DOI: 10.4049/jimmunol.180.11.7107] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Transcription factor NF-kappaB controls the expression of multiple genes involved in immunity and inflammation. The initial activation and duration of NF-kappaB signaling is regulated by posttranslational modifications to IkappaB kinase, which earmarks inhibitors of NF-kappaB for degradation. Prior studies suggest that K63-linked ubiquitination of NEMO (NF-kappaB essential modulator), an IkappaB kinase regulatory subunit, is critical for NF-kappaB and MAPK signaling following engagement of Ag receptors. We now demonstrate that NF-kappaB and MAPK pathways are largely unaffected in primary cells from mice harboring a ubiquitination-defective form of NEMO, NEMO-KR. TLR- but not Ag receptor-induced cellular responses are impaired in NEMO-KR mice, which are more resistant to LPS-induced endotoxic shock than wild-type animals. Thus, one function of NEMO ubiquitination is to fine tune innate immune responses under TLR control.
Collapse
Affiliation(s)
- Chang-Yuan Ni
- Department of Microbiology and Immunology, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1756
|
Kriete A, Mayo KL, Yalamanchili N, Beggs W, Bender P, Kari C, Rodeck U. Cell autonomous expression of inflammatory genes in biologically aged fibroblasts associated with elevated NF-kappaB activity. IMMUNITY & AGEING 2008; 5:5. [PMID: 18631391 PMCID: PMC2483950 DOI: 10.1186/1742-4933-5-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Accepted: 07/16/2008] [Indexed: 02/06/2023]
Abstract
BACKGROUND Chronic inflammation is a well-known corollary of the aging process and is believed to significantly contribute to morbidity and mortality of many age-associated chronic diseases. However, the mechanisms that cause age-associated inflammatory changes are not well understood. Particularly, the contribution of cell stress responses to age-associated inflammation in 'non-inflammatory' cells remains poorly defined. The present cross-sectional study focused on differences in molecular signatures indicative of inflammatory states associated with biological aging of human fibroblasts from donors aged 22 to 92 years. RESULTS Gene expression profiling revealed elevated steady-state transcript levels consistent with a chronic inflammatory state in fibroblast cell-strains obtained from older donors. We also observed enhanced NF-kappaB DNA binding activity in a subset of strains, and the NF-kappaB profile correlated with mRNA expression levels characteristic of inflammatory processes, which include transcripts coding for cytokines, chemokines, components of the complement cascade and MHC molecules. This intrinsic low-grade inflammatory state, as it relates to aging, occurs in cultured cells irrespective of the presence of other cell types or the in vivo context. CONCLUSION Our results are consistent with the view that constitutive activation of inflammatory pathways is a phenomenon prevalent in aged fibroblasts. It is possibly part of a cellular survival process in response to compromised mitochondrial function. Importantly, the inflammatory gene expression signature described here is cell autonomous, i.e. occurs in the absence of prototypical immune or pro-inflammatory cells, growth factors, or other inflammatory mediators.
Collapse
Affiliation(s)
- Andres Kriete
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Bossone Research Center, 3141 Chestnut Street, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | |
Collapse
|
1757
|
Vaira S, Alhawagri M, Anwisye I, Kitaura H, Faccio R, Novack DV. RelA/p65 promotes osteoclast differentiation by blocking a RANKL-induced apoptotic JNK pathway in mice. J Clin Invest 2008; 118:2088-97. [PMID: 18464930 DOI: 10.1172/jci33392] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2007] [Accepted: 03/26/2008] [Indexed: 12/26/2022] Open
Abstract
Osteoclasts (OCs) function to reabsorb bone and are responsible for the bone loss associated with inflammatory arthritis and osteoporosis. OC numbers are elevated in most disorders of accelerated bone destruction, reflecting altered rates of precursor differentiation and apoptosis. Both of these processes are regulated by the JNK family of MAP kinases. In this study, we have demonstrated that the NF-kappaB subunit RelA/p65 inhibits JNK-mediated apoptosis during a critical period of commitment to the OC phenotype in response to the cytokine RANKL. This RelA/p65-mediated arrest of cell death led to enhanced OC differentiation. Hence, Rela-/- OC precursors displayed prolonged JNK activation in response to RANKL, and this was accompanied by an increase in cell death that prevented efficient differentiation. Although complete blockade of JNK activity inhibits osteoclastogenesis, both short-term blockade in RelA-deficient cultures and suppression of the downstream mediator, Bid rescued apoptosis and differentiation. These antiapoptotic effects were RelA specific, as overexpression of RelA, but not RelB, blocked apoptosis and rescued differentiation in Rela-/- precursors. Thus, RelA blocks a RANKL-induced, apoptotic JNK-Bid pathway, thereby promoting OC differentiation. Consistent with this, mice lacking RelA/p65 in the hematopoietic compartment were shown to have a deficient osteoclastogenic response to RANKL and were protected from arthritis-induced osteolysis.
Collapse
Affiliation(s)
- Sergio Vaira
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | | | | |
Collapse
|
1758
|
IKK1 and IKK2 cooperate to maintain bile duct integrity in the liver. Proc Natl Acad Sci U S A 2008; 105:9733-8. [PMID: 18606991 DOI: 10.1073/pnas.0800198105] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Inflammatory destruction of intrahepatic bile ducts is a common cause of vanishing bile duct syndrome and cholestasis, often progressing to biliary cirrhosis and liver failure. However, the molecular mechanisms underlying the pathogenesis of inflammatory biliary disease are poorly understood. Here, we show that the two IkappaB kinases, IKK1/IKKalpha and IKK2/IKKbeta, display distinct collaborative and specific functions that are essential to protect the liver from cytokine toxicity and bile duct disease. Combined conditional ablation of IKK1 and IKK2, but not of each kinase alone, sensitized the liver to in vivo LPS challenge, uncovering a redundant function of the two IkappaB kinases in mediating canonical NF-kappaB signaling in hepatocytes and protecting the liver from TNF-induced failure. Unexpectedly, mice with combined ablation of IKK1 and IKK2 or IKK1 and NEMO spontaneously developed severe jaundice and fatal cholangitis characterized by inflammatory destruction of small portal bile ducts. This bile duct disease was caused by the combined impairment of canonical NF-kappaB signaling together with inhibition of IKK1-specific functions affecting the bile-blood barrier. These results reveal a novel function of the two IkappaB kinases in cooperatively regulating liver immune homeostasis and bile duct integrity and suggest that IKK signaling may be implicated in human biliary diseases.
Collapse
|
1759
|
Lousse JC, Van Langendonckt A, González-Ramos R, Defrère S, Renkin E, Donnez J. Increased activation of nuclear factor-kappa B (NF-κB) in isolated peritoneal macrophages of patients with endometriosis. Fertil Steril 2008; 90:217-20. [PMID: 17889859 DOI: 10.1016/j.fertnstert.2007.06.015] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Revised: 06/04/2007] [Accepted: 06/04/2007] [Indexed: 11/25/2022]
Abstract
Endometriosis is characterized by pelvic inflammation that shows an increased number of activated peritoneal macrophages and their secreted products such as cytokines, growth factors, and angiogenic factors. Our results show that activation of nuclear factor-kappa B (NF-kappaB), a pro-inflammatory transcription factor, is statistically significantly increased in peritoneal macrophages from patients with endometriosis when compared with controls.
Collapse
|
1760
|
Kleino A, Myllymäki H, Kallio J, Vanha-aho LM, Oksanen K, Ulvila J, Hultmark D, Valanne S, Rämet M. Pirk is a negative regulator of the Drosophila Imd pathway. THE JOURNAL OF IMMUNOLOGY 2008; 180:5413-22. [PMID: 18390723 DOI: 10.4049/jimmunol.180.8.5413] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
NF-kappaB transcription factors are involved in evolutionarily conserved signaling pathways controlling multiple cellular processes including apoptosis and immune and inflammatory responses. Immune response of the fruit fly Drosophila melanogaster to Gram-negative bacteria is primarily mediated via the Imd (immune deficiency) pathway, which closely resembles the mammalian TNFR signaling pathway. Instead of cytokines, the main outcome of Imd signaling is the production of antimicrobial peptides. The pathway activity is delicately regulated. Although many of the Imd pathway components are known, the mechanisms of negative regulation are more elusive. In this study we report that a previously uncharacterized gene, pirk, is highly induced upon Gram-negative bacterial infection in Drosophila in vitro and in vivo. pirk encodes a cytoplasmic protein that coimmunoprecipitates with Imd and the cytoplasmic tail of peptidoglycan recognition protein LC (PGRP-LC). RNA interference-mediated down-regulation of Pirk caused Imd pathway hyperactivation upon infection with Gram-negative bacteria, while overexpression of pirk reduced the Imd pathway response both in vitro and in vivo. Furthermore, pirk-overexpressing flies were more susceptible to Gram-negative bacterial infection than wild-type flies. We conclude that Pirk is a negative regulator of the Imd pathway.
Collapse
Affiliation(s)
- Anni Kleino
- Institute of Medical Technology, University of Tampere, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
1761
|
de Seigneux S, Leroy V, Ghzili H, Rousselot M, Nielsen S, Rossier BC, Martin PY, Féraille E. NF-kappaB inhibits sodium transport via down-regulation of SGK1 in renal collecting duct principal cells. J Biol Chem 2008; 283:25671-25681. [PMID: 18586672 DOI: 10.1074/jbc.m803812200] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tubulointerstitial inflammation is a common feature of renal diseases. We have investigated the relationship between inflammation and Na(+) transport in the collecting duct (CD) using the mCCD(cl1) and mpkCDD(cl4) principal cell models. Lipopolysaccharide (LPS) decreased basal and aldosterone-stimulated amiloride-sensitive transepithelial current in a time-dependent manner. This effect was associated with a decrease in serum and glucocorticoid-regulated kinase 1 (SGK1) mRNA and protein levels followed by a decrease in epithelial sodium channel (ENaC) alpha-subunit mRNA levels. The LPS-induced decrease in SGK1 expression was confirmed in isolated rat CD. This decreased expression of either SGK1 or the ENaC alpha-subunit was not due to enhanced degradation of mRNA. In contrast, LPS inhibited transcriptional activity of the SGK1 promoter measured by luciferase-reporter gene assay. The effect of LPS was not mediated by inhibition of mineralocorticoid or glucocorticoid receptor, because expression of both receptors was unchanged and blockade of either receptor by spironolactone or RU486, respectively, did not prevent the down-regulation of SGK1. The effect of LPS was mediated by the canonical NF-kappaB pathway, as overexpression of a constitutively active mutant, IKKbeta (inhibitor of nuclear factor kappaB kinase-beta) decreased SGK1 mRNA levels, and knockdown of p65 NF-kappaB subunit by small interfering RNA increased SGK1 mRNA levels. Chromatin immunoprecipitation showed that LPS increased p65 binding to two NF-kappaB sites along the SGK1 promoter. In conclusion, we show that activation of the NF-kappaB pathway down-regulates SGK1 expression, which might lead to decreased ENaC alpha-subunit expression, ultimately resulting in decreased Na(+) transport.
Collapse
Affiliation(s)
- Sophie de Seigneux
- Service de Néphrologie, Fondation pour Recherches Médicales, 1211 Genève 4, Switzerland
| | - Valérie Leroy
- Service de Néphrologie, Fondation pour Recherches Médicales, 1211 Genève 4, Switzerland
| | - Hafida Ghzili
- Service de Néphrologie, Fondation pour Recherches Médicales, 1211 Genève 4, Switzerland
| | - Martine Rousselot
- Service de Néphrologie, Fondation pour Recherches Médicales, 1211 Genève 4, Switzerland
| | - Søren Nielsen
- The Water and Salt Research Center, Institute of Anatomy, University of Aarhus, 8000 Aarhus, Denmark
| | - Bernard C Rossier
- Department of Pharmacology and Toxicology, University of Lausanne, 1005 Lausanne, Switzerland
| | - Pierre-Yves Martin
- Service de Néphrologie, Fondation pour Recherches Médicales, 1211 Genève 4, Switzerland
| | - Eric Féraille
- Service de Néphrologie, Fondation pour Recherches Médicales, 1211 Genève 4, Switzerland.
| |
Collapse
|
1762
|
Wittwer T, Schmitz ML. NIK and Cot cooperate to trigger NF-kappaB p65 phosphorylation. Biochem Biophys Res Commun 2008; 371:294-7. [PMID: 18439422 DOI: 10.1016/j.bbrc.2008.04.069] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2008] [Accepted: 04/15/2008] [Indexed: 10/22/2022]
Abstract
The serine/threonine kinase Cot triggers NF-kappaB-dependent transactivation and activation of various MAPKinases. Here we identify Cot as a novel p65 interacting protein kinase. Cot expression induces p65 phosphorylation at serines 536 and 468 in dependence from its kinase function. Accordingly, shRNA-mediated knockdown of Cot expression interferes with TNF-induced NF-kappaB-dependent gene expression. Also the C-terminally truncated, oncogenic form of Cot is able to trigger p65 phosphorylation. In vitro kinase assays and dominant negative mutants revealed that NIK functions downstream of Cot to mediate p65 phosphorylation.
Collapse
Affiliation(s)
- Tobias Wittwer
- Institute of Biochemistry, Medical Faculty, Friedrichstrasse 24, Justus-Liebig-University, D-35392 Giessen, Germany
| | | |
Collapse
|
1763
|
Induction of a pro-apoptotic ATM-NF-kappaB pathway and its repression by ATR in response to replication stress. EMBO J 2008; 27:1963-73. [PMID: 18583959 DOI: 10.1038/emboj.2008.127] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Accepted: 06/02/2008] [Indexed: 11/08/2022] Open
Abstract
The transcription factor NF-kappaB has critical functions in biologic responses to genotoxic stimuli. Activation of NF-kappaB in response to DNA double strand break (DSB) inducers can be mediated by ATM (ataxia telangiectasia mutated)-dependent phosphorylation of NEMO (NF-kappaB essential modulator). Here, we show that the replication stress inducers hydroxyurea (HU) and aphidicolin also activate this ATM-dependent signalling pathway. We further show that ATR (ATM- and Rad3-related) interacts with NEMO but surprisingly does not cause NEMO phosphorylation. Consequently, ATR represses NF-kappaB activation induced by replication stress. Reduction or increase of ATR expression by RNA interference or overexpression increased or reduced ATM-NEMO association and NF-kappaB activation induced by HU. Apoptosis gene expression and chromatin immunoprecipitation analyses indicated that HU and the DSB inducer etoposide caused complex patterns of NF-kappaB-dependent pro- and antiapoptotic gene expression with the overall outcome for the former being pro-apoptotic, whereas the latter antiapoptotic. Thus, replication stress and DSB inducers activate NF-kappaB through a conserved pathway with opposite biologic outcomes, and ATR antagonizes ATM function at least in part by competing for NEMO association.
Collapse
|
1764
|
Cvek B, Dvorak Z. The value of proteasome inhibition in cancer. Can the old drug, disulfiram, have a bright new future as a novel proteasome inhibitor? Drug Discov Today 2008; 13:716-22. [PMID: 18579431 DOI: 10.1016/j.drudis.2008.05.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 05/12/2008] [Accepted: 05/13/2008] [Indexed: 12/22/2022]
Abstract
The major approach to the development of anticancer drugs involves searching for new compounds, efficient against malignancies, which are not, as yet, used clinically. This strategy is time-consuming and expensive. Recent studies have disclosed a surprising, but mechanistically consistent, anticancer activity of disulfiram (antabuse), a drug used for about 50 years in the treatment of alcoholism. Disulfiram has been successfully used to suppress hepatic metastases originating from ocular melanoma. The pharmacokinetics of disulfiram and its pharmacological profile in cancer cell lines and in cancer cells obtained from patients is well known. Disulfiram is a readily available and inexpensive substance whose adverse effects are negligible, compared to classical cancerostatics. In addition, the inhibitory potency of disulfiram against the proteasome conforms to current anticancer strategies and represents a new, promising approach to proteasome inhibition.
Collapse
Affiliation(s)
- Boris Cvek
- Department of Medical Chemistry and Biochemistry at Palacky University, Hnevotinska 3, Olomouc 77515, Czech Republic.
| | | |
Collapse
|
1765
|
The death domain-containing kinase RIP1 regulates p27(Kip1) levels through the PI3K-Akt-forkhead pathway. EMBO Rep 2008; 9:766-73. [PMID: 18566599 DOI: 10.1038/embor.2008.102] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 05/07/2008] [Accepted: 05/07/2008] [Indexed: 11/08/2022] Open
Abstract
Elucidating the cross-talk between inflammatory and cell proliferation pathways might provide important insights into the pathogenesis of inflammation-induced cancer. Here, we show that the receptor-interacting protein 1 (RIP1)-an essential mediator of inflammation-induced nuclear factor-kappaB (NF-kappaB) activation-regulates p27(Kip1) levels and cell-cycle progression. RIP1 regulates p27(Kip1) levels by an NF-kappaB-independent signal that involves activation of the phosphatidylinositol 3-kinase (PI3K)-Akt-forkhead pathway. Mouse embryonic fibroblasts (MEFs) from RIP1-knockout mice express high levels of p27(Kip1). Reconstitution of MEFs with RIP1 downregulates p27(Kip1) levels in a PI3K-dependent manner. RIP1 regulates p27(Kip1) at the messenger RNA level by regulating the p27(Kip1) promoter through the forkhead transcription factors. RIP1 expression blocks accumulation of cells in G(1) in response to serum starvation and favours cell-cycle progression. Finally, we show that overexpression of p27(Kip1) blocks the effects of RIP1 on the cell cycle. Thus, our study provides a new insight into how components of inflammatory and immune signalling pathways regulate cell-cycle progression.
Collapse
|
1766
|
Otero JE, Dai S, Foglia D, Alhawagri M, Vacher J, Pasparakis M, Abu-Amer Y. Defective osteoclastogenesis by IKKbeta-null precursors is a result of receptor activator of NF-kappaB ligand (RANKL)-induced JNK-dependent apoptosis and impaired differentiation. J Biol Chem 2008; 283:24546-53. [PMID: 18567579 DOI: 10.1074/jbc.m800434200] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
It has been reported previously that inhibitory kappaB kinase (IKK) supports osteoclastogenesis through NF-kappaB-mediated prevention of apoptosis. This finding suggests that the ligand for receptor activator of NF-kappaB (RANKL), the master osteoclastogenic cytokine, induces apoptosis of osteoclast precursors (OCPs) in the absence of IKKbeta/NF-kappaB competency. To validate this hypothesis, we sought to determine the pro-apoptotic signaling factors induced by RANKL in IKKbeta-null osteoclast OCPs and to rescue osteoclast differentiation in the absence of IKKbeta through their inhibition. To accomplish this, we generated mice that lack IKKbeta in multiple hematopoietic lineages, including OCPs. We found that these mice possess both in vitro and in vivo defects in osteoclast generation, in concurrence with previous reports, and that this defect is a result of susceptibility to RANKL-mediated apoptosis as a result of gain-of-function of JNK activation. We demonstrate that differentiation of OCPs depends on IKKbeta because reduced IKKbeta mRNA expression correlates with impaired induction of osteoclast differentiation markers in response to RANKL stimulation. We further show that fine-tuned inhibition of JNK activation in these cells inhibits RANKL-induced apoptosis and restores the ability of IKKbeta-null OCPs to become mature osteoclasts. Our data highlight the pro-osteoclastogenic and anti-apoptotic roles of IKKbeta in OCPs and identify a pro-apoptotic mechanism activated within the RANK signalosome.
Collapse
Affiliation(s)
- Jesse E Otero
- Department of Orthedic Surgery, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | |
Collapse
|
1767
|
Schön M, Wienrich BG, Kneitz S, Sennefelder H, Amschler K, Vöhringer V, Weber O, Stiewe T, Ziegelbauer K, Schön MP. KINK-1, a Novel Small-Molecule Inhibitor of IKKβ, and the Susceptibility of Melanoma Cells to Antitumoral Treatment. ACTA ACUST UNITED AC 2008; 100:862-75. [DOI: 10.1093/jnci/djn174] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
1768
|
Cousins DJ, McDonald J, Lee TH. Therapeutic approaches for control of transcription factors in allergic disease. J Allergy Clin Immunol 2008; 121:803-9; quiz 810-1. [PMID: 18395546 DOI: 10.1016/j.jaci.2008.02.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 02/08/2008] [Accepted: 02/12/2008] [Indexed: 11/29/2022]
Abstract
The inflammatory response observed in allergic disease involves multiple cell types but is orchestrated in part by the T(H)2 cytokines IL-4, IL-5, and IL-13. In recent years, the transcription factors that control the expression and function of these cytokines have been elucidated, including signal transducer and activator of transcription 6, GATA3, nuclear factor of activated T cells, and nuclear factor kappaB. These molecules are attractive targets for therapeutic intervention because they regulate the expression of numerous effector molecules and functions simultaneously. For instance, the immunosuppressive agents glucocorticoids and cyclosporin A both function by repressing the activity of transcription factors through a variety of mechanisms. In this review we examine the role of each transcription factor in allergic disease and discuss approaches that have been taken to therapeutically interfere with transcription factor function in allergic disease.
Collapse
Affiliation(s)
- David J Cousins
- MRC-Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, United Kingdom.
| | | | | |
Collapse
|
1769
|
Niederberger E, Geisslinger G. The IKK-NF-kappaB pathway: a source for novel molecular drug targets in pain therapy? FASEB J 2008; 22:3432-42. [PMID: 18559989 DOI: 10.1096/fj.08-109355] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Several studies indicate that the nuclear factor-kappa B (NF-kappaB) -activation cascade plays a crucial role not only in immune responses, inflammation, and apoptosis but also in the development and processing of pathological pain. Accordingly, a pharmacological intervention into this pathway may have antinociceptive effects and could provide novel treatment strategies for pain and inflammation. In this review we summarize the role of NF-kappaB in the nervous system, its impact on nociception, and several approaches that investigated the effects of various modulators of the classical I-kappaB-kinase-NF-kappaB signal transduction pathway in inflammatory nociception and neuropathic pain. The results indicate that NF-kappaB has an impact on nociceptive transmission and processing and that a number of substances that inhibit the NF-kappaB-activating cascade are capable of reducing the nociceptive response in different animal models. Therefore, a modulation of specific participants in the NF-kappaB signal transduction might exert a useful approach for the development of new painkillers.
Collapse
Affiliation(s)
- Ellen Niederberger
- Pharmazentrum Frankfurt/ZAFES, Institut für Klinische Pharmakologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany.
| | | |
Collapse
|
1770
|
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: 129] [Impact Index Per Article: 7.6] [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 IκB kinase (IKK) complex is a key regulator of signal transduction pathways leading to the induction of NF-κB-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-κB activation in response to TNFα, IL-1β, poly(I:C), and PMA. In cells infected with VACV lacking gene B14R (vΔB14) there was a higher level of phosphorylated IκBα but a similar level of IκBα 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 IKKβ but not IKKα, suggesting the interaction occurs via IKKβ. B14 inhibited NF-κB activation induced by overexpression of IKKα, IKKβ, and a constitutively active mutant of IKKα, S176/180E, but did not inhibit a comparable mutant of IKKβ, S177/181E. This suggested that phosphorylation of these serine residues in the activation loop of IKKβ is targeted by B14, and this was confirmed using Ab specific for phospho-IKKβ. Vaccinia virus (VACV) is the live vaccine used to eradicate smallpox and is also the most intensively studied poxvirus. Like many poxviruses, VACV produces a wide variety of proteins that inhibit parts of the host response to infection. Consequently, the virus can escape destruction by the immune system and be passed on to additional hosts. Here we report a new VACV immune evasion mechanism mediated by protein B14, a protein that contributes to virus virulence. B14 functions by interacting with a cellular protein called IKKβ, which is critical for mounting an innate immune response to infection, and also plays important roles in cancer and cell death. B14 prevents IKKβ being activated and consequently the cellular signaling pathway leading to activation of nuclear factor kappa B (NF-κB) is not induced. Without activation of NF-κB the host cell cannot produce other molecules that amplify the innate immune response to infection. This mechanism of action of B14 fits nicely with the observed increase in the host response to infection by a VACV strain lacking the B14R gene. Lastly, an increased understanding of how B14 inhibits IKKβ function may lead to development of novel drugs against this important cellular enzyme.
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
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
1771
|
Dodeller F, Gottar M, Huesken D, Iourgenko V, Cenni B. The lysosomal transmembrane protein 9B regulates the activity of inflammatory signaling pathways. J Biol Chem 2008; 283:21487-94. [PMID: 18541524 DOI: 10.1074/jbc.m801908200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The intracellular signaling pathway by which tumor necrosis factor (TNF) induces its pleiotropic actions is well characterized and includes unique components as well as modules shared with other signaling pathways. In addition to the currently known key effectors, further molecules may however modulate the biological response to TNF. In our attempt to characterize novel regulators of the TNF signaling cascade, we have identified transmembrane protein 9B (TMEM9B, c11orf15) as an important component of TNF signaling and a module shared with the interleukin 1beta (IL-1beta) and Toll-like receptor (TLR) pathways. TMEM9B is a glycosylated protein localized in membranes of the lysosome and partially in early endosomes. The expression of TMEM9B is required for the production of proinflammatory cytokines induced by TNF, IL-1beta, and TLR ligands but not for apoptotic cell death triggered by TNF or Fas ligand. TMEM9B is essential in TNF activation of both the NF-kappaB and MAPK pathways. It acts downstream of RIP1 and upstream of the MAPK and IkappaB kinases at the level of the TAK1 complex. These findings indicate that TMEM9B is a key component of inflammatory signaling pathways and suggest that endosomal or lysosomal compartments regulate these pathways.
Collapse
|
1772
|
|
1773
|
MacKenzie EL, Iwasaki K, Tsuji Y. Intracellular iron transport and storage: from molecular mechanisms to health implications. Antioxid Redox Signal 2008; 10:997-1030. [PMID: 18327971 PMCID: PMC2932529 DOI: 10.1089/ars.2007.1893] [Citation(s) in RCA: 388] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Revised: 12/03/2007] [Accepted: 12/04/2007] [Indexed: 12/21/2022]
Abstract
Maintenance of proper "labile iron" levels is a critical component in preserving homeostasis. Iron is a vital element that is a constituent of a number of important macromolecules, including those involved in energy production, respiration, DNA synthesis, and metabolism; however, excess "labile iron" is potentially detrimental to the cell or organism or both because of its propensity to participate in oxidation-reduction reactions that generate harmful free radicals. Because of this dual nature, elaborate systems tightly control the concentration of available iron. Perturbation of normal physiologic iron concentrations may be both a cause and a consequence of cellular damage and disease states. This review highlights the molecular mechanisms responsible for regulation of iron absorption, transport, and storage through the roles of key regulatory proteins, including ferroportin, hepcidin, ferritin, and frataxin. In addition, we present an overview of the relation between iron regulation and oxidative stress and we discuss the role of functional iron overload in the pathogenesis of hemochromatosis, neurodegeneration, and inflammation.
Collapse
Affiliation(s)
- Elizabeth L MacKenzie
- Department of Environmental and Molecular Toxicology, North Carolina State University, Raleigh, North Carolina 27695, USA
| | | | | |
Collapse
|
1774
|
The TWEAK-Fn14 cytokine-receptor axis: discovery, biology and therapeutic targeting. Nat Rev Drug Discov 2008; 7:411-25. [PMID: 18404150 DOI: 10.1038/nrd2488] [Citation(s) in RCA: 453] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
TWEAK is a multifunctional cytokine that controls many cellular activities including proliferation, migration, differentiation, apoptosis, angiogenesis and inflammation. TWEAK acts by binding to Fn14, a highly inducible cell-surface receptor that is linked to several intracellular signalling pathways, including the nuclear factor-kappaB (NF-kappaB) pathway. The TWEAK-Fn14 axis normally regulates various physiological processes, in particular it seems to play an important, beneficial role in tissue repair following acute injury. Furthermore, recent studies have indicated that TWEAK-Fn14 axis signalling may contribute to cancer, chronic autoimmune diseases and acute ischaemic stroke. This Review provides an overview of TWEAK-Fn14 axis biology and summarizes the available data supporting the proposal that both TWEAK and Fn14 should be considered as potential targets for the development of novel therapeutics.
Collapse
|
1775
|
Shukla M, Gupta K, Rasheed Z, Khan KA, Haqqi TM. Consumption of hydrolyzable tannins-rich pomegranate extract suppresses inflammation and joint damage in rheumatoid arthritis. Nutrition 2008; 24:733-43. [PMID: 18490140 DOI: 10.1016/j.nut.2008.03.013] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Revised: 03/05/2008] [Accepted: 03/12/2008] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Although consumption of dietary supplements containing pomegranate extract (POMx) by patients with arthritis is on the rise, the efficacy of such preparations in suppressing joint inflammation and damage is not known. The present study was designed to evaluate a standardized preparation of POMx using collagen-induced arthritis (CIA) in mice, a widely used animal model of rheumatoid arthritis. METHODS CIA-susceptible DBA/1 mice were fed POMx by gavage before and after immunization with chicken type II collagen. Severity of clinical arthritis was scored using a visual scoring system. Arthritic joints were analyzed by histopathology and graded. Lysates were generated from mouse joints and levels of anti-type II collagen immunoglobulin G and inflammatory cytokines interleukin (IL)-1beta, IL-6, and tumor necrosis factor-alpha were quantified by enzyme-linked immunosorbent assay. The effect of POMx on lipopolysaccharide-induced nitric oxide production was determined by Griess reaction and mitogen-activated protein kinase activation was studied by western immunoblotting in mouse macrophages. RESULTS Consumption of POMx potently delayed the onset and reduced the incidence of CIA in mice. Severity of arthritis was also significantly lower in POMx-fed animals. Histopathology of the arthritic joints from POMx-fed mice demonstrated reduced joint infiltration by the inflammatory cells, and the destruction of bone and cartilage were alleviated. Levels of IL-6 were significantly decreased in the joints of POMx-fed mice with CIA. In mouse macrophages, POMx abrogated multiple signal transduction pathways and downstream mediators implicated in the pathogenesis of rheumatoid arthritis. CONCLUSION Our studies suggest that inhibition of a spectrum of signal transduction pathways and the downstream pathogenic cellular response by POMx or compounds derived from it may be a useful approach for the prevention of the onset and severity of inflammatory arthritis.
Collapse
Affiliation(s)
- Meenakshi Shukla
- Division of Rheumatic Diseases, Department of Medicine, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, Ohio, USA
| | | | | | | | | |
Collapse
|
1776
|
Cheung KL, Khor TO, Yu S, Kong ANT. PEITC induces G1 cell cycle arrest on HT-29 cells through the activation of p38 MAPK signaling pathway. AAPS JOURNAL 2008; 10:277-81. [PMID: 18473178 DOI: 10.1208/s12248-008-9032-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2007] [Accepted: 03/12/2008] [Indexed: 11/30/2022]
Abstract
Phenethyl isothiocyanate (PEITC), an isothiocyanate abundantly found in cruciferous vegetables have been shown to induce apoptosis through MAPK pathway in prostate and colon cancer cells. In the present study, we investigate the effect of PEITC on cell cycle regulation of HT-29 colon cancer cells. Using flow cytometry and Western blot analyses, we found that PEITC significantly induced G1 cell cycle arrest in HT-29 cells. We showed that the cell cycle arrest was not related to beta-catenin translocation into the nucleus. Interestingly, inhibition of p38 attenuated the cell cycle arrest, suggesting that cell cycle arrest by PEITC was caused by the activation of MAPK pathway. Treatments of PEITC resulted in a dose-dependent down-regulation of cyclins A, D, E and pRb protein expression. The down-regulation can be attributed to the activation of the p38 pathway, since inhibition of its activities by specific inhibitor blocked PEITC's ability to decrease the expression level of cyclins A and D and attenuate cell cycle arrest effect of PEITC. In conclusion, this study shows for the first time that PEITC can arrest HT-29 cells in G1 phase by down-regulation of cyclins through the activation of p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Ka Lung Cheung
- Graduate Program in Pharmaceutical Science, Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | | | | | | |
Collapse
|
1777
|
White DE, Burchill SA. BAY 11-7082 induces cell death through NF-kappaB-independent mechanisms in the Ewing's sarcoma family of tumours. Cancer Lett 2008; 268:212-24. [PMID: 18471963 DOI: 10.1016/j.canlet.2008.03.045] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Revised: 03/27/2008] [Accepted: 03/28/2008] [Indexed: 10/22/2022]
Abstract
The role of NF-kappaB in the Ewing's sarcoma family of tumours (ESFT) and their response to fenretinide has been investigated. Basal levels of phosphorylated NF-kappaB were low in all ESFT cells. BAY 11-7082 decreased cell viability, which was accompanied by caspase-3 cleavage. This was independent of the increase in reactive oxygen species, p38(MAPK) phosphorylation and expression of NF-kappaB target proteins. NF-kappaB knockdown did not induce death under normal growth conditions, but did reduce TNFalpha-dependent cell survival. Fenretinide-induced apoptosis was independent of NF-kappaB. BAY 11-7082-induced cell death through an NF-kappaB-independent mechanism and enhanced cell death when combined with fenretinide.
Collapse
Affiliation(s)
- Danielle E White
- Candlelighter's Children's Cancer Research Group, Leeds Institute of Molecular Medicine, Cancer Research UK Clinical Centre, St James's University Hospital, Leeds, UK.
| | | |
Collapse
|
1778
|
Prickett TD, Ninomiya-Tsuji J, Broglie P, Muratore-Schroeder TL, Shabanowitz J, Hunt DF, Brautigan DL. TAB4 stimulates TAK1-TAB1 phosphorylation and binds polyubiquitin to direct signaling to NF-kappaB. J Biol Chem 2008; 283:19245-54. [PMID: 18456659 DOI: 10.1074/jbc.m800943200] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Responses to transforming growth factor beta and multiple cytokines involve activation of transforming growth factor beta-activated kinase-1 (TAK1) kinase, which activates kinases IkappaB kinase (IKK) and MKK3/6, leading to the parallel activation of NF-kappaB and p38 MAPK. Activation of TAK1 by autophosphorylation is known to involve three different TAK1-binding proteins (TABs). Here we report a protein phosphatase subunit known as type 2A phosphatase-interacting protein (TIP) that also acts as a TAB because it co-precipitates with and directly binds to TAK1, enhances TAK1 autophosphorylation at unique sites, and promotes TAK1 phosphorylation of IKKbeta and signaling to NF-kappaB. Mass spectrometry demonstrated that co-expression of TAB4 protein significantly increased phosphorylation of four sites in TAK1, in a linker region between the kinase and TAB2/3 binding domains, and two sites in TAB1. Recombinant GST-TAB4 bound in an overlay assay directly to inactive TAK1 and activated TAK1 but not TAK1 phosphorylated in the linker sites, suggesting a bind and release mechanism. In kinase assays using TAK1 immune complexes, added GST-TAB4 selectively stimulated IKK phosphorylation. TAB4 co-precipitated polyubiquitinated proteins dependent on a Phe-Pro motif that was required to enhance phosphorylation of TAK1. TAB4 mutated at Phe-Pro dominantly interfered with IL-1beta activation of NF-kappaB involving IKK-dependent but not p38 MAPK-dependent signaling. The results show that TAB4 binds TAK1 and polyubiquitin chains to promote specific sites of phosphorylation in TAK1-TAB1, which activates IKK signaling to NF-kappaB.
Collapse
Affiliation(s)
- Todd D Prickett
- Center for Cell Signaling and Department of Microbiology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | | | |
Collapse
|
1779
|
Vassou D, Bakogeorgou E, Kampa M, Dimitriou H, Hatzoglou A, Castanas E. Opioids modulate constitutive B-lymphocyte secretion. Int Immunopharmacol 2008; 8:634-44. [DOI: 10.1016/j.intimp.2008.01.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 12/17/2007] [Accepted: 01/03/2008] [Indexed: 11/26/2022]
|
1780
|
Yan G, Huang J, Jarbadan NR, Jiang Y, Cheng H. Sequestration of NF-κB Signaling Complexes in Lipid Rafts Contributes to Repression of NF-κB in T Lymphocytes under Hyperthermia Stress. J Biol Chem 2008; 283:12489-500. [DOI: 10.1074/jbc.m707988200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
1781
|
Liu Z, Zhang HM, Yuan J, Lim T, Sall A, Taylor GA, Yang D. Focal adhesion kinase mediates the interferon-gamma-inducible GTPase-induced phosphatidylinositol 3-kinase/Akt survival pathway and further initiates a positive feedback loop of NF-kappaB activation. Cell Microbiol 2008; 10:1787-800. [PMID: 18452580 DOI: 10.1111/j.1462-5822.2008.01165.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Interferon-gamma-inducible GTPase (IGTP) expression is upregulated in coxsackievirus B3 (CVB3)-infected murine heart and inhibits CVB3-induced apoptosis through activation of the PI3 kinase/Akt pathway. However, the mechanism of this pathway activation is unknown. In this study, using doxcycycline-inducible Tet-On HeLa cells that overexpress IGTP, we have demonstrated that focal adhesion kinase (FAK) is phosphorylated in response to IGTP expression and that transfection of the Tet-On HeLa cells with a dominant negative FAK (FRNK) blocks Akt activation. Furthermore, induction of IGTP also promoted the NF-kappaB activation as evidenced by its enhanced nuclear translocation, binding to transcriptional promoters and increased transcriptional activity. However, FRNK transfection and phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 both blocked the IGTP-induced translocation and NF-kappaB activation. Furthermore, silencing NF-kappaB with siRNAs significantly inhibited the phosphorylation of FAK and Akt, but not their total expression levels, indicating that NF-kappaB activation is required for the IGTP-induced activation of FAK and PI3K/Akt. Finally, blocking this survival pathway by transfection of FRNK or silencing of NF-kappaB reduced CVB3 replication and enhanced cell death during CVB3 infection. Taken together, these results suggest that FAK is a mediator upstream of PI3K/Akt and NF-kappaB functions as a downstream effector and also positively regulates the activity of upstream kinases.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Pathology and Laboratory Medicine, University of British Columbia, The James Hogg iCAPTURE Center - St. Paul's Hospital, Vancouver, Canada
| | | | | | | | | | | | | |
Collapse
|
1782
|
Singh NN, Ramji DP. Protein kinase CK2, an important regulator of the inflammatory response? J Mol Med (Berl) 2008; 86:887-97. [PMID: 18437331 DOI: 10.1007/s00109-008-0352-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Revised: 03/11/2008] [Accepted: 03/12/2008] [Indexed: 01/01/2023]
Abstract
Casein kinase 2 (CK2) is a highly conserved serine-threonine kinase that uses both adenosine triphosphate and guanosine triphosphate as phosphate donors. This constitutively active and ubiquitously expressed enzyme is often present as a tetrameric holoenzyme complex of two catalytic subunits (alpha and/or alpha') and two regulatory beta subunits. The enzyme is known to phosphorylate more than 300 substrates and controls a wide range of processes, including the regulation of cell cycle, apoptosis, transformation, and circadian rhythm. Several lines of recent evidence also suggest a potentially important role for CK2 in the control of the inflammatory response. This review will give an overview of CK2 and its regulation and describe the evidence implicating its role in inflammation.
Collapse
Affiliation(s)
- Nishi N Singh
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | | |
Collapse
|
1783
|
Gutti RK, Tsai-Morris CH, Dufau ML. Gonadotropin-regulated testicular helicase (DDX25), an essential regulator of spermatogenesis, prevents testicular germ cell apoptosis. J Biol Chem 2008; 283:17055-64. [PMID: 18430733 DOI: 10.1074/jbc.m708449200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Gonadotropin-regulated testicular helicase (GRTH)/DDX25 is an essential post-transcriptional regulator of spermatogenesis. In GRTH null mice severe apoptosis was observed in spermatocytes entering the metaphase of meiosis. Pro- and anti-apoptotic factors were found to be under GRTH regulation in comparative studies of spermatocytes from wild type and GRTH(-/-) knock-out (KO) mice. KO mice displayed decreased levels of Bcl-2 and Bcl-xL (anti-apoptotic factors), an increase in Bid, Bak, and Bad (pro-apoptotic), reduced phospho-Bad, and release of cytochrome c. Also, an increase on Smac, a competitor of inhibitor apoptotic proteins that release caspases, was observed. These changes caused an increase in cleavage of caspases 9 and 3, activation of caspase 3 and increases in cleavage products of PARP. The half-life of caspase 3 transcripts was markedly increased in KO, indicating that GRTH had a negative role on its mRNA stability. IkappaBalpha, which sequesters NF-kappaB from its transcriptional activation of pro-apoptotic genes, was highly elevated in KO, and its phospho-form, which promotes its dissociation, was reduced. The increase of HDAC1 and abolition of p300 expression in KO indicated a nuclear action of GRTH on the NF-kappaB-mediated transcription of anti-apoptotic genes. It also regulates the associated death domain pathway and caspase 8-mediated events. GRTH-mediated apoptotic regulation was further indicated by its selective binding to pro- and anti-apoptotic mRNAs. These studies have demonstrated that GRTH, as a component of mRNP particles, acts as a negative regulator of the tumor necrosis factor receptor 1 and caspase pathways and promotes NF-kappaB function to control apoptosis in spermatocytes of adult mice.
Collapse
Affiliation(s)
- Ravi K Gutti
- Section on Molecular Endocrinology, Endocrinology and Reproduction Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892-4510, USA
| | | | | |
Collapse
|
1784
|
Abstract
OBJECTIVE Since imiquimod, a nucleoside analogue of the imidazoquinoline family, has shown efficacy against many tumour entities, its mode of action has become a focus of scientific interest. RESULTS The major biologic effects of imiquimod are mediated through agonistic activity towards toll-like receptors (TLR) 7 and 8, and consecutively, activation of nuclear factor-kappa B (NF-kappaB). The result of this activity is the induction of pro-inflammatory cytokines, chemokines and other mediators leading to activation of antigen-presenting cells and other components of innate immunity and, eventually, the mounting of a profound T-helper (Th1)-weighted antitumoral cellular immune response. Several secondary effects on the molecular and cellular level may also be explained, at least in part, by the activation of NF-kappaB. Moreover, independent of TLR-7 and TLR-8, imiquimod appears to interfere with adenosine receptor signalling pathways, and the compound causes receptor-independent reduction of adenylyl cyclase activity. This novel mechanism may augment the pro-inflammatory activity of the compound through suppression of a negative regulatory feedback mechanism which normally limits inflammatory responses. Finally, imiquimod induces apoptosis of tumour cells at higher concentrations. The pro-apoptotic activity of imiquimod involves caspase activation and appears to depend on B cell lymphoma/leukemia protein (Bcl)-2 proteins. CONCLUSIONS Overall, imiquimod acts on several levels, which appear to synergistically underlie the profound antitumoral activity of the compound.
Collapse
Affiliation(s)
- M P Schön
- Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine and Department of Dermatology, Julius-Maximilians-University, Würzburg, Germany.
| | | |
Collapse
|
1785
|
|
1786
|
Guan YS, He Q, Wang MQ, Li P. Nuclear factor kappa B and hepatitis viruses. Expert Opin Ther Targets 2008; 12:265-80. [PMID: 18269337 DOI: 10.1517/14728222.12.3.265] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Hepatitis can be caused by a number of viruses, which have similar clinical manifestations and render infected individuals at high risk of death from cirrhosis and liver cancer. Current therapies for hepatitis have limited effects and unsatisfactory patient outcomes. Nuclear factor kappa B (NF-kappaB) is critical for immune and inflammatory responses. During its lifetime the cell demands specific and highly regulated control of NF-kappaB activity. OBJECTIVE To develop novel strategies to overcome various hepatitides and related liver cancer with NF-kappaB as the key point. METHODS All aspects of NF-kappaB control with regard to hepatitis are covered. RESULTS/CONCLUSION NF-kappaB plays an important role in the process of hepatitis and is hypothesized to be an anti-cancer factor in the subsequent inflammation-associated hepatocarcinogenesis.
Collapse
Affiliation(s)
- Yong-Song Guan
- West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China.
| | | | | | | |
Collapse
|
1787
|
Higuchi Y, Kawakami S, Hashida M. [Development of cell-selective targeting systems of NFkappaB decoy for inflammation therapy]. YAKUGAKU ZASSHI 2008; 128:209-18. [PMID: 18239368 DOI: 10.1248/yakushi.128.209] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
NFkappaB regulate several inflammatory related molecules and evoke immune and inflammatory response by several stimuli, therefore inhibition of NFkappaB activation would be a novel therapeutic strategy. To date, there are many conventional drugs including nonsteroldal or steroldal anti-inflammatory drugs or immune suppressors etc. were known to inhibit NFkappaB activation, however, several side effects were also reported. Recently, double stranded oligonucleotide including NFkappaB binding sequence, called NFkappaB decoy, was developed to prevent NFkappaB activation, which is powerful tool in a new class of anti-gene strategy for molecular therapy with low side effect. However, NFkappaB decoy is easily degraded by nuclease and rapidly excreted to urine, therefore it is necessary to develop carrier for NFkappaB decoy therapy. Here, we shall review delivery system for NFkappaB decoy and introduce our cell-selective delivery system for NFkappaB decoy using sugar decorated cationic liposomes.
Collapse
Affiliation(s)
- Yuriko Higuchi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Japan
| | | | | |
Collapse
|
1788
|
Sharma CS, Sarkar S, Periyakaruppan A, Ravichandran P, Sadanandan B, Ramesh V, Thomas R, Hall JC, Wilson BL, Ramesh GT. Simulated microgravity activates apoptosis and NF-kappaB in mice testis. Mol Cell Biochem 2008; 313:71-8. [PMID: 18385949 DOI: 10.1007/s11010-008-9743-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2008] [Accepted: 03/13/2008] [Indexed: 10/22/2022]
Abstract
Microgravity is known to have significant effect on all aspects of reproductive function in animal models. Recent studies have also shown that microgravity induces changes at the cellular level, including apoptosis. Our effort here was to study the effect of simulated microgravity on caspase-8 and the caspase-3 activities, the effectors of the apoptotic pathway and on the transcription factor NF-kappaB a signaling molecule in mouse testis. Morey-Holton hind limb suspension model was used to simulate microgravity. Caspase-8 and 3 fluorometric assays were carried out and HLS mice testis exhibited a 51% increase in caspase-8 and caspase-3 compared to the controls. A sandwich ELISA-based immunoassay was carried out for detection of NF-kappaB which again significantly increased in the test mice. Testosterone levels were measured using an ELISA kit and in HLS mice the decrease was significant. There was also a significant decrease in testis weight in the test mice. Simulated microgravity activates caspase 8, 3 and NF-kappaB necessary to stimulate the apoptotic pathway in mice testis. This may account for the drop in testis weight and testosterone level further affecting testicular physiology and function.
Collapse
Affiliation(s)
- Chidananda S Sharma
- Department of Biology, Molecular Toxicology Laboratory, Center for Biotechnology & Biomedical Sciences, Norfolk State University, Norfolk, VA 23504, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1789
|
Chae M, Kim K, Park SM, Jang IS, Seo T, Kim DM, Kim IC, Lee JH, Park J. IRF-2 regulates NF-kappaB activity by modulating the subcellular localization of NF-kappaB. Biochem Biophys Res Commun 2008; 370:519-24. [PMID: 18395009 DOI: 10.1016/j.bbrc.2008.03.136] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2008] [Accepted: 03/28/2008] [Indexed: 01/28/2023]
Abstract
Nuclear Factor-kappa B (NF-kappaB) is a transcription factor essential to the control of cell proliferation, survival, differentiation, immune response, and inflammation. Constitutive NF-kappaB activation has been observed in a broad variety of solid tumors and hematological malignancies, which suggests that NF-kappaB signaling may perform a critical role in the development of human cancers. Interferon regulatory factor-2 (IRF-2), an antagonistic transcriptional repressor of IRF-1, evidences oncogenic potential, but little is currently known regarding the mechanism underlying the oncogenic activities of IRF-2. In this study, we report that IRF-2 recruits RelA/p65 transcription factors into the nucleus via physical interaction. While the nuclear recruitment of RelA by IRF-2 augments TNFalpha-induced NF-kappaB dependent transcription, the N-terminal truncated mutant form of IRF-2 inhibits the nuclear localization of RelA, and thus interferes with NF-kappaB activation. Furthermore, the knockdown of IRF-2 by IRF-2 siRNA attenuates TNFalpha-induced NF-kappaB dependent transcription by inhibiting the nuclear localization of RelA. Thus, these results show that IRF-2 regulates NF-kappaB activity via the modulation of NF-kappaB subcellular localization.
Collapse
Affiliation(s)
- Myounghee Chae
- Korea Basic Science Institute, Gwangju Center, 300, Yongbong-Dong, Book-Ku, Gwangju 500-757, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
1790
|
Anticarcinogenesis by dietary phytochemicals: Cytoprotection by Nrf2 in normal cells and cytotoxicity by modulation of transcription factors NF-κB and AP-1 in abnormal cancer cells. Food Chem Toxicol 2008; 46:1257-70. [DOI: 10.1016/j.fct.2007.09.082] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2007] [Revised: 08/20/2007] [Accepted: 09/03/2007] [Indexed: 01/09/2023]
|
1791
|
Nakajima H, Fujiwara H, Furuichi Y, Tanaka K, Shimbara N. A novel small-molecule inhibitor of NF-κB signaling. Biochem Biophys Res Commun 2008; 368:1007-13. [DOI: 10.1016/j.bbrc.2008.01.166] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Accepted: 01/26/2008] [Indexed: 10/22/2022]
|
1792
|
Salminen A, Huuskonen J, Ojala J, Kauppinen A, Kaarniranta K, Suuronen T. Activation of innate immunity system during aging: NF-kB signaling is the molecular culprit of inflamm-aging. Ageing Res Rev 2008; 7:83-105. [PMID: 17964225 DOI: 10.1016/j.arr.2007.09.002] [Citation(s) in RCA: 410] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2007] [Revised: 09/14/2007] [Accepted: 09/14/2007] [Indexed: 10/22/2022]
Abstract
Innate and adaptive immunity are the major defence mechanisms of higher organisms against inherent and environmental threats. Innate immunity is present already in unicellular organisms but evolution has added novel adaptive immune mechanisms to the defence armament. Interestingly, during aging, adaptive immunity significantly declines, a phenomenon called immunosenescence, whereas innate immunity seems to be activated which induces a characteristic pro-inflammatory profile. This process is called inflamm-aging. The recognition and signaling mechanisms involved in innate immunity have been conserved during evolution. The master regulator of the innate immunity is the NF-kB system, an ancient signaling pathway found in both insects and vertebrates. The NF-kB system is in the nodal point linking together the pathogenic assault signals and cellular danger signals and then organizing the cellular resistance. Recent studies have revealed that SIRT1 (Sir2 homolog) and FoxO (DAF-16), the key regulators of aging in budding yeast and Caenorhabditis elegans models, regulate the efficiency of NF-kB signaling and the level of inflammatory responses. We will review the role of innate immunity signaling in the aging process and examine the function of NF-kB system in the organization of defence mechanisms and in addition, its interactions with the protein products of several gerontogenes. Our conclusion is that NF-kB signaling seems to be the culprit of inflamm-aging, since this signaling system integrates the intracellular regulation of immune responses in both aging and age-related diseases.
Collapse
|
1793
|
Subramaniam D, Ramalingam S, May R, Dieckgraefe BK, Berg DE, Pothoulakis C, Houchen CW, Wang TC, Anant S. Gastrin-mediated interleukin-8 and cyclooxygenase-2 gene expression: differential transcriptional and posttranscriptional mechanisms. Gastroenterology 2008; 134:1070-82. [PMID: 18395088 DOI: 10.1053/j.gastro.2008.01.040] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Accepted: 01/04/2008] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Gastrin induces the expression of cyclooxygenase (COX)-2 and interleukin (IL)-8; however, the mechanism(s), especially in gastric epithelial cells, is not well understood. Here, we have determined the intracellular mechanisms mediating gastrin-dependent gene expression. METHODS AGS-E human gastric cancer cell line stably expressing cholecystokinin-2 receptor was treated with amidated gastrin-17. Real-time polymerase chain reaction, Western blot, and enzyme-linked immunosorbent assay were performed to determine COX-2 and IL-8 expression and Akt, Erk, and p38 phosphorylation. Gene promoter activity was determined by luciferase assay. Electrophoretic mobility shift assay analysis was performed for nuclear factor kappaB (NF-kappaB) and activator protein-1 activity. RNA stability was determined after actinomycin D treatment. HuR localization was determined by immunocytochemistry. RESULTS Gastrin induced COX-2 and IL-8 expression in AGS-E cells, which was inhibited by phosphatidylinositol 3' kinase (PI3K) and p38 inhibitors. Gastrin-mediated Akt activation was observed to be downstream of p38. IL-8 expression was dependent on COX-2-mediated prostaglandin E(2) synthesis. In the presence of an NF-kappaB inhibitor MG132, IL-8 transcription was inhibited, but not that of COX-2. This was confirmed after knockdown of the p65 RelA subunit of NF-kappaB. Further studies showed that COX-2 gene transcription is regulated by activator protein-1. Gastrin increased the stability of both COX-2 and IL-8 messenger RNA (mRNA) in a p38-dependent manner, the half-life increasing from 31 minutes to 8 hours and approximately 4 hours, respectively. Gastrin, through p38 activity, also enhanced HuR expression, nucleocytoplasmic translocation, and enhanced COX-2 mRNA binding. CONCLUSIONS Gastrin differentially induces COX-2 and IL-8 expression at the transcriptional and posttranscriptional levels by PI3K and p38 mitogen-activated protein kinase pathways, respectively.
Collapse
Affiliation(s)
- Dharmalingam Subramaniam
- Department of Internal Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
1794
|
Watson MR, Wallace K, Gieling RG, Manas DM, Jaffray E, Hay RT, Mann DA, Oakley F. NF-kappaB is a critical regulator of the survival of rodent and human hepatic myofibroblasts. J Hepatol 2008; 48:589-97. [PMID: 18279996 DOI: 10.1016/j.jhep.2007.12.019] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Revised: 12/06/2007] [Accepted: 12/26/2007] [Indexed: 12/04/2022]
Abstract
BACKGROUND/AIMS Hepatic myofibroblast activation during injury causes deposition of extracellular matrix within the liver and promotes development of fibrosis. Hepatic myofibroblast apoptosis is associated with remodelling of fibrotic extracellular matrix and regression of fibrosis. Previous work showed that inhibition of constitutive NF-kappaB signaling promotes hepatic myofibroblast apoptosis and resolution of fibrosis in rodent models. However, to date agents used to target constitutive NF-kappaB transcriptional activity in hepatic myofibroblasts have been relatively non-specific with potential for off-target effects that may complicate data interpretation. Likewise, rat chronic liver disease models may not accurately recapitulate the activation of human hepatic myofibroblasts. METHODS We used a mutant recombinant IkappaBalpha super-repressor fused to the HIV-TAT domain to specifically target NF-kappaB signaling in hepatic myofibroblasts. Inhibition of NF-kappaB activity was measured using reporter assay. Apoptosis of hepatic myofibroblasts was assessed by morphological changes, cleavage of the PARP-1 protein and Caspase 3 activation. RESULTS TAT-IkappaBalphaSR reduced NF-kappaB dependent transcription, Bcl-2 expression and promoted Jun-N-terminal kinase-dependent apoptosis in human and rat hepatic myofibroblasts. CONCLUSIONS These data highlight the conserved role of NF-kappaB during fibrogenesis. Our data validate the use of rodent models for pre-clinical testing of NF-kappaB inhibitors as anti-fibrotics and stimulators of fibrotic extracellular matrix remodelling.
Collapse
Affiliation(s)
- Martha R Watson
- Liver Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle NE2 4HH, UK
| | | | | | | | | | | | | | | |
Collapse
|
1795
|
|
1796
|
Chau TL, Gioia R, Gatot JS, Patrascu F, Carpentier I, Chapelle JP, O'Neill L, Beyaert R, Piette J, Chariot A. Are the IKKs and IKK-related kinases TBK1 and IKK-epsilon similarly activated? Trends Biochem Sci 2008; 33:171-80. [PMID: 18353649 DOI: 10.1016/j.tibs.2008.01.002] [Citation(s) in RCA: 177] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Revised: 01/04/2008] [Accepted: 01/17/2008] [Indexed: 02/06/2023]
Abstract
The IkappaB kinases (IKKs) IKK-alpha and IKK-beta, and the IKK-related kinases TBK1 and IKK-epsilon, have essential roles in innate immunity through signal-induced activation of NF-kappaB, IRF3 and IRF7, respectively. Although the signaling events within these pathways have been extensively studied, the mechanisms of IKK and IKK-related complex assembly and activation remain poorly defined. Recent data provide insight into the requirement for scaffold proteins in complex assembly; NF-kappaB essential modulator coordinates some IKK complexes, whereas TANK, NF-kappaB-activating kinase-associated protein 1 (NAP1) or similar to NAP1 TBK1 adaptor (SINTBAD) assemble TBK1 and IKK-epsilon complexes. The different scaffold proteins undergo similar post-translational modifications, including phosphorylation and non-degradative polyubiquitylation. Moreover, increasing evidence indicates that distinct scaffold proteins assemble IKK, and potentially TBK1 and IKK-epsilon subcomplexes, in a stimulus-specific manner, which might be a mechanism to achieve specificity.
Collapse
Affiliation(s)
- Tieu-Lan Chau
- Interdisciplinary Cluster for Applied Genoproteomics, University of Liège, Sart-Tilman, 4000 Liège, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1797
|
Olivotto E, Borzi RM, Vitellozzi R, Pagani S, Facchini A, Battistelli M, Penzo M, Li X, Flamigni F, Li J, Falcieri E, Facchini A, Marcu KB. Differential requirements for IKKalpha and IKKbeta in the differentiation of primary human osteoarthritic chondrocytes. ACTA ACUST UNITED AC 2008; 58:227-39. [PMID: 18163512 DOI: 10.1002/art.23211] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Osteoarthritic (OA) chondrocytes behave in an intrinsically deregulated manner, characterized by chronic loss of healthy cartilage and inappropriate differentiation to a hypertrophic-like state. IKKalpha and IKKbeta are essential kinases that activate NF-kappaB transcription factors, which in turn regulate cell differentiation and inflammation. This study was undertaken to investigate the differential roles of each IKK in chondrocyte differentiation and hypertrophy. METHODS Expression of IKKalpha or IKKbeta was ablated in primary human chondrocytes by retro-transduction of specific short-hairpin RNAs. Micromass cultures designed to reproduce chondrogenesis with progression to the terminal hypertrophic stage were established, and anabolism and remodeling of the extracellular matrix (ECM) were investigated in the micromasses using biochemical, immunohistochemical, and ultrastructural techniques. Cellular parameters of hypertrophy (i.e., proliferation, viability, and size) were also analyzed. RESULTS The processes of ECM remodeling and mineralization, both characteristic of terminally differentiated hypertrophic cells, were defective following the loss of IKKalpha or IKKbeta. Silencing of IKKbeta markedly enhanced accumulation of glycosaminoglycan in conjunction with increased SOX9 expression. Ablation of IKKalpha dramatically enhanced type II collagen deposition independent of SOX9 protein levels but in association with suppressed levels of runt-related transcription factor 2. Moreover, IKKalpha-deficient cells retained the phenotype of cells in a pre-hypertrophic-like state, as evidenced by the smaller size and faster proliferation of these cells prior to micromass seeding, along with the enhanced viability of their differentiated micromasses. CONCLUSION IKKalpha and IKKbeta exert differential roles in ECM remodeling and endochondral ossification, which are events characteristic of hypertrophic chondrocytes and also complicating factors often found in OA. Because the effects of IKKalpha were more profound and pleotrophic in nature, our observations suggest that exacerbated IKKalpha activity may be responsible, at least in part, for the characteristic abnormal phenotypes of OA chondrocytes.
Collapse
|
1798
|
Souto-Carneiro MM, Fritsch R, Sepúlveda N, Lagareiro MJ, Morgado N, Longo NS, Lipsky PE. The NF-kappaB canonical pathway is involved in the control of the exonucleolytic processing of coding ends during V(D)J recombination. THE JOURNAL OF IMMUNOLOGY 2008; 180:1040-9. [PMID: 18178844 DOI: 10.4049/jimmunol.180.2.1040] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
V(D)J recombination is essential to produce an Ig repertoire with a large range of Ag specificities. Although NF-kappaB-binding sites are present in the human and mouse IgH, Igkappa, and Iglambda enhancer modules and RAG expression is controlled by NF-kappaB, it is not known whether NF-kappaB regulates V(D)J recombination mechanisms after RAG-mediated dsDNA breaks. To clarify the involvement of NF-kappaB in human V(D)J recombination, we amplified Ig gene rearrangements from individual peripheral B cells of patients with X-linked anhidrotic ectodermal dysplasia with hyper-IgM syndrome (HED-ID) who have deficient expression of the NF-kappaB essential modulator (NEMO/Ikkgamma). The amplification of nonproductive Ig gene rearrangements from HED-ID B cells reflects the influence of the Ikkgamma-mediated canonical NF-kappaB pathway on specific molecular mechanisms involved in V(D)J recombination. We found that the CDR3(H) from HED-ID B cells were abnormally long, as a result of a marked reduction in the exonuclease activity on the V, D, and J germline coding ends, whereas random N-nucleotide addition and palindromic overhangs (P nucleotides) were comparable to controls. This suggests that an intact canonical NF-kappaB pathway is essential for normal exonucleolytic activity during human V(D)J recombination, whereas terminal deoxynucleotide transferase, Artemis, and DNA-dependent protein kinase catalytic subunit activity are not affected. The generation of memory B cells and somatic hypermutation were markedly deficient confirming a role for NF-kappaB in these events of B cell maturation. However, selection of the primary B cell repertoire appeared to be intact and was partially able to correct the defects generated by abnormal V(D)J recombination.
Collapse
Affiliation(s)
- M Margarida Souto-Carneiro
- Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892-1560, USA
| | | | | | | | | | | | | |
Collapse
|
1799
|
Schmid JA, Birbach A. IkappaB kinase beta (IKKbeta/IKK2/IKBKB)--a key molecule in signaling to the transcription factor NF-kappaB. Cytokine Growth Factor Rev 2008; 19:157-65. [PMID: 18308615 DOI: 10.1016/j.cytogfr.2008.01.006] [Citation(s) in RCA: 186] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
IKKbeta/IKBKB (IkappaB kinase beta), also designated as IKK2, was named after its function of phosphorylating IkappaB molecules, the inhibitors of NF-kappaB transcription factors. The kinase activity of IKKbeta targets two adjacent serine residues of IkappaB leading to ubiquitination and proteasomal degradation of the inhibitor, followed by release and activation of NF-kappaB. Many signaling pathways that activate NF-kappaB converge at the level of IKKbeta. Examples of stimuli leading to IKKbeta and subsequent NF-kappaB activation include inflammatory cytokines (IL-1, TNFalpha), endotoxins (lipopolysaccharide), viral infection and double strand RNA as well as physical signals such as UV-irradiation. Transcription factors of the NF-kappaB protein family have a great variety of functions in regulating the immune system, cellular differentiation, survival and proliferation. NF-kappaB is an essential factor in acute as well as chronic inflammation, a pathological state which is either cause or co-factor in a great variety of diseases. Moreover, recent data suggest that many variants of cancer are characterized by elevated constitutive activity of NF-kappaB, which can act as a survival factor for malignant cells by its predominantly anti-apoptotic function. Given the tight regulation of NF-kappaB by IkappaB molecules and the central role of IKKbeta in phosphorylation and degradation of the inhibitor, IKKbeta is a very promising target for pharmaceutical substances aiming at interfering with NF-kappaB activation.
Collapse
Affiliation(s)
- Johannes A Schmid
- Center for Biomolecular Medicine and Pharmacology, Medical University Vienna, Austria.
| | | |
Collapse
|
1800
|
Saha A, Hammond CE, Trojanowska M, Smolka AJ. Helicobacter pylori-induced H,K-ATPase alpha-subunit gene repression is mediated by NF-kappaB p50 homodimer promoter binding. Am J Physiol Gastrointest Liver Physiol 2008; 294:G795-807. [PMID: 18202112 DOI: 10.1152/ajpgi.00431.2007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Infection of human gastric body mucosa by the gram-negative, microaerophilic bacterium Helicobacter pylori induces an inflammatory response and a transitory hypochlorhydria that progresses in approximately 2% of patients to atrophic gastritis, dysplasia, and gastric adenocarcinoma. We have previously shown that H. pylori infection of cultured gastric epithelial cells (AGS) represses the activity of the transfected alpha-subunit (HKalpha) promoter of H,K-ATPase, the parietal cell enzyme mediating acid secretion. However, the mechanistic details of H. pylori-mediated repression of HKalpha and ensuing hypochlorhydria are unknown. H. pylori is known to upregulate the transcription factor NF-kappaB through the ERK1/2 MAPK pathway. We identified NF-kappaB-binding regions in the HKalpha promoter and found that H. pylori inoculation of AGS cells increased NF-kappaB p50 binding to the transfected HKalpha promoter and repressed its transcriptional activity. Immunoblot and DNA-protein interaction studies showed that although active phosphorylated NF-kappaB p65 is present in H. pylori-infected AGS cells, an NF-kappaB p50/p65 heterodimeric complex fails to bind to the HKalpha promoter. Point mutations at -159 and -161 bp in the HKalpha promoter NF-kappaB binding sequence prevented binding of NF-kappaB p50 and prevented H. pylori repression of point-mutated HKalpha promoter activity in transfected AGS cells. Small interfering RNA-mediated knockdown of NF-kappaB p50 in H. pylori-infected AGS cells also abrogated H. pylori-induced HKalpha repression, whereas NF-kappaB p65 knockdown did not. We conclude that H. pylori inhibits HKalpha gene expression by ERK1/2-mediated NF-kappaB p50 homodimer binding to the HKalpha promoter. This study identifies a novel pathogen-dependent mechanism of H,K-ATPase inhibition and contributes to understanding of H. pylori pathophysiology.
Collapse
Affiliation(s)
- Arindam Saha
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | |
Collapse
|