51
|
Jiao BY, Lin WS, She FF, Chen WN, Lin X. Hepatitis B virus X protein enhances activation of nuclear factor κB through interaction with valosin-containing protein. Arch Virol 2011; 156:2015-21. [PMID: 21918864 DOI: 10.1007/s00705-011-1099-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 08/29/2011] [Indexed: 12/26/2022]
Abstract
Hepatitis B virus X protein (HBx protein) is a multifunctional regulatory protein. The transactivation of nuclear factor kappa B (NF-κB) by HBx protein has been shown to be of importance in the pathogenesis of HBV-related diseases. However, the mechanism involved remains largely unclear. In this study, a CytoTrap yeast two-hybrid system was employed to screen binding partners of the HBx protein; 29 cellular proteins, including valosin-containing protein (VCP), were identified. The interaction between HBx protein and VCP was further confirmed in vitro and in vivo using a glutathione S-transferase pull-down assay and co-immunoprecipitation, respectively. It was also shown that this interaction is mediated by amino acid residues 51-120 of the HBx protein. In Huh-7 hepatoma cells, HBx protein enhanced the VCP-mediated activation of NF-κB. Our findings provide new insights into the molecular mechanisms that lead to the activation of NF-κB by HBx protein.
Collapse
Affiliation(s)
- Bo-Yan Jiao
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Research Center of Molecular Medicine, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350004, People's Republic of China
| | | | | | | | | |
Collapse
|
52
|
Wang Y, Yang YS, Tang XC, Zhang HY. T33, a novel peroxisome proliferator-activated receptor γ/α agonist, exerts neuroprotective action via its anti-inflammatory activities. Acta Pharmacol Sin 2011; 32:1100-8. [PMID: 21804572 DOI: 10.1038/aps.2011.69] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIM To examine the neuroprotective effects of T33, a peroxisome proliferator-activated receptor gamma/alpha (PPARγ/α) agonist, in acute ischemic models in vitro and in vivo. METHODS Primary astrocytes subjected to oxygen-glucose deprivation/reperfusion (O/R) and BV-2 cells subjected to hypoxia were used as a model simulating the ischemic core and penumbra, respectively. The mRNA levels of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) were measured using qPCR. The levels of TNF-α secreted by BV-2 cells were measured using ELISA. Protein levels of cyclooxygenase-2 (COX-2), p65, phosphorylated I-κBα/I-κBα, phosphorylated I-κB kinase (pIKK), phosphorylated eukaryote initiation factor 2α (p-eIF-2α)/eIF-2α and p-p38/p38 were detected using Western blot. PPARγ activity was measured using EMSA. The neuroprotection in vivo was examined in rat middle cerebral artery occlusion (MCAO) model with neurological scoring and TTC staining. RESULTS Addition of T33 (0.5 μmol/L) increased the level of I-κBα protein in primary astrocytes subjected to O/R, which was due to promoting protein synthesis without affecting degradation. In primary astrocytes subjected to O/R, addition of T33 amplified I-κBα gene transcription and mRNA translation, thus suppressing the nuclear factor-kappa B (NF-κB) pathway and reducing inflammatory mediators (TNF-α, IL-1β, and COX-2). In BV-2 cells subjected to hypoxia, T33 (0.5 μmol/L) reduced TNF-α, COX-2, and p-P38 production, which was antagonized by pre-administration of the specific PPARγ antagonist GW9662 (30 μmol/L). T33 (2 mg/kg, ip) attenuated MCAO-induced inflammatory responses and brain infarction, which was antagonized by pre-administered GW9662 (4 mg/kg, ip). CONCLUSION T33 exerted anti-inflammatory effects in the ischemic core and penumbra via PPARγ activation, which contributed to its neuroprotective action.
Collapse
|
53
|
Plant polyphenols attenuate hepatic injury after hemorrhage/resuscitation by inhibition of apoptosis, oxidative stress, and inflammation via NF-kappaB in rats. Eur J Nutr 2011; 51:311-21. [PMID: 21698494 DOI: 10.1007/s00394-011-0216-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 06/01/2011] [Indexed: 01/04/2023]
Abstract
PURPOSE Oxidative stress and inflammation contribute to hepatic injury after hemorrhage/resuscitation (H/R). Natural plant polyphenols, i.e., green tea extract (GTE) possess high anti-oxidant and anti-inflammatory activities in various models of acute inflammation. However, possible protective effects and feasible mechanisms by which plant polyphenols modulate pro-inflammatory, apoptotic, and oxidant signaling after H/R in the liver remain unknown. Therefore, we investigated the effects of GTE and its impact on the activation of NF-kappaB in the pathogenesis of hepatic injury induced by H/R. METHODS Twenty-four female LEWIS rats (180-250 g) were fed a standard chow (ctrl) or a diet containing 0.1% polyphenolic extracts (GTE) from Camellia sinensis starting 5 days before H/R. Rats were hemorrhaged to a mean arterial pressure of 30 ± 2 mmHg for 60 min and resuscitated (H/R and GTE H/R groups). Control groups (sham, ctrl, and GTE) underwent surgical procedures without H/R. Two hours after resuscitation, tissues were harvested. RESULTS Plasma alanine aminotransferase (ALT) and lactate dehydrogenase (LDH) increased 3.5-fold and fourfold, respectively, in vehicle-treated rats as compared to GTE-fed rats. Histopathological analysis revealed significantly decreased hepatic necrosis and apoptosis in GTE-fed rats after H/R. Real-time PCR showed that GTE diminished gene expression of pro-apoptotic caspase-8 and Bax, while anti-apoptotic Bcl-2 was increased after H/R. Hepatic oxidative (4-hydroxynonenal) and nitrosative (3-nitrotyrosine) stress as well as systemic IL-6 level and hepatic IL-6 mRNA were markedly reduced in GTE-fed rats compared with controls after H/R. Plant polyphenols also decreased the activation of both JNK and NFκB. CONCLUSIONS Taken together, GTE application blunts hepatic damage, apoptotic, oxidative, and pro-inflammatory changes after H/R. These results underline the important roles of JNK and NF-kappaB in inflammatory processes after H/R and the beneficial impact of plant polyphenols in preventing their activation.
Collapse
|
54
|
Effects of green tea catechins on the pro-inflammatory response after haemorrhage/resuscitation in rats. Br J Nutr 2011; 105:1791-7. [PMID: 21294935 DOI: 10.1017/s000711451000560x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Plant polyphenols, i.e. green tea extract (GTE), possess high antioxidative and anti-inflammatory capacity, thus being protective in various models of acute inflammation. However, their anti-inflammatory effect and a feasible mechanism in haemorrhage/resuscitation (H/R)-induced liver injury remain unknown. We investigated the effects of GTE and the role of NF-κB in the pathogenesis of liver injury induced by H/R, and their effects on intercellular adhesion molecule-1 (ICAM-1) expression and neutrophil infiltration. Female Lewis rats were fed a standard chow diet (control, ctrl) or a diet containing 0·1 % polyphenolic GTE for five consecutive days before H/R. Rats were haemorrhaged to a mean arterial pressure of 30 (sem 2) mmHg for 60 min and resuscitated. Control groups (sham_ctrl and sham_GTE) underwent surgical procedures without H/R. At 2 h after resuscitation, tissues were harvested. Serum alanine aminotransferase (ALT) and IL-6 were measured. Hepatic necrosis, ICAM-1 expression and polymorphonuclear leucocyte (PMNL) infiltration were assessed. Hepatic expression of IκBα (phospho) was measured. H/R induced strong liver damage with increased necrosis and serum ALT levels. Compared with both sham groups, inflammatory markers (serum IL-6 and hepatic PMNL infiltration) were elevated after H/R (P < 0·05). Also, H/R increased IκBα phosphorylation. GTE administration markedly (P < 0·05) decreased serum ALT and IL-6 levels, hepatic necrosis as well as PMNL infiltration and the expression of ICAM-1 and phosphorylated IκBα compared with H/R. In conclusion, we observed that NF-κB activation plays an important role in the pathogenesis of liver injury after H/R through the up-regulation of hepatic ICAM-1 expression and subsequent PMNL infiltration. GTE pre-treatment prevents liver damage in this model of acute inflammation through a NF-κB-dependent mechanism.
Collapse
|
55
|
NF-kappaB activation stimulates transcription and replication of retrovirus XMRV in human B-lineage and prostate carcinoma cells. J Virol 2011; 85:3179-86. [PMID: 21270144 DOI: 10.1128/jvi.02333-10] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Xenotropic murine leukemia virus-related virus (XMRV) is a gammaretrovirus linked to prostate carcinoma and chronic fatigue syndrome. Here we report that NF-κB activation can markedly increase XMRV production. The inflammatory cytokine tumor necrosis factor alpha (TNF-α), which activates NF-κB, significantly augmented viral Gag protein production in XMRV-infected cells. Reporter assays showed that TNF-α and Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1), an intrinsic NF-κB activator, increased long terminal repeat (LTR)-dependent XMRV transcription. We identified two NF-κB binding sites (designated κB-1 and κB-2) in the LTR U3 region of XMRV and demonstrated that both sites bind to the NF-κB component p65/RelA. Mutation of the κB-1 site, but not the κB-2 site, impaired responsiveness to TNF-α and LMP1 in reporter assays. A mutant XMRV with a mutation at the κB-1 site replicated significantly less efficiently than the wild-type XMRV in the prostate carcinoma LNCaP, DU145, and PC-3 cell lines, HEK293 cells, the EBV-immortalized cell line IB4, and the Burkitt's lymphoma cell line BJAB. These results demonstrate that TNF-α and EBV LMP1 enhance XMRV replication in prostate carcinoma and B-lineage cells through the κB-1 site in the XMRV LTR, suggesting that inflammation, EBV infection, and other conditions leading to NF-κB activation may promote XMRV spread in humans.
Collapse
|
56
|
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: 24] [Impact Index Per Article: 1.6] [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]
|
57
|
Regulation of IkappaBalpha function and NF-kappaB signaling: AEBP1 is a novel proinflammatory mediator in macrophages. Mediators Inflamm 2010; 2010:823821. [PMID: 20396415 PMCID: PMC2855089 DOI: 10.1155/2010/823821] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2009] [Accepted: 01/12/2010] [Indexed: 02/08/2023] Open
Abstract
NF-κB comprises a family of transcription factors that are critically involved in various inflammatory processes. In this paper, the role of NF-κB in inflammation and atherosclerosis and the regulation of the NF-κB signaling pathway are summarized. The structure, function, and regulation of the NF-κB inhibitors, IκBα and IκBβ, are reviewed. The regulation of NF-κB activity by glucocorticoid receptor (GR) signaling and IκBα sumoylation is also discussed. This paper focuses on the recently reported regulatory function that adipocyte enhancer-binding protein 1 (AEBP1) exerts on NF-κB transcriptional activity in macrophages, in which AEBP1 manifests itself as a potent modulator of NF-κB via physical interaction with IκBα and a critical mediator of inflammation. Finally, we summarize the regulatory roles that recently identified IκBα-interacting proteins play in NF-κB signaling. Based on its proinflammatory roles in macrophages, AEBP1 is anticipated to serve as a therapeutic target towards the treatment of various inflammatory conditions and disorders.
Collapse
|
58
|
Kim EJ, Raval AP, Hirsch N, Perez-Pinzon MA. Ischemic preconditioning mediates cyclooxygenase-2 expression via nuclear factor-kappa B activation in mixed cortical neuronal cultures. Transl Stroke Res 2010; 1:40-7. [PMID: 20606709 PMCID: PMC2893355 DOI: 10.1007/s12975-009-0006-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nuclear factor-kappaB (NF-κB) activation occurs following ischemic preconditioning (IPC) in brain. However, the upstream signaling messengers and down-stream targets of NF-κB required for induction of IPC remain undefined. In a previous study, we demonstrated that epsilon protein kinase c (εPKC) was a key mediator of IPC in brain. Activation of εPKC induced cyclooygenase-2 (COX-2) expression and conferred ischemic tolerance in the neuronal and hippocampal slice models. Here, we hypothesized that IPC-mediated COX-2 expression was mediated by NF-κB. We tested this hypothesis in mixed cortical neuron/astrocyte cell cultures. To simulate IPC or ischemia, cell cultures were exposed to 1 or 4 h of oxygen-glucose deprivation, respectively. Our results demonstrated translocation of p65 and p50 subunits of NF-κB into nucleus following IPC or εPKC activation. NF-κB inhibition with pyrrolidine dithiocarbamate (10 μM) abolished IPC or εPKC activator-mediated neuroprotection indicating that NF-κB activation was involved in ischemic tolerance. In parallel studies, inhibition of either εPKC or the extracellular signal-regulated kinase (ERK 1/2) pathway reduced IPC-induced NF-κB activation. Finally, inhibition of NF-κB blocked IPC-induced COX-2 expression. In conclusion, we demonstrated that IPC-signaling cascade comprises εPKC activation→ERK1/2 activation→NF-κB translocation to nucleus→COX-2 expression resulting in neuroprotection in mixed neuronal culture.
Collapse
Affiliation(s)
- Eun Joo Kim
- Cerebral Vascular Disease Research Center, Department of Neurology and Neuroscience Program (D4-5), University of Miami Miller School of Medicine, P.O. Box 016960, Miami, FL 33101, USA
| | - Ami P. Raval
- Cerebral Vascular Disease Research Center, Department of Neurology and Neuroscience Program (D4-5), University of Miami Miller School of Medicine, P.O. Box 016960, Miami, FL 33101, USA
| | - Nina Hirsch
- Cerebral Vascular Disease Research Center, Department of Neurology and Neuroscience Program (D4-5), University of Miami Miller School of Medicine, P.O. Box 016960, Miami, FL 33101, USA
| | - Miguel A. Perez-Pinzon
- Cerebral Vascular Disease Research Center, Department of Neurology and Neuroscience Program (D4-5), University of Miami Miller School of Medicine, P.O. Box 016960, Miami, FL 33101, USA
| |
Collapse
|
59
|
Lee SM, Jang YS, Lee CT, Kim YW, Han SK, Shim YS, Yoo CG. Role of Insulin in the Activation of NF-κB/IκB Pathway in Macrophage Cells. Tuberc Respir Dis (Seoul) 2010. [DOI: 10.4046/trd.2010.68.3.168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Sang-Min Lee
- Department of Internal Medicine and Lung Institute of Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Yeon Sil Jang
- Department of Internal Medicine and Lung Institute of Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Choon-Taek Lee
- Respiratory Center, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Young Whan Kim
- Department of Internal Medicine and Lung Institute of Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Koo Han
- Department of Internal Medicine and Lung Institute of Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Young-Soo Shim
- Department of Internal Medicine and Lung Institute of Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Chul-Gyu Yoo
- Department of Internal Medicine and Lung Institute of Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
60
|
Grimaldo S, Tian F, Li LY. Sensitization of endothelial cells to VEGI-induced apoptosis by inhibiting the NF-kappaB pathway. Apoptosis 2009; 14:788-95. [PMID: 19418226 DOI: 10.1007/s10495-009-0351-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Vascular endothelial growth inhibitor (VEGI) is an endogenous inhibitor of endothelial cell growth and a promising candidate for cancer therapy. VEGI is able to inhibit tumor growth by specifically targeting the tumor neovasculature. Increasing the anti-angiogenic potential of this cytokine is of great interest for its therapeutic potential. NF-kappaB is known to have an integral role in TNF superfamily signaling, acting as a pro-survival factor. A role of VEGI-induced NF-kappaB activation in endothelial cells has yet to be described. Here we show that suppression of the NF-kappaB pathway can increase the apoptotic potential of VEGI. We used siRNA to deplete NF-kappaB or its activator IKK2 from adult bovine aortic endothelial cells. The siRNA treatments diminished VEGI-induced NF-kappaB activation, evidenced from a reduced extent of NF-kappaB nuclear translocation and diminished expression of NF-kappaB-target genes such as interleukins-6 and -1beta. The siRNA-treated endothelial cells when exposed to VEGI exhibited a marked decrease in cell viability and a significant increase in apoptosis. These results confirm that VEGI utilizes NF-kappaB as a pro-survival role factor in endothelial cells. We then examined whether a combination of VEGI with NF-kappaB inhibitors would constitute a more potential therapeutic regiment. We found that in the presence of the NF-kappaB inhibitors curcumin or BMS-345541 there was a marked increase in the apoptotic potential of VEGI on endothelial cells. These findings indicate that a combination therapy using VEGI and NF-kappaB inhibitors could be a potent approach for cancer treatment.
Collapse
Affiliation(s)
- Sammy Grimaldo
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | |
Collapse
|
61
|
Ban JO, Oh JH, Hwang BY, Moon DC, Jeong HS, Lee S, Kim S, Lee H, Kim KB, Han SB, Hong JT. Inflexinol inhibits colon cancer cell growth through inhibition of nuclear factor-kappaB activity via direct interaction with p50. Mol Cancer Ther 2009; 8:1613-24. [PMID: 19509257 DOI: 10.1158/1535-7163.mct-08-0694] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Kaurane diterpene compounds have been known to be cytotoxic against several cancer cells through inhibition of nuclear factor-kappaB (NF-kappaB) activity. Here, we showed that inflexinol, a novel kaurane diterpene compound, inhibited the activity of NF-kappaB and its target gene expression as well as cancer cell growth through induction of apoptotic cell death in vitro and in vivo. These inhibitory effects on NF-kappaB activity and on cancer cell growth were suppressed by the reducing agents DTT and glutathione and were abrogated in the cells transfected with mutant p50 (C62S). Sol-gel biochip and surface plasmon resonance analysis showed that inflexinol binds to the p50 subunit of NF-kappaB. These results suggest that inflexinol inhibits colon cancer cell growth via induction of apoptotic cell death through inactivation of NF-kappaB by a direct modification of cysteine residue in the p50 subunit of NF-kappaB.
Collapse
Affiliation(s)
- Jung Ok Ban
- College of Pharmacy, Chungbuk National University, 48 Gaeshin-dong, Heungduk-gu, Cheonju, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Cloning and characterization of DULP, a novel ubiquitin-like molecule from human dendritic cells. Cell Mol Immunol 2009; 6:27-33. [PMID: 19254477 DOI: 10.1038/cmi.2009.4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
We identified a novel ubiquitin-like molecule DULP from human dendritic cells. DULP contains a domain that shares 26% identity and 34% similarity with ubiquitin, and it possesses the corresponding Ile-44 hydrophobic patch used by mono- or poly-ubiquitin to interact with a ubiquitin-interaction motif (UIM) or ubiquitin-associated domain (UBA). Lysine residue corresponding to 6 of ubiquitin, which is involved in the formation of a multi-ubiquitin chain that can bind proteasomal subunit Rpn10/S5a, is also conserved in its ubiquitin-homology domain. However, DULP does not possess the highly conserved C-terminus Gly-Gly required for ubiquitin conjugation or the Lys-48 required for the formation of polyubiquitin chain to target substrates for degradation, suggesting it might be a novel ubiquitin-domain protein (UDP). DULP was found widely expressed in many cells and the ubiquitin-homology domain was not cleaved. We also confirmed that DULP expression was enriched in the nucleus and much weaker in the cytosol. Besides, we found that overexpression of DULP in 293T cells induced apoptosis, which might not be associated with the mitochondrial or proteasome pathway, with the specific mechanism remaining unclear. Further investigations are needed to identify the precise biological functions of DULP.
Collapse
|
63
|
Burnett JC, Miller-Jensen K, Shah PS, Arkin AP, Schaffer DV. Control of stochastic gene expression by host factors at the HIV promoter. PLoS Pathog 2009; 5:e1000260. [PMID: 19132086 PMCID: PMC2607019 DOI: 10.1371/journal.ppat.1000260] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Accepted: 12/09/2008] [Indexed: 12/21/2022] Open
Abstract
The HIV promoter within the viral long terminal repeat (LTR) orchestrates many aspects of the viral life cycle, from the dynamics of viral gene expression and replication to the establishment of a latent state. In particular, after viral integration into the host genome, stochastic fluctuations in viral gene expression amplified by the Tat positive feedback loop can contribute to the formation of either a productive, transactivated state or an inactive state. In a significant fraction of cells harboring an integrated copy of the HIV-1 model provirus (LTR-GFP-IRES-Tat), this bimodal gene expression profile is dynamic, as cells spontaneously and continuously flip between active (Bright) and inactive (Off) expression modes. Furthermore, these switching dynamics may contribute to the establishment and maintenance of proviral latency, because after viral integration long delays in gene expression can occur before viral transactivation. The HIV-1 promoter contains cis-acting Sp1 and NF-κB elements that regulate gene expression via the recruitment of both activating and repressing complexes. We hypothesized that interplay in the recruitment of such positive and negative factors could modulate the stability of the Bright and Off modes and thereby alter the sensitivity of viral gene expression to stochastic fluctuations in the Tat feedback loop. Using model lentivirus variants with mutations introduced in the Sp1 and NF-κB elements, we employed flow cytometry, mRNA quantification, pharmacological perturbations, and chromatin immunoprecipitation to reveal significant functional differences in contributions of each site to viral gene regulation. Specifically, the Sp1 sites apparently stabilize both the Bright and the Off states, such that their mutation promotes noisy gene expression and reduction in the regulation of histone acetylation and deacetylation. Furthermore, the NF-κB sites exhibit distinct properties, with κB site I serving a stronger activating role than κB site II. Moreover, Sp1 site III plays a particularly important role in the recruitment of both p300 and RelA to the promoter. Finally, analysis of 362 clonal cell populations infected with the viral variants revealed that mutations in any of the Sp1 sites yield a 6-fold higher frequency of clonal bifurcation compared to that of the wild-type promoter. Thus, each Sp1 and NF-κB site differentially contributes to the regulation of viral gene expression, and Sp1 sites functionally “dampen” transcriptional noise and thereby modulate the frequency and maintenance of this model of viral latency. These results may have biomedical implications for the treatment of HIV latency. After HIV genome integration into the host chromosome, the viral promoter coordinates a complex set of inputs to control the establishment of viral latency, the onset of viral gene expression, and the ensuing gene expression levels. Among these inputs are chromatin structure at the site of integration, host transcription factors, and the virally encoded transcriptional regulator Tat. Importantly, transcriptional noise from host and viral transcriptional regulators may play a critical role in the decision between replication versus latency, because stochastic fluctuations in gene expression are amplified by a Tat-mediated positive transcriptional feedback loop. To evaluate the individual contributions of key transcription factor binding elements in gene expression dynamics, we employ model HIV viruses with mutations introduced into numerous promoter elements. Extensive analysis of gene expression dynamics and transcription factor recruitment to the viral promoter reveals that each site differentially contributes to viral gene expression and to the establishment of a low expression state that may contribute to viral latency. This systems-level approach elucidates the synergistic contributions of host and viral factors to the dynamics, magnitudes, and stochastic effects in viral gene expression, as well as provides insights into mechanisms that contribute to proviral latency.
Collapse
Affiliation(s)
- John C Burnett
- Department of Chemical Engineering and the Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California, United States of America
| | | | | | | | | |
Collapse
|
64
|
Murakami T, Harada H, Suico MA, Shuto T, Suzu S, Kai H, Okada S. Ephedrae herba, a component of Japanese herbal medicine Mao-to, efficiently activates the replication of latent human immunodeficiency virus type 1 (HIV-1) in a monocytic cell line. Biol Pharm Bull 2008; 31:2334-2337. [PMID: 19043222 DOI: 10.1248/bpb.31.2334] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The persistence of latent human immunodeficiency virus type 1 (HIV-1)-infected cellular reservoirs, despite prolonged treatment with highly active antiretroviral therapy (HAART), represents a major hurdle to virus eradication. In this study, we evaluated the effect of Japanese herbal medicine on the induction of HIV-1 replication in latently infected monocytic cell line, U1, in order to eradicate virus efficiently. We found that Mao-to was able to induce HIV-1 replication either alone or in combination with tumor necrosis factor-alpha (TNF-alpha). Among the four components of Mao-to, only Ephedrae herba had strong effects in inducing HIV-1 replication. Analysis by Western blotting revealed that Ephedrae herba induced the nuclear translocation of nuclear factor-kappa B (NF-kappaB). Reporter assay data also showed that Ephedrae herba and, slightly, Mao-to activated the NF-kappaB promoter, indicating that these herbal agents may induce HIV-1 replication through NF-kappaB activation. These findings suggest that Mao-to and its component, Ephedrea herba, may be good candidates to augment HAART by inducing the expression of latent HIV-1 with the ultimate goal of eliminating persistent viral reservoirs in individuals infected with HIV-1.
Collapse
Affiliation(s)
- Toru Murakami
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Global COE Cell Fate Regulation Research and Education Unit, Kumamoto University, Japan
| | | | | | | | | | | | | |
Collapse
|
65
|
Singh K, Sinha S, Malonia SK, Bist P, Tergaonkar V, Chattopadhyay S. Tumor suppressor SMAR1 represses IkappaBalpha expression and inhibits p65 transactivation through matrix attachment regions. J Biol Chem 2008; 284:1267-78. [PMID: 18981184 DOI: 10.1074/jbc.m801088200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Aberrant NF-kappaB activity promotes tumorigenesis. However, NF-kappaB also inhibits tumor growth where tumor suppressor pathways remain unaltered. Thus, its role in tumorigenesis depends upon the function of other cellular factors. Tumor suppressor SMAR1 down-modulated in high grade breast cancers is regulated by p53 and is reported to interact and stabilize p53. Because both SMAR1 and NF-kappaB are involved in tumorigenesis, we investigated the effect of SMAR1 upon NF-kappaB activity. We show that SMAR1 induction by doxorubicin or overexpression produces functional NF-kappaB complexes that are competent for binding to NF-kappaB consensus sequence. However, SMAR1 induced p65-p50 complex is phosphorylation- and transactivation-deficient. Induction of functional NF-kappaB complexes stems from down-regulation of IkappaBalpha transcription through direct binding of SMAR1 to the matrix attachment region site present in IkappaBalpha promoter and recruitment of corepressor complex. Real time PCR array for NF-kappaB target genes revealed that SMAR1 down-regulates a subset of NF-kappaB target genes that are involved in tumorigenesis. We also show that SMAR1 inhibits tumor necrosis factor alpha-induced induction of NF-kappaB suggesting that activation of NF-kappaB by SMAR1 is independent and different from classical pathway. Thus, for the first time we report that a tumor suppressor protein SMAR1 can modulate NF-kappaB transactivation and inhibit tumorigenesis by regulating NF-kappaB target genes.
Collapse
Affiliation(s)
- Kamini Singh
- National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune 411007, Maharastra, India
| | | | | | | | | | | |
Collapse
|
66
|
Li K, Ma Q, Shi L, Dang C, Hong Y, Wang Q, Li Y, Fan W, Zhang L, Cheng J. NS5ATP9 gene regulated by NF-kappaB signal pathway. Arch Biochem Biophys 2008; 479:15-9. [PMID: 18727915 DOI: 10.1016/j.abb.2008.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2008] [Revised: 07/29/2008] [Accepted: 08/01/2008] [Indexed: 10/21/2022]
Abstract
NS5ATP9 was previously identified as p15 PAF [proliferating cell nuclear antigen (PCNA)-associated factor] to bind with PCNA. We earlier identified the promoter region of NS5ATP9 and found NS5ATP9 is a NS5A up-regulation gene. However little is known about how it is regulated. To investigate the gene regulation of NS5ATP9, we screened NS5ATP9 promoter binding proteins using phage display and verified by electrophoretic mobility shift assay (EMSA). We found that the nuclear protein rhNF-kappaB (p50) could bind to the NS5ATP9 promoter and the binding region contained within a 156 bp (nucleotides -5 to -161bp) immediately upstream of the transcription initiation site. Our results suggest that NF-kappaB could participate in the regulation of NS5ATP9 gene expression in carcinogenesis.
Collapse
Affiliation(s)
- Kang Li
- The First Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Natoli G, Chiocca S. Nuclear Ubiquitin Ligases, NF- B Degradation, and the Control of Inflammation. Sci Signal 2008; 1:pe1. [DOI: 10.1126/stke.11pe1] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
68
|
Thompson C, Cloutier A, Bossé Y, Poisson C, Larivée P, McDonald PP, Stankova J, Rola-Pleszczynski M. Signaling by the Cysteinyl-Leukotriene Receptor 2. J Biol Chem 2008; 283:1974-84. [DOI: 10.1074/jbc.m608197200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
69
|
DeLotto R, DeLotto Y, Steward R, Lippincott-Schwartz J. Nucleocytoplasmic shuttling mediates the dynamic maintenance of nuclear Dorsal levels during Drosophila embryogenesis. Development 2007; 134:4233-41. [PMID: 17978003 DOI: 10.1242/dev.010934] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
In Drosophila, the NF-kappaB/REL family transcription factor, Dorsal, redistributes from the cytoplasm to nuclei, forming a concentration gradient across the dorsoventral axis of the embryo. Using live imaging techniques in conjunction with embryos expressing a chimeric Dorsal-GFP, we demonstrate that the redistribution of Dorsal from cytoplasm to nucleus is an extremely dynamic process. Nuclear Dorsal concentration changes continuously over time in all nuclei during interphase. While Dorsal appears to be nuclearly localized primarily in ventral nuclei, it is actively shuttling into and out of all nuclei, including nuclei on the dorsal side. Nuclear export is blocked by leptomycin B, a potent inhibitor of Exportin 1 (CRM1)-mediated nuclear export. We have developed a novel in vivo assay revealing the presence of a functional leucine-rich nuclear export signal within the carboxyterminal 44 amino acids of Dorsal. We also find that diffusion of Dorsal is partially constrained to cytoplasmic islands surrounding individual syncitial nuclei. A model is proposed in which the generation and maintenance of the Dorsal gradient is a consequence of an active process involving both restricted long-range diffusion and the balancing of nuclear import with nuclear export.
Collapse
Affiliation(s)
- Robert DeLotto
- Department of Molecular Biology, University of Copenhagen, Ole Maaløes Vej 5, DK-2200 Copenhagen N, Denmark.
| | | | | | | |
Collapse
|
70
|
Abstract
Over the past few years, the transcription factor nuclear factor (NF)-kappaB and the proteins that regulate it have emerged as a signaling system of pre-eminent importance in human physiology and in an increasing number of pathologies. While NF-kappaB is present in all differentiated cell types, its discovery and early characterization were rooted in understanding B-cell biology. Significant research efforts over two decades have yielded a large body of literature devoted to understanding NF-kappaB's functioning in the immune system. NF-kappaB has been found to play roles in many different compartments of the immune system during differentiation of immune cells and development of lymphoid organs and during immune activation. NF-kappaB is the nuclear effector of signaling pathways emanating from many receptors, including those of the inflammatory tumor necrosis factor and Toll-like receptor superfamilies. With this review, we hope to provide historical context and summarize the diverse physiological functions of NF-kappaB in the immune system before focusing on recent advances in elucidating the molecular mechanisms that mediate cell type-specific and stimulus-specific functions of this pleiotropic signaling system. Understanding the genetic regulatory circuitry of NF-kappaB functionalities involves system-wide measurements, biophysical studies, and computational modeling.
Collapse
Affiliation(s)
- Alexander Hoffmann
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA.
| | | |
Collapse
|
71
|
Raymond L, Eck S, Hays E, Tomek I, Kantor S, Vincenti M. RelA is required for IL-1beta stimulation of Matrix Metalloproteinase-1 expression in chondrocytes. Osteoarthritis Cartilage 2007; 15:431-41. [PMID: 17097317 PMCID: PMC1865522 DOI: 10.1016/j.joca.2006.09.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2006] [Accepted: 09/24/2006] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Interleukin-1beta (IL-1beta) stimulates collagenase-1 (Matrix Metalloproteinase-1 (MMP-1)) expression in articular chondrocytes, leading to cleavage of type II collagen and irreversible cartilage degradation. The nuclear factor-kappa B (NF-kappaB) pathway is potently activated in IL-1beta-stimulated cells and has been implicated as an intermediate in MMP-1 gene expression. However, the roles of individual NF-kappaB family members during IL-1beta-induced MMP-1 gene expression have not been defined. RESULTS To address the relationship between the NF-kappaB pathway and MMP-1 gene activation in chondrocytes, primary cultured human articular chondrocyte cultures (HAC) and SW-1353 cells were stimulated with IL-1beta over a 24-h time course and MMP-1, NF-kappaB1, NF-kappaB2 and RelA gene expression was assayed. IL-1beta-induced MMP-1 expression was comparable in HAC and SW-1353 cells both temporally and quantitatively. MMP-1 gene expression was mirrored by increases in NF-kappaB gene expression, and inhibition of NF-kappaB nuclear translocation with dominant-negative IkappaBalpha reduced IL-1beta-dependent MMP-1 gene expression. IL-1beta activated the NF-kappaB pathway in chondrocytes, both through phosphorylation and transient degradation of IkappaBalpha, as well as through sustained phosphorylation of RelA. Small inhibitory RNAs (siRNA) specific for RelA resulted in significant reduction of MMP-1 mRNA, whereas siRNA for NF-kappaB1 and NF-kappaB2 augmented IL-1beta-induced MMP-1 expression. CONCLUSIONS Our data demonstrate that IL-1beta activation of the NF-kappaB pathway is required for IL-1beta induction of MMP-1 in chondrocytes and that RelA can work independently of NF-kappaB1 or NF-kappaB2 to activate this gene expression program.
Collapse
Affiliation(s)
- L Raymond
- Research Service, VA Medical Center, White River Junction, VT 05009, and Department of Medicine, Dartmouth Medical School, Lebanon, NH, USA
| | | | | | | | | | | |
Collapse
|
72
|
Abstract
NF-kappaB transcription factors and the signaling pathways that activate them play a critical role in cancer development, progression and therapy, and recently have become a focal point for intense drug discovery and development efforts. This article presents a critical review on the different types of inhibitors targeting the NF-kappaB pathway at several stages.
Collapse
Affiliation(s)
- Davide Melisi
- The University of Texas, MD Anderson Cancer Center, Department of Gastrointestinal Medical Oncology, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | |
Collapse
|
73
|
Majdalawieh A, Zhang L, Ro HS. Adipocyte enhancer-binding protein-1 promotes macrophage inflammatory responsiveness by up-regulating NF-kappaB via IkappaBalpha negative regulation. Mol Biol Cell 2007; 18:930-42. [PMID: 17202411 PMCID: PMC1805081 DOI: 10.1091/mbc.e06-03-0217] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2006] [Revised: 12/18/2006] [Accepted: 12/22/2006] [Indexed: 11/11/2022] Open
Abstract
Nuclear factor kappaB (NF-kappaB) subunits comprise a family of eukaryotic transcription factors that are critically involved in cell proliferation, inflammation, and apoptosis. Under basal conditions, NF-kappaB subunits are kept under inhibitory regulation by physical interaction with NF-kappaB inhibitors (IkappaB subunits) in the cytosol. Upon stimulation, IkappaB subunits become phosphorylated, ubiquitinated, and subsequently degraded, allowing NF-kappaB subunits to translocate to the nucleus and bind as dimers to kappaB responsive elements of target genes. Previously, we have shown that AEBP1 enhances macrophage inflammatory responsiveness by inducing the expression of various proinflammatory mediators. Herein, we provide evidence suggesting that AEBP1 manifests its proinflammatory function by up-regulating NF-kappaB activity via hampering IkappaBalpha, but not IkappaBbeta, inhibitory function through protein-protein interaction mediated by the discoidin-like domain (DLD) of AEBP1. Such interaction renders IkappaBalpha susceptible to enhanced phosphorylation and degradation, subsequently leading to augmented NF-kappaB activity. Collectively, we propose a novel molecular mechanism whereby NF-kappaB activity is modulated by means of protein-protein interaction involving AEBP1 and IkappaBalpha. Moreover, our study provides a plausible mechanism explaining the differential regulatory functions exhibited by IkappaBalpha and IkappaBbeta in various cell types. We speculate that AEBP1 may serve as a potential therapeutic target for the treatment of various chronic inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Amin Majdalawieh
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada B3H 1X5
| | - Lei Zhang
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada B3H 1X5
| | - Hyo-Sung Ro
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada B3H 1X5
| |
Collapse
|
74
|
Cohen-Lahav M, Douvdevani A, Chaimovitz C, Shany S. The anti-inflammatory activity of 1,25-dihydroxyvitamin D3 in macrophages. J Steroid Biochem Mol Biol 2007; 103:558-62. [PMID: 17267205 DOI: 10.1016/j.jsbmb.2006.12.093] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In a previous study we demonstrated a down-regulatory effect of vitamin D active metabolite (1,25(OH)(2)D(3)) and its vitamin D(2) analog (1,24(OH)(2)D(2)) on TNFalpha expression in macrophages. We also found an inhibitory effect in the physiological concentration (10(-10)M) of 1,25(OH)(2)D(3) which was dose-dependent. This down-regulation, caused by the decrease in NFkappaB activity by 1,25(OH)(2)D(3) and 1,24(OH)(2)D(2), was demonstrated in P388D1 cells transfected with NFkappaB reporter gene (p NFkappaB-Luc) and by EMSA. In our present study we investigated the processes leading to reduced NFkappaB activity on P388D1 cells. A decrease in nuclei NFkappaB-p65 and an increase in cytosolic NFkappaB-p65, were measured, while no changes in total NFkappaB-p65 mRNA and protein levels were observed. Simultaneously, a significant increase in both mRNA and protein levels of the NFkappaB-cytosolic inhibitor, IkappaBalpha, were determined. The half-life of IkappaBalpha-mRNA increased, with a parallel decrease in the phosphorylation of its protein, as the first step of ubiquitinization and degradation. The present results demonstrate that 1,25(OH)(2)D(3) and 1,24(OH)(2)D(2) inhibit TNFalpha expression in macrophages, by increasing IkappaBalpha and decreasing NFkappaB activity. Since NFkappaB is a major transcription factor for TNFalpha and other inflammatory mediators, these findings suggest that 1,25(OH)(2)D(3) and 1,24(OH)(2)D(2) may be used therapeutically as anti-inflammatory agents.
Collapse
Affiliation(s)
- Merav Cohen-Lahav
- Department of Clinical Biochemistry, Soroka Medical Center, Ben-Gurion University, Faculty of Health Sciences, P.O. Box 151, Beer Sheva 84101, Israel
| | | | | | | |
Collapse
|
75
|
Hansberger MW, Campbell JA, Danthi P, Arrate P, Pennington KN, Marcu KB, Ballard DW, Dermody TS. IkappaB kinase subunits alpha and gamma are required for activation of NF-kappaB and induction of apoptosis by mammalian reovirus. J Virol 2006; 81:1360-71. [PMID: 17121808 PMCID: PMC1797491 DOI: 10.1128/jvi.01860-06] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Reoviruses induce apoptosis both in cultured cells and in vivo. Apoptosis plays a major role in the pathogenesis of reovirus encephalitis and myocarditis in infected mice. Reovirus-induced apoptosis is dependent on the activation of transcription factor NF-kappaB and downstream cellular genes. To better understand the mechanism of NF-kappaB activation by reovirus, NF-kappaB signaling intermediates under reovirus control were investigated at the level of Rel, IkappaB, and IkappaB kinase (IKK) proteins. We found that reovirus infection leads initially to nuclear translocation of p50 and RelA, followed by delayed mobilization of c-Rel and p52. This biphasic pattern of Rel protein activation is associated with the degradation of the NF-kappaB inhibitor IkappaBalpha but not the structurally related inhibitors IkappaBbeta or IkappaBepsilon. Using IKK subunit-specific small interfering RNAs and cells deficient in individual IKK subunits, we demonstrate that IKKalpha but not IKKbeta is required for reovirus-induced NF-kappaB activation and apoptosis. Despite the preferential usage of IKKalpha, both NF-kappaB activation and apoptosis were attenuated in cells lacking IKKgamma/Nemo, an essential regulatory subunit of IKKbeta. Moreover, deletion of the gene encoding NF-kappaB-inducing kinase, which is known to modulate IKKalpha function, had no inhibitory effect on either response in reovirus-infected cells. Collectively, these findings indicate a novel pathway of NF-kappaB/Rel activation involving IKKalpha and IKKgamma/Nemo, which together mediate the expression of downstream proapoptotic genes in reovirus-infected cells.
Collapse
Affiliation(s)
- Mark W Hansberger
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Song JJ, An JY, Kwon YT, Lee YJ. Evidence for two modes of development of acquired tumor necrosis factor-related apoptosis-inducing ligand resistance. Involvement of Bcl-xL. J Biol Chem 2006; 282:319-28. [PMID: 17110373 DOI: 10.1074/jbc.m608065200] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Previous studies have shown that repeated application of TRAIL induces acquired resistance to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Using human prostate adenocarcinoma DU-145 and human pancreatic carcinoma MiaPaCa-2 cells as a model, we now demonstrate for the first time that two states of acquired TRAIL resistance can be developed after TRAIL treatment. Data from survival assay and Western blot analysis show that acquired TRAIL resistance was developed within 1 day and gradually decayed within 6 days after TRAIL treatment in both cell lines. After TRAIL treatment, the level of Bcl-xL increased and reached a maximum within 2 days and gradually decreased in both cell lines. Bcl-xL-mediated development of acquired TRAIL resistance was suppressed by knockdown of Bcl-xL expression. Protein interaction assay revealed that during the development of TRAIL resistance, Bcl-xL dissociated from Bad and then associated with Bax. Overexpression of mutant-type Bad (S136A), which prevents this dissociation, partially suppressed the development of acquired TRAIL resistance. Thus, our results suggest that (a) dissociation of Bad from Bcl-xL and (b) an increase in the intracellular level of Bcl-xL are responsible for development of acquired TRAIL resistance.
Collapse
Affiliation(s)
- Jae J Song
- Department of Surgery and Pharmacology, School of Medicine, University of Pittsburgh, Pennsylvania 15213, USA
| | | | | | | |
Collapse
|
77
|
Park SS, Kim JM, Kim DS, Kim IH, Kim SY. Transglutaminase 2 Mediates Polymer Formation of I-κBα through C-terminal Glutamine Cluster. J Biol Chem 2006; 281:34965-72. [PMID: 16987813 DOI: 10.1074/jbc.m604150200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Recently we reported that transglutaminase 2 (TGase 2) activates nuclear factor-kappaB (NF-kappaB) independently of I-kappaB kinase (IKK) activation, by inducing cross-linking and protein polymer formation of inhibitor of nuclear factor-kappaBalpha (I-kappaBalpha). TGase 2 catalyzes covalent isopeptide bond formation between the peptide bound-glutamine and the lysine residues. Using matrix-assisted laser desorption ionization time-of-flight mass spectra analysis of I-kappaBalpha polymers cross-linked by TGase 2, as well as synthetic peptides in an in vitro competition assay, we identified a glutamine cluster at the C terminus of I-kappaBalpha (amino acids 266-268) that appeared to play a key role in the formation of I-kappaBalpha polymers. Although there appeared to be no requirement for specific lysine residues, we found a considerably higher preference for the use of lysine residues at positions 21, 22, and 177 in TGase 2-mediated cross-linking of I-kappaBalpha. We demonstrated that synthetic peptides encompassing the glutamine cluster at amino acid positions 266-268 reversed I-kappaBalpha polymerization in vitro. Furthermore, the depletion of free I-kappaBalpha in EcR/TG cells was completely rescued in vivo by transfection of mutant I-kappaBalphas in glutamine sites (Q266G, Q267G, and Q313G) as well as in a lysine site (K177G). These findings provide additional clues into the mechanism by which TGase 2 contributes to the inflammatory process via activation of NF-kappaB.
Collapse
Affiliation(s)
- Sung-Soo Park
- Molecular Oncology Branch, Division of Basic Sciences, Research Institute, National Cancer Center, 809 Madu-1-dong, Ilsandong-gu, Goyang, Gyeonggi-do 411-769, Republic of Korea
| | | | | | | | | |
Collapse
|
78
|
|
79
|
Tsuchiya K, Yoshimoto T, Hirono Y, Tateno T, Sugiyama T, Hirata Y. Angiotensin II induces monocyte chemoattractant protein-1 expression via a nuclear factor-kappaB-dependent pathway in rat preadipocytes. Am J Physiol Endocrinol Metab 2006; 291:E771-8. [PMID: 16705055 DOI: 10.1152/ajpendo.00560.2005] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Both monocyte chemoattractant protein-1 (MCP-1), a member of chemokine family, and angiotensinogen, a precursor of angiotensin (ANG) II, are produced by adipose tissue and increased in obese state. MCP-1 has been shown to decrease insulin-stimulated glucose uptake and several adipogenic genes expression in adipocytes in vitro, suggesting its pathophysiological significance in obesity. However, the pathophysiological interaction between MCP-1 and ANG II in adipose tissue remains unknown. The present study was undertaken to investigate the potential mechanisms by which ANG II affects MCP-1 gene expression in rat primary cultured preadipocytes and adipose tissue in vivo. ANG II significantly increased steady-state MCP-1 mRNA levels in a time- and dose-dependent manner. The ANG II-induced MCP-1 mRNA and protein expression was completely abolished by ANG II type 1 (AT1)-receptor antagonist (valsartan). An antioxidant/NF-kappaB inhibitor (pyrrolidine dithiocarbamate) and an inhibitor of 1kappaB-alpha phosphorylation (Bay 11-7085) also blocked ANG II-induced MCP-1 mRNA expression. ANG II induced translocation of NF-kappaB p65 subunit from cytoplasm to nucleus by immunocytochemical study. Luciferase assay using reporter constructs containing MCP-1 promoter region revealed that two NF-kappaB binding sites in its enhancer region were essential for the ANG II-induced promoter activities. Furthermore, basal mRNA and protein of MCP-1 during preadipocyte differentiation were significantly greater in preadipocytes than in differentiated adipocytes, whose effect was more pronounced in the presence of ANG II. Exogenous administration of ANG II to rats led to increased MCP-1 expression in epididymal, subcutaneous, and mesenteric adipose tissue. In conclusion, our present study demonstrates that ANG II increases MCP-1 gene expression via ANG II type 1 receptor-mediated and NF-kappaB-dependent pathway in rat preadipocytes as well as adipose MCP-1 expression in vivo. Thus the augmented MCP-1 expression by ANG II in preadipocytes may provide a new link between obesity and cardiovascular disease.
Collapse
Affiliation(s)
- Kyoichiro Tsuchiya
- Department of Clinical and Molecular Endocrinology, Tokyo Medical and Dental University Graduate School, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8513, Japan
| | | | | | | | | | | |
Collapse
|
80
|
Thompson C, Cloutier A, Bossé Y, Thivierge M, Gouill CL, Larivée P, McDonald PP, Stankova J, Rola-Pleszczynski M. CysLT1 receptor engagement induces activator protein-1- and NF-kappaB-dependent IL-8 expression. Am J Respir Cell Mol Biol 2006; 35:697-704. [PMID: 16809637 DOI: 10.1165/rcmb.2005-0407oc] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Because cysteinyl-leukotrienes (cysLTs) are major protagonists in the pathophysiology of human asthma, and because neutrophils are involved in the more severe form of asthma, we studied the potential for leukotriene (LT) D(4) to induce synthesis of the chemokine IL-8 through activation of the CysLT1 receptor. We found LTD(4) to induce IL-8 gene expression in monocytic THP-1 cells and human dendritic cells with complete abrogation by selective CysLT1 antagonists. Human embryonic kidney-293 cells stably transfected with CysLT1 were used to better study the transcriptional regulation of the IL-8 promoter. Stimulation of the cells with graded concentrations of LTD(4) resulted in a time- and concentration-dependent induction of IL-8 transcription and protein synthesis. Use of IL-8 promoter mutants with substitutions in their NF-kappaB, activator protein (AP)-1, and NF-IL-6 binding elements revealed a requirement for NF-kappaB and AP-1, but not NF-IL-6, in LTD(4)-induced activation of the IL-8 promoter. Overexpression of dominant-negative IkappaBalpha inhibited the IL-8 transactivation induced by LTD(4). NF-kappaB DNA binding activity was induced by LTD(4), as determined by electrophoretic mobility shift assays, and could be supershifted by antibodies against p50 and p65. Supershift assays after LTD(4) stimulation also indicated the formation of a c-Jun/c-Fos complex. Moreover, our results demonstrate that LTD(4) upregulates the expression of c-fos and c-jun at the mRNA level. Our data show for the first time that LTD(4), via the CysLT1 receptor, can transcriptionally activate IL-8 production, with involvement of the transcription factors p50, p65, Fos, and Jun. These findings provide mechanistic and potentially therapeutic elements for modulation of the inflammatory component of asthma.
Collapse
Affiliation(s)
- Charles Thompson
- Immunology Division, Department of Pediatrics, Faculty of Medicine, Université de Sherbrooke, 3001 North 12th Avenue, Sherbrooke, PQ, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Shalini S, Bansal MP. Alterations in selenium status influences reproductive potential of male mice by modulation of transcription factor NFkappaB. Biometals 2006; 20:49-59. [PMID: 16758115 DOI: 10.1007/s10534-006-9014-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2005] [Accepted: 04/11/2006] [Indexed: 01/12/2023]
Abstract
Selenium (Se), an essential dietary trace element, is required for the maintenance of male fertility. In order to study its role in spermatogenesis, Balb/c mice with different Se status (Se deficient, group I; adequate, group II and excess, group III) were generated by feeding yeast based Se deficient diet for group I and deficient diet supplemented with Se as sodium selenite at adequate (0.2 ppm) and excess (1 ppm) for group II and III, respectively, for a period of 4 and 8 weeks. Percentage fertility was reduced in group I and III as compared to group II. A significant decrease in Se levels and glutathione peroxidase (GSH-Px) activity were observed in group I animals, whereas increase in GSH-Px activity was seen in group III. Further, significant increase in lipid peroxidation was observed in both Se deficient and excess groups. This indicated that dietary manipulation of Se levels either deficiency or excess leads to increased oxidative stress. Nuclear factor kappa B (NFkappaB), a well-known redox regulated transcription factor has also been suggested to play a crucial role in spermatogenesis. The expression of both p65 and p50 genes (components of NFkappaB) increased in Se deficient group I mice while the expression of the inhibitory IkappaBalpha declined significantly. This indicated activation of NFkappaB in Se deficiency. We also studied iNOS expression, which is a known target gene of NFkappaB, by RT-PCR. Significant elevation in the iNOS levels as well as NO levels was recorded. Both enhanced NO levels and NFkappaB are harmful in the progression of normal spermatogenic cycle. Therefore, present result clearly demonstrates the effect of reduced supply of Se on up-regulation and activation of NFkappaB in testis and its influence on spermatogenesis.
Collapse
Affiliation(s)
- Sonia Shalini
- Department of Biophysics, Panjab University, Chandigarh, 160 014, India
| | | |
Collapse
|
82
|
Lee JH, Jung HS, Giang PM, Jin X, Lee S, Son PT, Lee D, Hong YS, Lee K, Lee JJ. Blockade of nuclear factor-kappaB signaling pathway and anti-inflammatory activity of cardamomin, a chalcone analog from Alpinia conchigera. J Pharmacol Exp Ther 2006; 316:271-8. [PMID: 16183703 DOI: 10.1124/jpet.105.092486] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nuclear factor-kappaB (NF-kappaB) and the signaling pathways that regulate its activity have become a focal point for intense drug discovery and development efforts. NF-kappaB regulates the transcription of a large number of genes, particularly those involved in immune, inflammatory, and antiapoptotic responses. In our search for NF-kappaB inhibitors from natural resources, we identified cardamomin, 2',4'-dihydroxy-6'-methoxychalcone, as an inhibitor of NF-kappaB activation from Alpinia conchigera Griff (Zingiberaceae). In present study, we demonstrated the effect of cardamomin on NF-kappaB activation in lipopolysaccharide (LPS)-stimulated RAW264.7 cells and LPS-induced mortality. This compound significantly inhibited the induced expression of NF-kappaB reporter gene by LPS or tumor necrosis factor (TNF)-alpha in a dose-dependent manner. LPS-induced production of TNF-alpha and NO as well as expression of inducible nitric-oxide synthase and cyclooxygenase-2 was significantly suppressed by the treatment of cardamomin in RAW264.7 cells. Also, cardamomin inhibited not only LPS-induced degradation and phosphorylation of inhibitor kappaBalpha (IkappaBalpha) but also activation of inhibitor kappaB (IkappaB) kinases and nuclear translocation of NF-kappaB. Further analyses revealed that cardamomin did not directly inhibit IkappaB kinases, but it significantly suppressed LPS-induced activation of Akt. Moreover, cardamomin suppressed transcriptional activity and phosphorylation of Ser536 of RelA/p65 subunit of NF-kappaB. However, this compound did not inhibit LPS-induced activation of extracellular signal-regulated kinase and stress-activated protein kinase/c-Jun NH(2)-terminal kinase, but significantly impaired activation of p38 mitogen-activated protein kinase. We also demonstrated that pretreatment of cardamomin rescued C57BL/6 mice from LPS-induced mortality in conjunction with decreased serum level of TNF-alpha. Together, cardamomin could be valuable candidate for the intervention of NF-kappaB-dependent pathological condition such as inflammation.
Collapse
Affiliation(s)
- Jeong-Hyung Lee
- Anticancer Research Laboratory, Korea Research Institute of Bioscience and Biotechnology, P.O. Box 115, Yuseong, Daejeon 305-600, Korea.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Pfeffer U, Ferrari N, Dell'Eva R, Indraccolo S, Morini M, Noonan DM, Albini A. Molecular mechanisms of action of angiopreventive anti-oxidants on endothelial cells: microarray gene expression analyses. Mutat Res 2005; 591:198-211. [PMID: 16084531 DOI: 10.1016/j.mrfmmm.2005.04.014] [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: 11/16/2004] [Revised: 04/09/2005] [Accepted: 04/09/2005] [Indexed: 05/03/2023]
Abstract
The anti-oxidants N-acetyl-l-cysteine (NAC) and (-)-epigallocatechin-3-gallate (EGCG) inhibit tumor vascularization by reducing endothelial cell migration and invasion in a similar, non additive and non synergistic manner but do not alter the growth of human umbilical vein endothelial cells. Here we address the effects of the two chemopreventive drugs on endothelial cell signaling by means of expression profiling and real-time PCR validation. We identify a series of angiogenesis related genes that are similarly regulated by the two drugs. Anti-oxidant treated endothelial cells show gene expression profiles compatible with a less activated, less apoptosis prone and less migratory phenotype. The anti-oxidants affect expression of several components of the TNFalpha response pathway including downstream genes that are regulated in the opposite direction in the absence of the inflammatory cytokine. The interference with the TNFalpha pathway is reflected by reduced NFkappaB activation in anti-oxidants treated cells but the compounds are not able to contrast TNFalpha mediated activation of NFkappaB. The chemopreventive action of these compounds thus relies on a reduction of basal levels of endothelial cell activation. Down-regulation of the TNFalpha responsive pro-metastatic, pro-inflammatory genes, urokinase plasminogen activator and selectin E, further implies anti-metastatic effects for these drugs.
Collapse
Affiliation(s)
- Ulrich Pfeffer
- Functional Genomics Unit, National Cancer Research Institute, Genova, Italy
| | | | | | | | | | | | | |
Collapse
|
84
|
Wegrzyn P, Jura J, Kupiec T, Piekoszewski W, Władyka B, Zarebski A, Koj A. A search for genes modulated by interleukin-6 alone or with interleukin-1beta in HepG2 cells using differential display analysis. Biochim Biophys Acta Mol Basis Dis 2005; 1762:319-28. [PMID: 16338121 DOI: 10.1016/j.bbadis.2005.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2005] [Revised: 10/11/2005] [Accepted: 11/01/2005] [Indexed: 10/25/2022]
Abstract
Interleukin-1 and interleukin-6 are principal cytokines involved in regulation of expression of acute-phase proteins. In the joint action of both cytokines IL-1 can suppress or enhance the IL-6-dependent induction of gene expression. Here, we report changes in the transcriptome profile of HepG2 cells exposed to IL-6 alone, or IL-1 and IL-6. Cytokine-responsive genes were identified by differential display analysis. Validation of observed changes in the transcript level was carried out using the slot blot method. Out of 88 cDNA species modulated by IL-6, only 38 represent different known genes whereas 18 clones match genomic clones in NCBI data with hypothetical cDNA sequences (the remaining 32 clones showed no homology with the database or represented several clones of the same gene). In the experiments with HepG2 cells prestimulated for 3 h with IL-1 and then stimulated with IL-6, 43 cDNA fragments were amplified. Twenty-three of them represent known genes while 10 clones have inserts matching hypothetical cDNA sequences in NCBI data. The identified transcripts modulated by IL-6 or both cytokines in HepG2 cells code for intracellular proteins of various function. The largest groups represent genes engaged in metabolism, protein synthesis and signaling pathways. Among all genes identified as differentially regulated under stimulation by IL-6, or IL-1/IL-6, six were detected in both types of stimulation. None of the typical genes coding for plasma acute phase proteins was identified in our experiments. This indicates that differential display cannot be used to characterize the profile of a given transcriptome. On the other hand, it is a useful technique for detection of new genes responding to IL-6 alone or IL-6 in combination with IL-1.
Collapse
Affiliation(s)
- Paulina Wegrzyn
- Department of Cell Biochemistry, Faculty of Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | | | | | | | | | | | | |
Collapse
|
85
|
Marui N, Medford R, Ahmad M. Activation of RelA homodimers by tumour necrosis factor alpha: a possible transcriptional activator in human vascular endothelial cells. Biochem J 2005; 390:317-24. [PMID: 15876188 PMCID: PMC1184585 DOI: 10.1042/bj20041659] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In vascular endothelial cells, cytokines induce genes that are expressed in inflammatory lesions partly through the activation of transcription factor NF-kappaB (nuclear factor-kappaB). Among the members of the NF-kappaB/rel protein family, homodimers of the RelA subunit of NF-kappaB can also function as strong transactivators when expressed in cells. However, the functional role of endogenous RelA homodimers has not been clearly elucidated. We investigated whether RelA homodimers are induced in cytokine-treated vascular endothelial cells. Gel mobility-shift and supershift assays revealed that a cytokine TNFalpha (tumour necrosis factor alpha) activated both NF-kappaB1/RelA heterodimers and RelA homodimers that bound to a canonical kappaB sequence, IgkappaB (immunoglobulin kappaB), in SV40 (simian virus 40) immortalized HMEC-1 (human dermal microvascular endothelial cell line 1). In HMEC-1 and HUVEC (human umbilical-vein endothelial cells), TNFalpha also induced RelA homodimers that bound to the sequence 65-2kappaB, which specifically binds to RelA homodimers but not to NF-kappaB1/RelA heterodimers in vitro. Deoxycholic acid, a detergent that can dissociate the NF-kappaB-IkappaB complex (where IkappaB stands for inhibitory kappaB), induced the binding of the RelA homodimers to 65-2kappaB from the cytosolic fraction of resting HMEC-1. Furthermore, TNFalpha induced the transcriptional activity of a reporter gene that was driven by 65-2kappaB in HMEC-1. These results suggest that in addition to NF-kappaB1/RelA heterodimers, TNFalpha also induces RelA homodimers that are functionally active. Thus RelA homodimers may actively participate in cytokine regulation of gene expression in human vascular endothelial cells.
Collapse
Affiliation(s)
- Nobuyuki Marui
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA 30322, U.S.A
| | - Russell M. Medford
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA 30322, U.S.A
| | - Mushtaq Ahmad
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA 30322, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
86
|
Kim KM, Song JJ, An JY, Kwon YT, Lee YJ. Pretreatment of acetylsalicylic acid promotes tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis by down-regulating BCL-2 gene expression. J Biol Chem 2005; 280:41047-56. [PMID: 16199534 DOI: 10.1074/jbc.m503713200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been shown to be selective in the induction of apoptosis in cancer cells with minimal toxicity to normal tissues. However, not all cancers are sensitive to TRAIL-mediated apoptosis. Thus, TRAIL-resistant cancer cells must be sensitized first to become responsive to TRAIL. In this study, we observed that pretreatment by acetylsalicylic acid (ASA) augmented TRAIL-induced apoptotic death in human prostate adenocarcinoma LNCaP and human colorectal carcinoma CX-1 cells. Western blot analysis showed that pretreatment of ASA followed by TRAIL treatment activated caspases (8, 9, and 3) and cleaved poly(ADP-ribose) polymerase, the hallmark feature of apoptosis. Most interestingly, at least 12 h of pretreatment with ASA was prerequisite for promoting TRAIL-induced apoptosis and was related to down-regulation of BCL-2. Biochemical analysis revealed that ASA inhibited NF-kappaB activity, which is known to regulate BCL-2 gene expression, by dephosphorylating IkappaB-alpha and inhibiting IKKbeta activity but not by affecting the HER-2/neu phosphatidylinositol 3-kinase-Akt signal pathway. Overexpression of BCL-2 suppressed the promotive effect of ASA on TRAIL-induced apoptosis and changes in mitochondrial membrane potential. Taken together, our studies suggested that ASA-promoted TRAIL cytotoxicity is mediated through down-regulating BCL-2 and by decreasing mitochondrial membrane potential.
Collapse
Affiliation(s)
- Ki M Kim
- Department of Surgery and Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | |
Collapse
|
87
|
Su G, Min W, Taylor EW. An HIV-1 encoded peptide mimics the DNA binding loop of NF-kappaB and binds thioredoxin with high affinity. Mutat Res 2005; 579:133-48. [PMID: 16054658 DOI: 10.1016/j.mrfmmm.2005.02.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2004] [Revised: 02/16/2005] [Accepted: 02/16/2005] [Indexed: 10/25/2022]
Abstract
Pro-fs is a human immunodeficiency virus type 1 (HIV-l)-encoded putative selenoprotein, predicted by a theoretical analysis of the viral genome; it is potentially expressed by a -1 frameshift from the protease coding region. Pro-fs has significant sequence similarity to the DNA binding loop of nuclear factor kappa B (NF-kappaB), which is known to bind thioredoxin (Trx). We hypothesize that the putative HIV-1 pro-fs gene product functions by mimicry of NF-kappaB via binding to Trx. The hypothesis was tested in vitro by co-immunoprecipitation and GST-pull down assays, using a purified mutant pro-fs protein, in which the two potential selenocysteine residues were mutated to cysteines, in order to permit expression in bacteria. Both experiments showed that pro-fs binds to human wild type Trx (Trx-wt) with high affinity. Mutation of the two conserved cysteine residues in the Trx active site redox center to serine (Ser) (Trx-CS) weakened but failed to abolish the interaction. In pro-fs-transfected 293T cells, using confocal microscopy and fluorescence resonance energy transfer (FRET), we have observed that pro-fs localizes in cell nuclei and forms oligomers. Upon stimulation by phorbol 12-myristate 13-acetate (PMA), Trx translocates into cell nuclei. Significant FRET efficiency was detected in the nuclei of PMA-stimulated 293T cells co-expressing fluorescence-tagged pro-fs and Trx-wt or Trx-CS. These results indicate that in living cells the double cysteine mutant of pro-fs binds to both Trx and Trx-CS with high affinity, suggesting that Trx-pro-fs binding is a structurally-specific interaction, involving more of the Trx molecule than just its active site cysteine residues. These results establish the capacity for functional mimicry of the Trx binding ability of the NF-kappaB/Rel family of transcription factors by the putative HIV-1 pro-fs protein.
Collapse
Affiliation(s)
- Guoping Su
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602-2352, USA.
| | | | | |
Collapse
|
88
|
Todorov VT, Völkl S, Friedrich J, Kunz-Schughart LA, Hehlgans T, Vermeulen L, Haegeman G, Schmitz ML, Kurtz A. Role of CREB1 and NFκB-p65 in the Down-regulation of Renin Gene Expression by Tumor Necrosis Factor α. J Biol Chem 2005; 280:24356-62. [PMID: 15857826 DOI: 10.1074/jbc.m502968200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Tumor necrosis factor-alpha (TNFalpha) is a potent inhibitor of renin gene expression in renal juxtaglomerular cells. We have found that TNFalpha suppresses renin transcription via transcription factor NFkappaB, which targets a cAMP responsive element (CRE) in the renin promoter. Here we aimed to further clarify the role of NFkappaB and the canonical CRE-binding proteins of the CRE-binding protein/activating transcription factor (CREB/ATF) family in the inhibition of renin gene expression by TNFalpha in the juxtaglomerular cell line As4.1. TNFalpha caused a moderate decrease in the binding of CREB1 to its cognate CRE DNA binding site. On the other hand, NFkappaB-p65 transcriptional activity was substantially reduced by TNFalpha, which targeted a trans-activation domain at the very C terminus of the p65 molecule. Our results suggest that TNFalpha inhibits renin gene expression by decreasing the transactivating capacity of NFkappaB-p65 and partially by attenuating CREB1 binding to CRE.
Collapse
Affiliation(s)
- Vladimir T Todorov
- Institute of Physiology, Institute of Pathology, and Department of Immunology, Regensburg University, D-93040 Regensburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Clarke P, Debiasi RL, Meintzer SM, Robinson BA, Tyler KL. Inhibition of NF-kappa B activity and cFLIP expression contribute to viral-induced apoptosis. Apoptosis 2005; 10:513-24. [PMID: 15909114 PMCID: PMC2394667 DOI: 10.1007/s10495-005-1881-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Virus-induced activation of nuclear factor-kappa B (NF-kappaB) is required for Type 3 (T3) reovirus-induced apoptosis. We now show that NF-kappaB is also activated by the prototypic Type 1 reovirus strain Lang (T1L), which induces significantly less apoptosis than T3 viruses, indicating that NF-kappaB activation alone is not sufficient for apoptosis in reovirus-infected cells. A second phase of virus-induced NF-kappaB regulation, where NF-kappaB activation is inhibited at later times following infection with T3 Abney (T3A), is absent in T1L-infected cells. This suggests that inhibition of NF-kappaB activation at later times post infection also contributes to reovirus-induced apoptosis. Reovirus-induced inhibition of stimulus-induced activation of NF-kappaB is significantly associated with apoptosis following infection of HEK293 cells with reassortant reoviruses and is determined by the T3 S1 gene segment, which is also the primary determinant of reovirus-induced apoptosis. Inhibition of stimulus-induced activation of NF-kappaB also occurs following infection of primary cardiac myocytes with apoptotic (8B) but not non-apoptotic (T1L) reoviruses. Expression levels of the NF-kappaB-regulated cellular FLICE inhibitory protein (cFLIP) reflect NF-kappaB activation in reovirus-infected cells. Further, inhibition of NF-kappaB activity and cFLIP expression promote T1L-induced apoptosis. These results demonstrate that inhibition of stimulus-induced activation of NF-kappaB and the resulting decrease in cFLIP expression promote reovirus-induced apoptosis.
Collapse
Affiliation(s)
- P Clarke
- Department of Neurology, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA.
| | | | | | | | | |
Collapse
|
90
|
Zaidi G, Panda H, Supakar PC. Increased phosphorylation and decreased level of IκBα during aging in rat liver. Biogerontology 2005; 6:141-5. [PMID: 16034681 DOI: 10.1007/s10522-005-3459-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2004] [Accepted: 01/26/2005] [Indexed: 10/25/2022]
Abstract
Nuclear factor kappa B (NFkappaB) is an evolutionary conserved transcription factor, which coordinates various metabolic processes triggered by innate and adaptive immune responses. Supakar et al. (J. Biol. Chem. 270: 837-842, 1995) had reported a 10-fold increase in DNA binding activity of NFkappaB in liver of old rats. In this study, we have analyzed the changes in the level of NFkappaB, inhibitor of NFkappaB (IkappaBalpha), phosphorylated-IkappaBalpha (p-IkappaBalpha) and IkappaB kinase (IKK) in rat liver during aging by reverse transcription polymerase chain reaction and/or western blotting. Here we demonstrate that there is an age-dependent increase in the level of p-IkappaBalpha with concomitant decrease in the level of IkappaBalpha, which may be correlated with increased inflammation, oxidative stress and higher level of activated NFkappaB in rat liver in old age.
Collapse
Affiliation(s)
- Ghazala Zaidi
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Orissa, India
| | | | | |
Collapse
|
91
|
Brown TT, Zelnik DL, Dobs AS, Evans DB, Chiao PJ. Fish oil supplementation in the treatment of cachexia in pancreatic cancer patients. INTERNATIONAL JOURNAL OF GASTROINTESTINAL CANCER 2005. [PMID: 15361649 DOI: 10.1385/ijgc:] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Patients with pancreatic cancer often experience a loss of weight and appetite, known as the anorexia-cachexia syndrome, which is associated with decreased quality of life and reduced survival. Research into the biological mechanisms of cachexia has demonstrated that an array of inflammatory mediators and tumor-derived factors cause appetite suppression, skeletal muscle proteolysis, and lipolysis,producing an overall hypercatabolic state that contributes to loss of fat and lean body mass. Omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been shown to modulate levels of proinflammatory cytokines, hepatic acute phase proteins, eicosanoids, and tumor-derived factors in animal models of cancer and may reverse some aspects of the process of cachexia. Results of clinical trials of n-3 PUFAs in the form of fish oils have been mixed, but should encourage further investigation into dietary fish oil supplementation, including the most effective route of administration and the proper dosage to promote optimal weight maintenance and to limit side effects. Concerns about standardization and quality control should also be considered. With the current available evidence, a recommendation for the use of omega 3 polyunsaturated fatty acids in pancreatic cancer cachexia is premature.
Collapse
Affiliation(s)
- Todd T Brown
- Johns Hopkins University School of Medicine, Division of Endocrinology and Metabolism, Center for Complementary and Alternative Medicine, Baltimore, MD 21287, USA
| | | | | | | | | |
Collapse
|
92
|
Kim KM, Lee YJ. Role of HER-2/neu signaling in sensitivity to tumor necrosis factor-related apoptosis-inducing ligand: Enhancement of TRAIL-mediated apoptosis by amiloride. J Cell Biochem 2005; 96:376-89. [PMID: 16059913 DOI: 10.1002/jcb.20512] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been shown to induce apoptosis in numerous transformed cell lines but not in most normal cells. Although this selectivity offers a potential therapeutic application in cancer, not all cancers are sensitive to TRAIL-mediated apoptosis. In this study, we observed that amiloride, a current clinically used diuretic drug, which had little or no cytotoxicity, sensitized TRAIL-resistant human prostate adenocarcinoma LNCaP and human ovarian adenocarcinoma SK-OV-3 cells. The TRAIL-mediated activation of caspase, and PARP cleavage, were promoted in the presence of amiloride. Western blot analysis showed that combined treatment with TRAIL and amiloride did not change the levels of TRAIL receptors (DR4, DR5, and DcR2) and anti-apoptotic proteins (FLIP, IAP, and Bcl-2). However, amiloride dephosphorylated HER-2/neu tyrosine kinase as well as Akt, an anti-apoptotic protein. Interestingly, amiloride also dephosphorylated PI3K and PDK-1 kinases along with PP1alpha phosphatase. In vitro kinase assay revealed that amiloride inhibited phosphorylation of kinase as well as phosphatase by competing with ATP. Taken together, the present studies suggest that amiloride enhances TRAIL-induced cytotoxicity by inhibiting phosphorylation of the HER-2/neu-PI3K-Akt pathway-associated kinases and phosphatase.
Collapse
Affiliation(s)
- Ki M Kim
- Department of Surgery and Pharmacology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | | |
Collapse
|
93
|
Park HJ, Lee YW, Lee SK. Baicalin induces NAD(P)H:quinone reductase through the transactivation of AP-1 and NF-κB in Hepa 1c1c7 cells. Eur J Cancer Prev 2004; 13:521-8. [PMID: 15548947 DOI: 10.1097/00008469-200412000-00010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Baicalin (5,6,7-trihydroxyflavone-7-O-D-glucuronic acid, BA) is a flavone isolated from Scutellariae radix. In our previous report BA was a major active principle of NAD(P)H:quinone reductase (QR) induction mediated by Scutellariae radix extract and the induction was related to the transcriptional activation of the QR gene in Hepa 1c1c7 cells. The primary aim of the present study was to determine the molecular mechanism of QR gene expression by baicalin. The antioxidant or electrophile response element (ARE/EpRE) found at the 5'-flanking region of phase II genes may play an important role in mediating their induction by xenobiotics, including chemopreventive agents. In accordance, to study the molecular mechanisms of QR gene expression by BA, electrophoretic mobility shift assay (EMSA), using nuclear extracts of treated and untreated cells against ARE, activator protein-1 (AP-1) or nuclear factor-kappaB (NF-kappaB) binding sites, showed that BA increased the binding levels of the parameters in a dose-dependent manner. Further, Hepa 1c1c7 cells were transiently transfected with a plasmid containing three copies of the AP-1- or NF-kappaB-binding site linked to a chloramphenicol acetyltransferase (CAT) reporter gene. Using the CAT reporter gene assay, a dose-dependent transactivation of AP-1- or NF-kappaB-mediated CAT expression was observed with the treatment of BA. These results clearly indicate that BA induces the QR gene expression and activity by transactivation of AP-1 and NF-kappaB, and thus BA may be considered as a potential cancer chemopreventive agent with the induction of phase II detoxification enzyme.
Collapse
Affiliation(s)
- H J Park
- College of Pharmacy, Ewha Womans University, 11-1 Daehyun-Dong, Seodaemun-Ku, Seoul 120-750, Korea
| | | | | |
Collapse
|
94
|
Minami T, Horiuchi K, Miura M, Abid MR, Takabe W, Noguchi N, Kohro T, Ge X, Aburatani H, Hamakubo T, Kodama T, Aird WC. Vascular Endothelial Growth Factor- and Thrombin-induced Termination Factor, Down Syndrome Critical Region-1, Attenuates Endothelial Cell Proliferation and Angiogenesis. J Biol Chem 2004; 279:50537-54. [PMID: 15448146 DOI: 10.1074/jbc.m406454200] [Citation(s) in RCA: 182] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation and dysfunction of the endothelium underlie many vascular disorders including atherosclerosis, tumor growth, and inflammation. Endothelial cell activation is mediated by many different extra-cellular signals, which result in overlapping yet distinct patterns of gene expression. Here we show, in DNA microarray analyses, that vascular endothelial growth factor (VEGF) and thrombin result in dramatic and rapid upregulation of Down syndrome critical region (DSCR)-1 gene encoding exons 4-7, a negative feedback regulator of calcium-calcineurin-NF-AT signaling. VEGF- and thrombin-mediated induction of DSCR-1 involves the cooperative binding of NF-ATc and GATA-2/3 to neighboring consensus motifs in the upstream promoter. Constitutive expression of DSCR-1 in endothelial cells markedly impaired NF-ATc nuclear localization, proliferation, and tube formation. Under in vivo conditions, overexpression of DSCR-1 reduced vascular density in matrigel plugs and melanoma tumor growth in mice. Taken together, these findings support a model in which VEGF- and thrombin-mediated induction of endothelial cell proliferation triggers a negative feedback loop consisting of DSCR-1 gene induction and secondary inhibition of NF-AT signaling. As a natural brake in the angiogenic process, this negative pathway may lend itself to therapeutic manipulation in pathological states.
Collapse
Affiliation(s)
- Takashi Minami
- The Research Center for Advanced Science and Technology, the University of Tokyo, Tokyo 153-8904, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Wenk J, Schüller J, Hinrichs C, Syrovets T, Azoitei N, Podda M, Wlaschek M, Brenneisen P, Schneider LA, Sabiwalsky A, Peters T, Sulyok S, Dissemond J, Schauen M, Krieg T, Wirth T, Simmet T, Scharffetter-Kochanek K. Overexpression of Phospholipid-hydroperoxide Glutathione Peroxidase in Human Dermal Fibroblasts Abrogates UVA Irradiation-induced Expression of Interstitial Collagenase/Matrix Metalloproteinase-1 by Suppression of Phosphatidylcholine Hydroperoxide-mediated NFκB Activation and Interleukin-6 Release. J Biol Chem 2004; 279:45634-42. [PMID: 15308634 DOI: 10.1074/jbc.m408893200] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phospholipid-hydroperoxide glutathione peroxidase (PHGPx) exhibits high specific activity in reducing phosphatidylcholine hydroperoxides (PCOOHs) and thus may play a central role in protecting the skin against UV irradiation-triggered detrimental long term effects like cancer formation and premature skin aging. Here we addressed the role of PHGPx in the protection against UV irradiation-induced expression of matrix metalloproteinase-1 (MMP-1). For this purpose, we created human dermal fibroblast cell lines overexpressing human PHGPx. Overexpression led to a significant increase in PHGPx activity. In contrast to a maximal 4.5-fold induction of specific MMP-1 mRNA levels in vector-transfected cells at 24 h after UVA irradiation, no MMP-1 induction occurred at any studied time point after UVA treatment of PHGPx-overexpressing fibroblasts. As interleukin-6 (IL-6) was earlier shown to mediate the UVA induction of MMP-1, we studied whether PHGPx overexpression might interfere with the NFkappaB-mediated IL-6 induction and downstream signaling. Using transient transfections of IL-6 promoter constructs containing NFkappaB binding sites, we observed a high induction of the reporter gene luciferase in vector-transfected control cells and a significantly lower induction in PHGPx-overexpressing fibroblasts following UVA irradiation. Consistently both UVA irradiation and treatment of fibroblasts with PCOOHs led to phosphorylation and nuclear translocation of the p65 subunit, whereas cells overexpressing PHGPx exhibited impaired NFkappaB activation, p65 phosphorylation, and nuclear translocation. In line with this, the PHGPx-overexpressing fibroblasts showed a reduced constitutive and UVA irradiation-induced IL-6 release. After incubating PHGPx-overexpressing cells with PCOOHs a reduced induction of IL-6 was observed. This together with the suppression of UVA irradiation-induced IL-6 release in the presence of Trolox, a chain breaker of PCOOH-initiated lipid peroxidation, indicates that UVA irradiation-induced PCOOHs and subsequent lipid peroxides initiate the NFkappaB-mediated induction of IL-6, which mediates the induction of MMP-1. Our finding is particularly relevant in light of the already available small molecule mimetics of PHGPx.
Collapse
Affiliation(s)
- Jutta Wenk
- Department of Dermatology, University of Cologne, 50924 Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Saccani S, Marazzi I, Beg AA, Natoli G. Degradation of promoter-bound p65/RelA is essential for the prompt termination of the nuclear factor kappaB response. ACTA ACUST UNITED AC 2004; 200:107-13. [PMID: 15226358 PMCID: PMC2213320 DOI: 10.1084/jem.20040196] [Citation(s) in RCA: 219] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Transcription factors of the nuclear factor (NF)-κB/Rel family translocate into the nucleus upon degradation of the IκBs. Postinduction repression of NF-κB activity depends on NF-κB–regulated resynthesis of IκBα, which dissociates NF-κB from DNA and exports it to the cytosol. We found that after activation, p65/RelA is degraded by the proteasome in the nucleus and in a DNA binding–dependent manner. If proteasome activity is blocked, NF-κB is not promptly removed from some target genes in spite of IκBα resynthesis and sustained transcription occurs. These results indicate that proteasomal degradation of p65/RelA does not merely regulate its stability and abundance, but also actively promotes transcriptional termination.
Collapse
Affiliation(s)
- Simona Saccani
- Institute for Research in Biomedicine, Via Vela 6, 6500 Bellinzona, Switzerland
| | | | | | | |
Collapse
|
97
|
Sclabas GM, Fujioka S, Schmidt C, Evans DB, Chiao PJ. NF-kappaB in pancreatic cancer. INTERNATIONAL JOURNAL OF GASTROINTESTINAL CANCER 2004; 33:15-26. [PMID: 12909735 DOI: 10.1385/ijgc:33:1:15] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Although the genetic profile of pancreatic cancer is emerging as a result of much research, the role of specific genetic alterations that initiate tumorigenesis and produce its cardinal clinical features of locally aggressive growth, metastasis, and chemotherapy resistance remains unresolved. Recently, a number of studies have shown that the inhibition of constitutive NF-kappaB activation, one of the frequent molecular alterations in pancreatic cancer, inhibits tumorigenesis and metastasis. It also sensitizes pancreatic cancer cell lines to anticancer agent-induced apoptosis. Therefore because of the crucial role of NF-kappaB in pancreatic cancer, it is a potential target for developing novel therapeutic strategies for the disease. In vivo and in vitro models that mimic the tumorigenic phenotypes in the appropriate histological and molecular concert would be very useful for confirming the suspected role of the pancreatic cancer signature genetic lesions and better understanding the molecular basis of this disease.
Collapse
Affiliation(s)
- Guido M Sclabas
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
98
|
Schmidt C, Peng B, Li Z, Sclabas GM, Fujioka S, Niu J, Schmidt-Supprian M, Evans DB, Abbruzzese JL, Chiao PJ. Mechanisms of proinflammatory cytokine-induced biphasic NF-kappaB activation. Mol Cell 2004; 12:1287-300. [PMID: 14636585 DOI: 10.1016/s1097-2765(03)00390-3] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The transcription factor NF-kappaB regulates genes involved in innate and adaptive immune response, inflammation, apoptosis, and oncogenesis. Proinflammatory cytokines induce the activation of NF-kappaB in both transient and persistent phases. We investigated the mechanism for this biphasic NF-kappaB activation. Our results show that MEKK3 is essential in the regulation of rapid activation of NF-kappaB, whereas MEKK2 is important in controlling the delayed activation of NF-kappaB in response to stimulation with the cytokines TNF-alpha and IL-1alpha. MEKK3 is involved in the formation of the IkappaBalpha:NF-kappaB/IKK complex, whereas MEKK2 participates in assembling the IkappaBbeta:NF-kappaB/IKK complex; these two distinct complexes regulate the proinflammatory cytokine-induced biphasic NF-kappaB activation. Thus, our study reveals a novel mechanism in which different MAP3K and IkappaB isoforms are involved in specific complex formation with IKK and NF-kappaB for regulating the biphasic NF-kappaB activation. These findings provide further insight into the regulation of cytokine-induced specific and temporal gene expression.
Collapse
Affiliation(s)
- Christian Schmidt
- Department of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Abstract
An IkappaBalpha-based NF-kappaB super repressor (sr) has been used widely for studying genes regulated by NF-kappaB transcription factors. Repression of NF-kappaB by IkappaBalpha(sr) also facilitates tumor necrosis factor alpha-induced apoptosis in the cell. However, IkappaBalpha primarily targets RelA and c-Rel-containing complexes, leaving other NF-kappaB/Rel protein complexes, such as p50 and p52 homodimers, and RelB heterodimers uninhibited. Because these atypical NF-kappaB complexes also contribute to gene regulation and are activated in pathological conditions, broad inhibition of all NF-kappaB species is of significant pharmacological and clinical interests. We have designed, generated, and tested a p105-based NF-kappaB super repressor. We showed that p105(sr), which no longer generates p50 and undergoes signal-induced degradation, effectively inhibits all NF-kappaB activities. In addition, we also demonstrated that p105(sr) significantly enhances tumor necrosis factor alpha-mediated killing of MT1/2 skin papilloma cells where p50 homodimer activity is elevated. Our results suggest that p105(sr) is a broader range and effective NF-kappaB super repressor and can potentially be used in cells where a noncanonical NF-kappaB activity is dominant or multiple NF-kappaB activities are activated.
Collapse
Affiliation(s)
- Dexue Fu
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
100
|
Abstract
BACKGROUND Recognition of pathogens by immune receptors leads to activation of macrophages, dendritic cells, and lymphocytes. Signals are communicated to enhance expression of target molecules such as cytokines and adhesion molecules, depending on activation of various inducible transcription factors, among which the family NF-kappaB transcription factors plays an evolutionarily conserved and critical role. Classical activation of NF-kappaB involves phosphorylation, polyubiquitination and subsequent degradation of the inhibitor molecules of NF-kappaB, referred to as IkappaB. Modification of IkappaBalpha, one of the mammalian IkappaB isoforms, with the small ubiquitin-like modifier (SUMO) results its protection from degradation. PRESENTATION OF THE HYPOTHESIS SUMO-IkappaBalpha localizes in the nucleus. The nuclear SUMO-IkappaBalpha pool may be dynamic. SUMO-IkappaBalpha functions as synergy control factor. TESTING THE HYPOTHESIS Immunoprecipitation from cellular fractions, 35S methionine pulse-chase, and FRET assays should reveal the localization of SUMO-IkappaBalpha and the dynamics of the pool. Expression of SUMOylation defective IkappaBalpha in an IkappaBalpha -/- background should yield insights into the function of SUMO-IkappaBalpha. IMPLICATION OF THE HYPOTHESIS IkappaBalpha contains the required SUMOylation motif but IkappaBbeta does not. The suggested study would provide evidence whether or not IkappaBalpha and IkappaBbeta can substitute each other. In addition, the suggested assays would reveal a possible redundancy in controlling transcriptional activity of NF-kappaB.
Collapse
Affiliation(s)
- Martin P Kracklauer
- Section of Molecular Cell and Developmental Biology, Institute for Cellular and Molecular Biology, The University of Texas at Austin, 1 University Station, A4800, 78712, Austin, TX, USA
| | - Christian Schmidt
- Department of Surgical Oncology and Molecular Oncology, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| |
Collapse
|