1801
|
Boersma MC, Meffert MK. Novel roles for the NF-kappaB signaling pathway in regulating neuronal function. Sci Signal 2008; 1:pe7. [PMID: 18272467 DOI: 10.1126/stke.16pe7] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Two new reports offer exciting evidence of novel roles for components of the nuclear factor kappaB (NF-kappaB) pathway in the nervous system. Transcriptional activation by NF-kappaB and chromatin remodeling by inhibitor of kappaB (IkappaB) kinase complex (IKK) have been linked to recall and reconsolidation of conditioned fear memories in the mammalian central nervous system. In the Drosophila neuromuscular junction, a member of the NF-kappaB family has been reported to regulate glutamate receptor clustering. Both reports could have important implications for the function of the NF-kappaB signaling pathway in neuronal plasticity.
Collapse
Affiliation(s)
- Matt C Boersma
- Departments of Biological Chemistry and Neuroscience, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | | |
Collapse
|
1802
|
Abstract
The NF-kappaB (nuclear factor kappaB) transcription factors control cell survival, proliferation and innate and adaptive immune response. Post-translational modifications of key components of the NF-kappaB pathway provide the molecular basis for signal transmission from the cell membrane to the nucleus. Here, we describe the involvement of different types of ubiquitin modification in the regulation of the NF-kappaB signalling pathway.
Collapse
|
1803
|
NF-kappaB signaling in skeletal muscle: prospects for intervention in muscle diseases. J Mol Med (Berl) 2008; 86:747-59. [PMID: 18246321 PMCID: PMC2480606 DOI: 10.1007/s00109-008-0308-4] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2007] [Revised: 11/23/2007] [Accepted: 12/10/2007] [Indexed: 01/01/2023]
Abstract
Muscle remodeling is an important physiological process that promotes adaptive changes in cytoarchitecture and protein composition after exercise, aging, or disease conditions. Numerous transcription factors have been reported to regulate skeletal muscle homeostasis. NF-κB is a major pleiotropic transcription factor modulating immune, inflammatory, cell survival, and proliferating responses; however, its role in muscle development, physiology, and disease has just started to be elucidated. The current review article aims to summarize the literature on the role of NF-κB signaling in skeletal muscle pathophysiology, investigated over the last years using in vitro and more recently in vivo systems. Understanding the exact role of NF-κB in muscle cells will allow better therapeutic manipulations in the setting of human muscle diseases.
Collapse
|
1804
|
Yu M, Han J, Cui P, Dai M, Li H, Zhang J, Xiu R. Cisplatin up-regulates ICAM-1 expression in endothelial cell via a NF-kappaB dependent pathway. Cancer Sci 2008; 99:391-7. [PMID: 18271937 PMCID: PMC11159323 DOI: 10.1111/j.1349-7006.2008.00696.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The anticancer drug cis-diammindichloroplatin (cisplatin) can cause severe side-effects, but to date, the mechanisms of action of these dangerous side-effects have not been completely elucidated. Since cellular adhesion molecules (CAMs), by mediating the recruitment of circulating leukocytes to the blood vessel wall and their subsequent migration into the subendothelial spaces, play a crucial role in several pathophysiologic processes, we sought potential proof for CAMs in the pathophysiology of cisplatin-induced vascular damage. In vitro, human umbilical vein endothelial cells (HUVECs) were subjected to various concentrations of cisplatin, considerable up-regulation of intercellular adhesion molecule-1 (ICAM-1) but not P-selectin, E-selectin, and vascular cell adhesion molecule 1 at both messenger mRNA and protein expression levels were observed. Electrophoretic mobility shift assays and Western blotting analysis revealed that cisplatin up-regulates ICAM-1 expression in HUVECs via an NF-kappaB-dependent pathway. Further intravital microscopy study demonstrated that significantly higher (P < 0.01) numbers of rolling and sticking leukocytes on the wall of postcapillary venules in male Golden Syrian hamster's cheek pouch bearing a human cervical carcinoma were observed, while inhibition of ICAM-1 by using specific anti-ICAM-1 antibody can attenuate cisplatin-stimulated leukocyte/endothelium interactions. These data suggest that ICAM-1 involves in the pathophisiologic process of cisplatin-induced vascular toxicity and may be exploited for therapeutic advantage.
Collapse
Affiliation(s)
- Minghua Yu
- Institute of Microcirculation, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
1805
|
Salminen A, Paimela T, Suuronen T, Kaarniranta K. Innate immunity meets with cellular stress at the IKK complex: regulation of the IKK complex by HSP70 and HSP90. Immunol Lett 2008; 117:9-15. [PMID: 18282612 DOI: 10.1016/j.imlet.2007.12.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Revised: 12/19/2007] [Accepted: 12/22/2007] [Indexed: 12/21/2022]
Abstract
Several research models have shown that if cellular stress induces the heat shock response then this will suppress the NF-kappaB-mediated inflammatory response. The NF-kappaB signaling pathway mediates both stress signals and innate immunity signals. Heat shock proteins HSP70 and HSP90 regulate several signaling cascades to maintain cellular homeostasis. Recent studies have revealed that HSP70 and HSP90 proteins regulate the function of the IKK complex which is the major activator of the NF-kappaB complex. The heat shock response can cause the dissociation of the IKK complex, composed of protein kinase subunits IKKalpha and IKKbeta and the regulatory unit NEMO, and inhibit the activation of NF-kappaB signaling. Suppression of immune signaling during cellular stress may be a useful feedback response for helping cells to survive tissue injury. Furthermore, IKKalpha and IKKbeta kinases are important activators of tumorigenesis and hence the inhibition of long-term activation of the IKK complex by HSP70 and HSP90 proteins may prevent cancer development during chronic inflammation.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neuroscience and Neurology, University of Kuopio, P.O. Box 1627, FIN-70211 Kuopio, Finland.
| | | | | | | |
Collapse
|
1806
|
Terragni J, Graham JR, Adams KW, Schaffer ME, Tullai JW, Cooper GM. Phosphatidylinositol 3-kinase signaling in proliferating cells maintains an anti-apoptotic transcriptional program mediated by inhibition of FOXO and non-canonical activation of NFkappaB transcription factors. BMC Cell Biol 2008; 9:6. [PMID: 18226221 PMCID: PMC2268685 DOI: 10.1186/1471-2121-9-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2007] [Accepted: 01/28/2008] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Phosphatidylinositol (PI) 3-kinase is activated by a variety of growth factor receptors and the PI 3-kinase/Akt signaling pathway is a key regulator of cell proliferation and survival. The downstream targets of PI 3-kinase/Akt signaling include direct regulators of cell cycle progression and apoptosis as well as a number of transcription factors. Growth factor stimulation of quiescent cells leads to robust activation of PI 3-kinase, induction of immediate-early genes, and re-entry into the cell cycle. A lower level of PI 3-kinase signaling is also required for the proliferation and survival of cells maintained in the presence of growth factors, but the gene expression program controlled by PI 3-kinase signaling in proliferating cells has not been elucidated. RESULTS We used microarray analyses to characterize the changes in gene expression resulting from inhibition of PI 3-kinase in proliferating cells. The genes regulated by inhibition of PI 3-kinase in proliferating cells were distinct from genes induced by growth factor stimulation of quiescent cells and highly enriched in genes that regulate programmed cell death. Computational analyses followed by chromatin immunoprecipitations demonstrated FOXO binding to both previously known and novel sites in promoter regions of approximately one-third of the up-regulated genes, consistent with activation of FOXO1 and FOXO3a in response to inhibition of PI 3-kinase. NFkappaB binding sites were similarly identified in promoter regions of over one-third of the down-regulated genes. RelB was constitutively bound to promoter regions in cells maintained in serum, however binding decreased following PI 3-kinase inhibition, indicating that PI 3-kinase signaling activates NFkappaB via the non-canonical pathway in proliferating cells. Approximately 70% of the genes targeted by FOXO and NFkappaB regulate cell proliferation and apoptosis, including several regulators of apoptosis that were not previously known to be targeted by these transcription factors. CONCLUSION PI 3-kinase signaling in proliferating cells regulates a novel transcriptional program that is highly enriched in genes that regulate apoptosis. At least one-third of these genes are regulated either by FOXO transcription factors, which are activated following PI 3-kinase inhibition, or by RelB, which is activated by PI 3-kinase via the non-canonical pathway in proliferating cells.
Collapse
Affiliation(s)
- Jolyon Terragni
- Department of Biology, Boston University, Boston MA 02215, USA.
| | | | | | | | | | | |
Collapse
|
1807
|
Torres J, Watt FM. Nanog maintains pluripotency of mouse embryonic stem cells by inhibiting NFkappaB and cooperating with Stat3. Nat Cell Biol 2008; 10:194-201. [PMID: 18223644 DOI: 10.1038/ncb1680] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Accepted: 12/18/2007] [Indexed: 12/12/2022]
Abstract
Embryonic stem (ES) cells are pluripotent cells derived from the inner cell mass of blastocysts. Self-renewal of mouse ES cells depends on activation of Stat3 by leukaemia inhibitory factor (LIF) in collaboration with bone morphogenetic protein signalling. The transcription factor Nanog is essential in maintaining pluripotency but the mechanisms involved are poorly understood. Here we examine the functional interactions of Nanog with the Stat3 and NFkappaB pathways. Nanog and Stat3 were found to bind to and synergistically activate Stat3-dependent promoters. We also found that Nanog binds to NFkappaB proteins; however, Nanog binding inhibited transcriptional activity of NFkappaB proteins. Endogenous NFkappaB activity and target-gene expression increased during differentiation of ES cells. Overexpression of NFkappaB proteins promoted differentiation, whereas inhibition of NFkappaB signalling, either by genetic ablation of the Ikbkg gene or overexpression of the IkappaBalpha super-repressor, increased expression of pluripotency markers. We conclude that Nanog represses the pro-differentiation activities of NFkappaB and cooperates with Stat3 to maintain pluripotency.
Collapse
Affiliation(s)
- Josema Torres
- Wellcome Trust Centre for Stem Cell Research, Tennis Court Road, Cambridge CB2 1QR, UK
| | | |
Collapse
|
1808
|
Arciniegas E, Carrillo LM, De Sanctis JB, Candelle D. Possible role of NFkappaB in the embryonic vascular remodeling and the endothelial mesenchymal transition process. Cell Adh Migr 2008; 2:17-29. [PMID: 19262121 DOI: 10.4161/cam.2.1.5789] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The NFkappaB family of transcription factors, particularly the activated p50/p65 heterodimer, is expressed in vascular cells during intimal thickening formation when hemodynamic conditions are altered. Here, we report that p50, p65, IkappaBalpha and IKKalpha display different spatial and temporal patterns of expression and distribution during both chicken embryo aortic wall remodeling and intimal thickening development. Additionally, we show that both p50 and p65 were located in the nucleus of some mesenchymal cells expressing alpha-smooth muscle actin which are present in the spontaneous intimal thickening observed at embryonic days 12-14 of development. We also demonstrated that both NFkappaB subunits are present in monolayers of primary embryonic aortic endothelial cells attached to fibronectin and stimulated with complete medium. This study demonstrates for the first time the presence of activated NFkappaB during the remodeling of the embryonic aortic wall and the formation of intimal thickening, providing evidence that suggest a possible role for this transcription factor in the EndoMT process.
Collapse
Affiliation(s)
- Enrique Arciniegas
- Facultad de Medicina, Servicio Autónomo Instituto de Biomedicina, Universidad Central de Venezuela, Caracas, Venezuela.
| | | | | | | |
Collapse
|
1809
|
Mutations in the zinc finger domain of IKK gamma block the activation of NF-kappa B and the induction of IL-2 in stimulated T lymphocytes. Mol Immunol 2008; 45:1633-45. [PMID: 18207244 DOI: 10.1016/j.molimm.2007.09.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2007] [Accepted: 09/18/2007] [Indexed: 01/06/2023]
Abstract
Mutations in the zinc finger of I kappa B kinase gamma (IKK gamma) are associated with hypohidrotic ectodermal dysplasia-immune deficiency (HED-ID) in which the major immune deficit is the inability to switch Ab heavy chain class. However, the pathophysiologic role of the mutations has not been fully delineated. Since help from activated Th cells is essential in Ab class switching, we sought to examine how these mutations affect T cell activation. Using a human T cell line that was null for IKK gamma, we generated cells stably expressing two of the reported mutations, namely, D406V and C417R. Cells expressing either mutation failed to induce IL-2 following stimulation with PMA/ionomycin while the induction of IL-2 was restored in cells reconstituted with the wild type IKK gamma. The lack of IL-2 upregulation correlated with the lack of NF-kappaB activation as evidenced by the inability to induce I kappa B alpha degradation, NF-kappaB binding to DNA and the expression of a reporter gene. However, both mutations did not prevent the incorporation of IKK gamma into the IKK complex and, interestingly, the induced phosphorylation of I kappa B alpha at S32 and S36 and its subsequent ubiquitination were not affected. The suppression of IL-2 induction was solely due to the inhibition of NF-kappaB activation as the mutations did not impair the activation of AP-1 and NFAT. Our data indicated that the failure of T cells to undergo activation in response to TCR stimuli may play a role in the pathophysiology of HED-ID and also showed that IKK gamma has a role in the post-ubiquitination processing of I kappa B alpha.
Collapse
|
1810
|
Clemo NK, Collard TJ, Southern SL, Edwards KD, Moorghen M, Packham G, Hague A, Paraskeva C, Williams AC. BAG-1 is up-regulated in colorectal tumour progression and promotes colorectal tumour cell survival through increased NF-κB activity. Carcinogenesis 2008; 29:849-57. [DOI: 10.1093/carcin/bgn004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
1811
|
Moss BL, Gross S, Gammon ST, Vinjamoori A, Piwnica-Worms D. Identification of a ligand-induced transient refractory period in nuclear factor-kappaB signaling. J Biol Chem 2008; 283:8687-98. [PMID: 18203717 DOI: 10.1074/jbc.m706831200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
In response to a variety of extracellular ligands, nuclear factor-kappaB (NF-kappaB) signaling regulates inflammation, cell proliferation, and apoptosis. It is likely that cells are not continuously exposed to stimulating ligands in vivo but rather experience transient pulses. To study the temporal regulation of NF-kappaB and its major regulator, inhibitor of NF-kappaBalpha (IkappaBalpha), in real time, we utilized a novel transcriptionally coupled IkappaBalpha-firefly luciferase fusion reporter and characterized the dynamics and responsiveness of IkappaBalpha processing upon a short 30-s pulse of tumor necrosis factor alpha (TNFalpha) or a continuous challenge of TNFalpha following a 30-s preconditioning pulse. Strikingly, a 30-s pulse of TNFalpha robustly activated inhibitor of NF-kappaB kinase (IKK), leading to IkappaBalpha degradation, NF-kappaB nuclear translocation, and strong transcriptional up-regulation of IkappaBalpha. Furthermore, we identified a transient refractory period (lasting up to 120 min) following preconditioning, during which the cells were not able to fully degrade IkappaBalpha upon a second TNFalpha challenge. Kinase assays of IKK activity revealed that regulation of IKK activity correlated in part with this transient refractory period. In contrast, experiments involving sequential exposure to TNFalpha and interleukin-1beta indicated that receptor dynamics could not explain this phenomenon. Utilizing a well accepted computational model of NF-kappaB dynamics, we further identified an additional layer of regulation, downstream of IKK, that may govern the temporal capacity of cells to respond to a second proinflammatory insult. Overall, the data suggested that nuclear export of NF-kappaB.IkappaBalpha complexes represented another rate-limiting step that may impact this refractory period, thereby providing an additional regulatory mechanism.
Collapse
Affiliation(s)
- Britney L Moss
- Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
1812
|
Salminen A, Suuronen T, Huuskonen J, Kaarniranta K. NEMO shuttle: a link between DNA damage and NF-kappaB activation in progeroid syndromes? Biochem Biophys Res Commun 2008; 367:715-8. [PMID: 18201555 DOI: 10.1016/j.bbrc.2007.11.189] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Accepted: 11/15/2007] [Indexed: 12/31/2022]
Abstract
Activation of NF-kappaB transcription factor signaling is one of the hallmarks of genotoxic stress. Recently, the NEMO shuttle was revealed to mediate this nucleo-cytoplasmic signaling linking DNA damage to the activation of NF-kappaB system. DNA damage is the causative factor of several segmental progeroid syndromes, such as Werner syndrome and Hutchinson-Gilford syndrome. Although the gene defects have been well characterized, the molecular mechanisms of premature aging process still need to be defined. Here we review the details of the NEMO shuttle, a dual-signal sensor linking DNA damage to NF-kappaB activation, and present evidence for the hypothesis that DNA damage in progeroid syndromes may activate the NEMO shuttle and subsequently increase the pressure on the activation of NF-kappaB system evoking a premature aging phenotype. The NEMO shuttle may link genotoxic stress to the activation of the innate immunity system and cause premature aging via inflamm-aging process.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neuroscience and Neurology, University of Kuopio, P.O. Box 1627, FIN-70211 Kuopio, Finland
| | | | | | | |
Collapse
|
1813
|
Dai Y, Chen S, Kramer LB, Funk VL, Dent P, Grant S. Interactions between bortezomib and romidepsin and belinostat in chronic lymphocytic leukemia cells. Clin Cancer Res 2008; 14:549-558. [PMID: 18223231 DOI: 10.1158/1078-0432.ccr-07-1934] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE The goal of this study was to characterize interactions between the proteasome inhibitor bortezomib and the histone deacetylase (HDAC) inhibitors (HDACI) romidepsin or belinostat in chronic lymphocytic leukemia (CLL) cells. EXPERIMENTAL DESIGN Primary and cultured (JVM-3 and MEC-2) CLL cells were exposed to agents alone or in combination, after which cell death was determined by 7-aminoactinomycin D staining/flow cytometry. Acetylation of target proteins, activation of caspase cascades, and expression of apoptosis-regulatory proteins were monitored by Western blot analysis. Nuclear factor-kappaB (NF-kappaB) activity was determined by luciferase reporter assay. Cells were transiently transfected with wild-type and acetylation site-mutated (inactive) RelA(p65) (e.g., K221R, K310R, or K281/221/310R) and assessed for HDACI sensitivity. RESULTS Combined exposure to very low concentrations of romidepsin or belinostat (i.e., low nanomolar and submicromolar, respectively) in combination with low nanomolar concentrations of bortezomib synergistically induced cell death in primary and cultured CLL cells. These events were likely associated with prevention of HDACI-mediated RelA acetylation and NF-kappaB activation by bortezomib, down-regulation of antiapoptotic proteins (i.e., Bcl-xL, Mcl-1, and XIAP), as well as up-regulation of the proapoptotic protein Bim, resulting in activation of caspase cascade. Finally, CLL cells transfected with inactive RelA displayed a significant increase in HDACI lethality. CONCLUSIONS Coadministration of the clinically relevant HDACIs romidepsin or belinostat with bortezomib synergistically induces cell death in CLL cells, likely through mechanisms involving, among other factors, NF-kappaB inactivation and perturbation in the expression of proapoptotic and antiapoptotic proteins. A strategy combining HDAC with proteasome inhibition warrants further attention in CLL.
Collapse
Affiliation(s)
- Yun Dai
- Department of Medicine, Virginia Commonwealth University/Massey Cancer Center, Richmond, VA 23298, USA
| | | | | | | | | | | |
Collapse
|
1814
|
Abstract
The NF-kappaB (nuclear factor kappaB) regulator A20 antagonises IKK [IkappaB (inhibitor of kappaB) kinase] activation by modulating Lys63-linked polyubiquitination of cytokine-receptor-associated factors including TRAF2/6 (tumour-necrosis-factor-receptor-associated factor 2/6) and RIP1 (receptor-interacting protein 1). In the present paper we describe the crystal structure of the N-terminal OTU (ovarian tumour) deubiquitinase domain of A20, which differs from other deubiquitinases but shares the minimal catalytic core with otubain-2. Analysis of conserved surface regions allows prediction of ubiquitin-binding sites for the proximal and distal ubiquitin molecules. Structural and biochemical analysis suggests a novel architecture of the catalytic triad, which might be present in a subset of OTU domains including Cezanne and TRABID (TRAF-binding domain). Biochemical analysis shows a preference of the isolated A20 OTU domain for Lys48-linked tetraubiquitin in vitro suggesting that additional specificity factors might be required for the physiological function of A20 in cells.
Collapse
|
1815
|
Vykhovanets EV, Shukla S, MacLennan GT, Resnick MI, Carlsen H, Blomhoff R, Gupta S. Molecular imaging of NF-kappaB in prostate tissue after systemic administration of IL-1 beta. Prostate 2008; 68:34-41. [PMID: 18004768 DOI: 10.1002/pros.20666] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Activation of nuclear-factor kappaB (NF-kappaB) influences the transcription of number of genes, many of which participate in inflammatory responses and tumor development. A wide range of human cancers and inflammatory disorders express inappropriate regulation of NF-kappaB. The role of NF-kappaB in intraprostatic inflammation has not been elucidated. METHODS Using transgenic NF-kappaB-Luciferase Tag mice coupled to the luciferase reporter gene, we performed serial, noninvasive in vivo and ex vivo molecular imaging of NF-kappaB activation in the mouse body after systemic administration of mouse pro-inflammatory cytokines: TNF-alpha, IL-6, and IL-1 beta at 10 microg/kg body weights. In some experiments, pretreatment with dexamethasone (10 mg/kg) was used to modulate the cytokine-induced NF-kappaB-dependent luminescence in vivo. RESULTS Treatment of NF-kappaB-Luc mice with cytokines increased luminescence in a time- and organ- specific manner. Highest levels of NF-kappaB-dependent luminescence were observed approximately 3-4 hr after IL-1 beta administration. An important finding was the cumulative effect of IL-1 beta to activate NF-kappaB in the prostate during chronic administration. CONCLUSIONS The molecular imaging of NF-kappaB activity might be an attractive approach to distinguish the role of cytokine-induced NF-kappaB signaling in intraprostatic inflammation and prostate cancer development. Since dexamethasone, a known NF-kappaB inhibitor, could reduce the IL-1 beta-induced NF-kappaB-dependent luminescence in the prostate, NF-kappaB-Luc mice might be useful tool to screen potential candidate drugs for treatment of inflammation and tumor associated with aberrant NF-kappaB activity.
Collapse
Affiliation(s)
- Eugene V Vykhovanets
- Department of Urology, Case Western Reserve University, University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA
| | | | | | | | | | | | | |
Collapse
|
1816
|
Xie Y, Deng S, Thomas CJ, Liu Y, Zhang YQ, Rinderspacher A, Huang W, Gong G, Wyler M, Cayanis E, Aulner N, Többen U, Chung C, Pompou S, Southall N, Vidović D, Schürer S, Branden L, Davis RE, Staudt LM, Inglese J, Austin CP, Landry DW, Smith DH, Auld DS. Identification of N-(quinolin-8-yl)benzenesulfonamides as agents capable of down-regulating NFkappaB activity within two separate high-throughput screens of NFkappaB activation. Bioorg Med Chem Lett 2008; 18:329-35. [PMID: 18024113 PMCID: PMC2275118 DOI: 10.1016/j.bmcl.2007.10.100] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2007] [Revised: 10/19/2007] [Accepted: 10/20/2007] [Indexed: 11/17/2022]
Abstract
We describe here a series of N-(quinolin-8-yl)benzenesulfonamides capable of suppressing the NFkappaB pathway identified from two high-throughput screens run at two centers of the NIH Molecular Libraries Initiative. These small molecules were confirmed in both primary and secondary assays of NFkappaB activation and expanded upon through analogue synthesis. The series exhibited potencies in the cell-based assays at as low as 0.6 microM, and several indications suggest that the targeted activity lies within a common region of the NFkappaB pathway.
Collapse
Affiliation(s)
- Yuli Xie
- Department of Medicine, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - ShiXian Deng
- Department of Medicine, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Craig J. Thomas
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH, 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| | - Yidong Liu
- Department of Medicine, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Ya-Qin Zhang
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH, 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| | - Alison Rinderspacher
- Department of Medicine, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Wenwei Huang
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH, 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| | - Gangli Gong
- Department of Medicine, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Michael Wyler
- MLSCN Center at Columbia University, Columbia Genome Center, 1150 St Nicholas Ave., New York, NY 10032, USA
| | - Efithia Cayanis
- MLSCN Center at Columbia University, Columbia Genome Center, 1150 St Nicholas Ave., New York, NY 10032, USA
| | - Nathalie Aulner
- MLSCN Center at Columbia University, Columbia Genome Center, 1150 St Nicholas Ave., New York, NY 10032, USA
| | - Udo Többen
- MLSCN Center at Columbia University, Columbia Genome Center, 1150 St Nicholas Ave., New York, NY 10032, USA
| | - Caty Chung
- Scientific Computing, The Scripps Research Institute, 5353 Parkside Drive, Jupiter FL 33458 USA
| | - Sergey Pompou
- MLSCN Center at Columbia University, Columbia Genome Center, 1150 St Nicholas Ave., New York, NY 10032, USA
| | - Noel Southall
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH, 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| | - Dušica Vidović
- Scientific Computing, The Scripps Research Institute, 5353 Parkside Drive, Jupiter FL 33458 USA
| | - Stephan Schürer
- Scientific Computing, The Scripps Research Institute, 5353 Parkside Drive, Jupiter FL 33458 USA
| | - Lars Branden
- MLSCN Center at Columbia University, Columbia Genome Center, 1150 St Nicholas Ave., New York, NY 10032, USA
| | - R. Eric Davis
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Building 10, Room 5A/02, Bethesda, MD 20892 USA
| | - Louis M. Staudt
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Building 10, Room 5A/02, Bethesda, MD 20892 USA
| | - James Inglese
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH, 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| | - Christopher P. Austin
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH, 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| | - Donald W. Landry
- Department of Medicine, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Deborah H. Smith
- MLSCN Center at Columbia University, Columbia Genome Center, 1150 St Nicholas Ave., New York, NY 10032, USA
| | - Douglas S. Auld
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH, 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| |
Collapse
|
1817
|
Osipo C, Golde TE, Osborne BA, Miele LA. Off the beaten pathway: the complex cross talk between Notch and NF-kappaB. J Transl Med 2008; 88:11-7. [PMID: 18059366 DOI: 10.1038/labinvest.3700700] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The canonical Notch pathway that has been well characterized over the past 25 years is relatively simple compared to the plethora of recently published data suggesting non-canonical signaling mechanisms and cross talk with other pathways. The manner in which other pathways cross talk with Notch signaling appears to be extraordinarily complex and, not surprisingly, context-dependent. While the physiological relevance of many of these interactions remains to be established, there is little doubt that Notch signaling is integrated with numerous other pathways in ways that appear increasingly complex. Among the most intricate cross talks described for Notch is its interaction with the NF-kappaB pathway, another major cell fate regulatory network involved in development, immunity, and cancer. Numerous reports over the last 11 years have described multiple cross talk mechanisms between Notch and NF-kappaB in diverse experimental models. This article will provide a brief overview of the published evidence for Notch-NF-kappaB cross talk, focusing on vertebrate systems.
Collapse
Affiliation(s)
- Clodia Osipo
- Breast Cancer Program, Cardinal Bernadin Cancer Center, Loyola University Medical Center, Maywood, IL 60513, USA
| | | | | | | |
Collapse
|
1818
|
Abstract
NF-kappaB is a major regulator of the first-line defense against invading pathogens, antigen-specific adaptive immune responses or chemical stress. Stimulation either by extracellular ligands (e.g., inflammatory cytokines, microbial pathogens, peptide antigens) or by intracellular Stressors (e.g., genotoxic drugs) initiates signal-specific pathways that all converge at the IkappaB kinase (IKK) complex, the gatekeeper for NF-kappaB activation. During recent years, considerable progress has been made in understanding the function of NF-kappaB in the regulation of cell growth, survival and apoptosis. In this review, we will focus on the regulation of large signaling complexes on the route to NF-kappaB. Recently published data demonstrate that the assembly, maintenance and activity of the IKK complex determine downstream activation of NF-kappaB. In addition, dynamic complexes upstream of IKK are formed in response to tumor necrosis factor (TNF), antigenic peptides or DNA-damaging agents. Clustering of signaling adaptors promotes the association and activation of ubiquitin ligases that trigger the conjugation of regulatory ubiquitin to target proteins. Ubiquitination serves as a platform to recruit the IKK complex and potentially other protein kinases to trigger IKK activation. These findings support a concept whereby protein complex assembly induces regulatory ubiquitination, which in turn recruits and activates protein kinases. Notably, the great interest in a detailed description of the mechanisms that regulate NF-kappaB activity stems from many observations that link dysregulated NF-kappaB signaling with the onset or progression of various diseases, including cancer, chronic inflammation, cardiovascular disorders and neurodegenerative diseases. Thus, the formation of large signaling clusters and regulatory ubiquitin chains represents promising targets for pharmacological intervention to modulate NF-kappaB signal transduction in disease.
Collapse
Affiliation(s)
- E Wegener
- GSF - Research Center for Environment and Health, Institute of Toxicology, Ingolstädter Landstr. 1, Neuherberg, Germany
| | | |
Collapse
|
1819
|
Nicholas C, Batra S, Vargo MA, Voss OH, Gavrilin MA, Wewers MD, Guttridge DC, Grotewold E, Doseff AI. Apigenin blocks lipopolysaccharide-induced lethality in vivo and proinflammatory cytokines expression by inactivating NF-kappaB through the suppression of p65 phosphorylation. THE JOURNAL OF IMMUNOLOGY 2007; 179:7121-7. [PMID: 17982104 DOI: 10.4049/jimmunol.179.10.7121] [Citation(s) in RCA: 252] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
LPS stimulates monocytes/macrophages through the activation of signaling events that modulate the production of inflammatory cytokines. Apigenin, a flavonoid abundantly found in fruits and vegetables, exhibits anti-proliferative and anti-inflammatory activities through poorly defined mechanisms. In this study, we demonstrate that apigenin inhibits the production of proinflammatory cytokines IL-1beta, IL-8, and TNF in LPS-stimulated human monocytes and mouse macrophages. The inhibitory effect on proinflammatory cytokine production persists even when apigenin is administered after LPS stimulation. Transient transfection experiments using NF-kappaB reporter constructs indicated that apigenin inhibits the transcriptional activity of NF-kappaB in LPS-stimulated mouse macrophages. The classical proteasome-dependent degradation of the NF-kappaB inhibitor IkappaBalpha was observed in apigenin LPS-stimulated human monocytes. Using EMSA, we found that apigenin does not alter NF-kappaB-DNA binding activity in human monocytes. Instead we show that apigenin, as part of a non-canonical pathway, regulates NF-kappaB activity through hypophosphorylation of Ser536 in the p65 subunit and the inactivation of the IKK complex stimulated by LPS. The decreased phosphorylation on Ser536 observed in LPS-stimulated mouse macrophages treated with apigenin was overcome by the over-expression of IKKbeta. In addition, our studies indicate that apigenin inhibits in vivo LPS-induced TNF and the mortality induced by lethal doses of LPS. Collectively, these findings suggest a molecular mechanism by which apigenin suppresses inflammation and modulates the immune response in vivo.
Collapse
Affiliation(s)
- Courtney Nicholas
- Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
1820
|
IKKγ (NEMO) is involved in the coordination of the AP-1 and NF-κB pathways. Mol Cell Biochem 2007; 310:181-90. [DOI: 10.1007/s11010-007-9679-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2007] [Accepted: 12/05/2007] [Indexed: 10/22/2022]
|
1821
|
Oem JK, Jackel-Cram C, Li YP, Kang HN, Zhou Y, Babiuk LA, Liu Q. Hepatitis C virus non-structural protein-2 activates CXCL-8 transcription through NF-kappaB. Arch Virol 2007; 153:293-301. [PMID: 18074095 DOI: 10.1007/s00705-007-1103-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Accepted: 10/24/2007] [Indexed: 01/27/2023]
Abstract
Hepatitis C is a devastating disease worldwide. Proteins encoded by the etiologic agent, hepatitis C virus (HCV), are believed to play important roles in HCV-associated pathogenesis. However, the biological functions of the non-structural protein-2 (NS2) encoded by HCV are not well characterized. Here, we show that HCV NS2 protein activates CXCL-8 (interleukin-8, IL-8) transcription in HepG2 cells as measured by reverse transcription-polymerase chain reaction and IL-8 promoter-luciferase reporter assays. Furthermore, when the kappaB site on the IL-8 promoter was eliminated by mutagenesis or when intracellular NF-kappaB activity was suppressed by an inhibitor, NS2 did not activate the IL-8 promoter, suggesting a role of NF-kappaB in this process. These results prompted us to hypothesize that HCV NS2 might be able to activate NF-kappaB. This hypothesis was tested by determination of NF-kappaB-driven reporter gene expression and NF-kappaB p65 subunit subcellular localization after HCV NS2 expression. Indeed, NS2 could up-regulate NF-kappaB-driven luciferase activity and was associated with p65 nuclear localization. These results demonstrate that HCV NS2 up-regulates IL-8 transcription through NF-kappaB. This newly identified function increases our understanding of the role of HCV NS2 protein in virus-host interactions.
Collapse
Affiliation(s)
- J-K Oem
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK, Canada
| | | | | | | | | | | | | |
Collapse
|
1822
|
Syed MM, Phulwani NK, Kielian T. Tumor necrosis factor-alpha (TNF-alpha) regulates Toll-like receptor 2 (TLR2) expression in microglia. J Neurochem 2007; 103:1461-71. [PMID: 17961202 DOI: 10.1111/j.1471-4159.2007.04838.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Microglia represent one effector arm of CNS innate immunity as evident by their role in pathogen recognition. We previously reported that exposure of microglia to Staphylococcus aureus (S. aureus), a prevalent CNS pathogen, led to elevated Toll-like receptor 2 (TLR2) expression, a pattern recognition receptor capable of recognizing conserved structural motifs associated with gram-positive bacteria such as S. aureus. In this study, we demonstrate that the proinflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) enhances TLR2 expression in microglia, whereas interleukin-1beta has no significant effect. To determine the downstream signaling events responsible for elevated microglial TLR2 expression in response to TNF-alpha, a series of signal transduction inhibitors were employed. Treatment with caffeic acid phenethyl ester, an inhibitor of redox-mediated nuclear factor-kappa B activation, significantly attenuated TNF-alpha-induced TLR2 expression. Similar results were observed with the IKK-2 and IkappaB-alpha inhibitors SC-514 and BAY 11-7082, respectively. In contrast, no significant alterations in TLR2 expression were observed with protein kinase C or p38 mitogen-activated protein kinase inhibitors. A definitive role for TNF-alpha was demonstrated by the inability of S. aureus to augment TLR2 expression in microglia isolated from TNF-alpha knockout mice. In addition, TLR2 expression was significantly attenuated in brain abscesses of TNF-alpha knockout mice. Collectively, these results indicate that in response to S. aureus, TNF-alpha acts in an autocrine/paracrine manner to enhance TLR2 expression in microglia and that this effect is mediated, in part, by activation of the nuclear factor-kappa B pathway.
Collapse
Affiliation(s)
- Mohsin Md Syed
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | | |
Collapse
|
1823
|
Baetas-Da-Cruz W, Castro P, Guimarães E, Koatz V, Corte-Real S, Cavalcante L. Increase in nuclear translocation of nuclear transcription factor-κB following infection of a human Schwann cell line with Leishmania amazonensis. Br J Dermatol 2007; 158:631-3. [DOI: 10.1111/j.1365-2133.2007.08368.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
1824
|
Geisel J, Kahl F, Müller M, Wagner H, Kirschning CJ, Autenrieth IB, Frick JS. IL-6 and maturation govern TLR2 and TLR4 induced TLR agonist tolerance and cross-tolerance in dendritic cells. THE JOURNAL OF IMMUNOLOGY 2007; 179:5811-8. [PMID: 17947654 DOI: 10.4049/jimmunol.179.9.5811] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Stimulation of naive mouse dendritic cells (DC) with LPS or Pam(3)CSK(4) (P3C) induces production of TNF-alpha via TLR4- or TLR2-signaling. Although tolerance in macrophages has been studied in detail, we investigated the role of TLR agonist concentration and IL-6 for tolerance in DC. P3C- or LPS-primed DC were nonresponsive to P3C or LPS restimulation in terms of TNF-alpha but not IL-6 production. The mechanisms involved in tolerance were dependent on the concentration of the TLR ligand used for DC priming. DC primed with LPS or P3C at high concentrations developed a maturation dependent, IL-6 independent tolerance associated with inhibition of TLR signaling upstream of IkappaB as indicated by decreased IkappaB degradation. In contrast, priming of DC with LPS or P3C at low concentrations resulted in IL-6-dependent tolerance, which was abolished in IL-6 deficient DC, and was not accompanied by maturation of DC or by down-regulation of TLR2 or TLR4. In homotolerogenic DC primed with LPS or P3C at high concentrations, degradation of IkappaB upon restimulation with LPS or P3C was inhibited suggesting tolerance mechanism(s) upstream of IkappaB; in contrast, cross-tolerance in DC primed with LPS or P3C at low concentrations was not associated with reduced IkappaB degradation suggesting tolerance mechanisms downstream of IkappaB. Our data indicate that in naive DC TLR4- and TLR2-stimulation results in homo- and cross-tolerance; the mechanisms involved in tolerance depend on the concentration of the TLR agonist used for DC priming and are governed by IL-6 and maturation.
Collapse
Affiliation(s)
- Julia Geisel
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
1825
|
Moussaieff A, Shohami E, Kashman Y, Fride E, Schmitz ML, Renner F, Fiebich BL, Munoz E, Ben-Neriah Y, Mechoulam R. Incensole acetate, a novel anti-inflammatory compound isolated from Boswellia resin, inhibits nuclear factor-kappa B activation. Mol Pharmacol 2007; 72:1657-64. [PMID: 17895408 DOI: 10.1124/mol.107.038810] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Boswellia resin is a major anti-inflammatory agent in herbal medical tradition, as well as a common food supplement. Its anti-inflammatory activity has been attributed to boswellic acid and its derivatives. Here, we re-examined the anti-inflammatory effect of the resin, using inhibitor of nuclear factor-kappaB alpha (IkappaB alpha) degradation in tumor necrosis factor (TNF) alpha-stimulated HeLa cells for a bioassay-guided fractionation. We thus isolated two novel nuclear factor-kappaB (NF-kappaB) inhibitors from the resin, their structures elucidated as incensole acetate (IA) and its nonacetylated form, incensole (IN). IA inhibited TAK/TAB-mediated IkappaB kinase (IKK) activation loop phosphorylation, resulting in the inhibition of cytokine and lipopolysaccharide-mediated NF-kappaB activation. It had no effect on IKK activity in vitro, and it did not suppress IkappaB alpha phosphorylation in costimulated T-cells, indicating that the kinase inhibition is neither direct nor does it affect all NF-kappaB activation pathways. The inhibitory effect seems specific; IA did not interfere with TNFalpha-induced activation of c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase. IA treatment had a robust anti-inflammatory effect in a mouse inflamed paw model. Cembrenoid diterpenoids, specifically IA and its derivatives, may thus constitute a potential novel group of NF-kappaB inhibitors, originating from an ancient anti-inflammatory herbal remedy.
Collapse
Affiliation(s)
- Arieh Moussaieff
- Department of Medicinal Chemistry and Natural Products, Medical Faculty, Hebrew University, Jerusalem 91120, Israel.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1826
|
Aberrant activation of stress-response pathways leads to TNF-alpha oversecretion in Fanconi anemia. Blood 2007; 111:1913-23. [PMID: 18055871 DOI: 10.1182/blood-2007-07-099218] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Fanconi anemia (FA), an inherited syndrome that associates bone marrow failure, cancer predisposition, and genetic instability, is characterized by an overproduction of the myelosuppressive cytokine TNF-alpha through unknown mechanisms. We demonstrate here that FANC pathway loss-of-function results in the aberrant activation of 2 major stress-signaling pathways: NF-kappaB and MAPKs. These responses are independent on TNF-alpha expression. On the contrary, inhibition of the MAPK pathways normalizes TNF-alpha oversecretion in FA. Moreover, our data show that the overexpression of the matrix metalloproteinase MMP-7 is the key event directly responsible for the high rate of TNF-alpha shedding and release from the cytoplasmic membrane in FA. TNF-alpha overproduction is, indeed, normalized by MMP-7 inhibition. Finally, MAPK inhibition impacts on MMP-7 overexpression. Evidence is provided of the existence of a linear pathway in which FANC mutations activate MAPK signaling that induces MMP-7 overexpression leading, in fine, to TNF-alpha oversecretion. TNF-alpha may, in turn, sustain or amplify both MAPKs and NF-kappaB activation. Aberrant expression or activity of NF-kappaB and/or MAPKs has been already involved in bone marrow failure and leukemia, and their inhibition offered clinical benefit for patients. In conclusion, our data provide a strong rationale for new clinical trials on FA patients.
Collapse
|
1827
|
Barré B, Perkins ND. A cell cycle regulatory network controlling NF-kappaB subunit activity and function. EMBO J 2007; 26:4841-55. [PMID: 17962807 PMCID: PMC2099464 DOI: 10.1038/sj.emboj.7601899] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Accepted: 09/27/2007] [Indexed: 11/09/2022] Open
Abstract
Aberrantly active NF-kappaB complexes can contribute to tumorigenesis by regulating genes that promote the growth and survival of cancer cells. We have investigated NF-kappaB during the cell cycle and find that its ability to regulate the G1-phase expression of key proto-oncogenes is subject to regulation by the integrated activity of IkappaB kinase (IKK)alpha, IKKbeta, Akt and Chk1. The coordinated binding of NF-kappaB subunits to the Cyclin D1, c-Myc and Skp2 promoters is dynamic with distinct changes in promoter occupancy and RelA(p65) phosphorylation occurring through G1, S and G2 phases, concomitant with a switch from coactivator to corepressor recruitment. Akt activity is required for IKK-dependent phosphorylation of NF-kappaB subunits in G1 and G2 phases, where Chk1 is inactive. However, in S-phase, Akt is inactivated, while Chk1 phosphorylates RelA and associates with IKKalpha, inhibiting the processing of the p100 (NF-kappaB2) subunit, which also plays a critical role in the regulation of these genes. These data reveal a complex regulatory network integrating NF-kappaB with the DNA-replication checkpoint and the expression of critical regulators of cell proliferation.
Collapse
Affiliation(s)
- Benjamin Barré
- Division of Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee, Scotland, UK
| | - Neil D Perkins
- Division of Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee, Scotland, UK
| |
Collapse
|
1828
|
Fakhrzadeh L, Laskin JD, Laskin DL. Regulation of caveolin-1 expression, nitric oxide production and tissue injury by tumor necrosis factor-alpha following ozone inhalation. Toxicol Appl Pharmacol 2007; 227:380-9. [PMID: 18207479 DOI: 10.1016/j.taap.2007.11.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2007] [Revised: 11/06/2007] [Accepted: 11/08/2007] [Indexed: 01/15/2023]
Abstract
Alveolar macrophages (AM) and inflammatory mediators including nitric oxide and peroxynitrite contribute to ozone-induced lung injury. The generation of these mediators is regulated, in part, by the transcription factor NF-kappaB. We previously demonstrated a critical role for NF-kappaB p50 in ozone-induced injury. In the present studies mechanisms regulating NF-kappaB activation in the lung after ozone inhalation were analyzed. Treatment of wild type (WT) mice with ozone (0.8 ppm, 3 h) resulted in a rapid increase in NF-kappaB binding activity in AM, which persisted for at least 12 h. This was not evident in mice lacking TNFalpha which are protected from ozone-induced injury; there was also no evidence of nitric oxide or peroxynitrite production in lungs from these animals. These data demonstrate that TNFalpha plays a role in NF-kappaB activation and toxicity. TNFalpha signaling involves PI-3-kinase (PI3K)/protein kinase B (PKB), and p44/42 MAP kinase (MAPK) which are important in NF-kappaB activation. Ozone Inhalation resulted in rapid and transient increases in p44/42 MAPK and PI3K/PKB in AM from WT mice, which was evident immediately after exposure. Caveolin-1, a transmembrane protein that negatively regulates PI3K/PKB and p44/42 MAPK signaling, was downregulated in AM from WT mice after ozone exposure. In contrast, ozone had no effect on caveolin-1, PI3K/PKB or p44/42 MAPK expression in AM from TNFalpha knockout mice. These data, together with our findings that TNFalpha suppressed caveolin-1 expression in cultured AM, suggest that TNFalpha and downstream signaling mediate activation of NF-kappaB and the regulation of inflammatory genes important in ozone toxicity, and that this process is linked to caveolin-1.
Collapse
Affiliation(s)
- Ladan Fakhrzadeh
- Department of Pharmacology and Toxicology, Rutgers University, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | | | | |
Collapse
|
1829
|
R-Roscovitine simultaneously targets both the p53 and NF-κB pathways and causes potentiation of apoptosis: implications in cancer therapy. Cell Death Differ 2007; 15:263-73. [DOI: 10.1038/sj.cdd.4402257] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
1830
|
Abstract
Forkhead box (Fox) proteins are a superfamily of evolutionarily conserved transcriptional regulators, which control a wide spectrum of biological processes. As a consequence, a loss or gain of Fox function can alter cell fate and promote tumorigenesis as well as cancer progression. Here we discuss the evidence that the deregulation of Fox family transcription factors has a crucial role in the development and progression of cancer, and evaluate the emerging role of Fox proteins as direct and indirect targets for therapeutic intervention, as well as biomarkers for predicting and monitoring treatment responses.
Collapse
Affiliation(s)
- Stephen S Myatt
- Cancer Research UK laboratories, Department of Oncology, MRC Cyclotron Building, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | | |
Collapse
|
1831
|
Hinz M, Broemer M, Arslan SC, Otto A, Mueller EC, Dettmer R, Scheidereit C. Signal Responsiveness of IκB Kinases Is Determined by Cdc37-assisted Transient Interaction with Hsp90. J Biol Chem 2007; 282:32311-9. [PMID: 17728246 DOI: 10.1074/jbc.m705785200] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The IkappaB kinase (IKK) holocomplex, containing the kinases IKKalpha, IKKbeta, and the scaffold NEMO (NF-kappaB essential modifier), mediates activation of NF-kappaB by numerous physiological stimuli. Heat shock protein 90 (Hsp90) and the co-chaperone Cdc37 have been indicated as additional subunits, but their specific functions in signal transduction are indistinct. Using an RNA interference approach, we demonstrate that Cdc37 recruits Hsp90 to the IKK complex in a transitory manner, preferentially via IKKalpha. Binding is conferred by N-terminal as well as C-terminal residues of Cdc37. Cdc37 is essential for the maturation of de novo synthesized IKKs into enzymatically competent kinases but not for assembly of an IKK holocomplex. Mature IKKs, T-loop-phosphorylated after stimulation either by receptor-mediated signaling or upon DNA damage, further require Hsp90-Cdc37 to generate an activated state. Thus, the present data denote Hsp90-Cdc37 as a transiently acting essential regulatory component of IKK signaling.
Collapse
Affiliation(s)
- Michael Hinz
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
1832
|
Tudhope SJ, Catley MC, Fenwick PS, Russell REK, Rumsey WL, Newton R, Barnes PJ, Donnelly LE. The Role of IκB Kinase 2, but Not Activation of NF-κB, in the Release of CXCR3 Ligands from IFN-γ-Stimulated Human Bronchial Epithelial Cells. THE JOURNAL OF IMMUNOLOGY 2007; 179:6237-45. [DOI: 10.4049/jimmunol.179.9.6237] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
1833
|
Abstract
Cancer has been seriously threatening the health and life of humans for a long period. Despite the intensive effort put into revealing the underlying mechanisms of cancer, the detailled machinery of carcinogenesis is still far from fully understood. Numerous studies have illustrated that cell signaling is extensively involved in tumor initiation, promotion and progression. Therefore, targeting the key molecules in the oncogenic signaling pathway might be one of the most promising ways to conquer cancer. Some targeted drugs, such as imatinib mesylate (Gleevec), herceptin, gefitinib (Iressa), sorafenib (Nexavar) and sunitinib (Sutent), which evolve from monotarget drug into multitarget ones, have been developed with encouraging effects.
Collapse
Affiliation(s)
- Jin Ding
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai 200438, China
| | | | | |
Collapse
|
1834
|
Sebens Müerköster S, Rausch AV, Isberner A, Minkenberg J, Blaszczuk E, Witt M, Fölsch UR, Schmitz F, Schäfer H, Arlt A. The apoptosis-inducing effect of gastrin on colorectal cancer cells relates to an increased IEX-1 expression mediating NF-κB inhibition. Oncogene 2007; 27:1122-34. [PMID: 17704804 DOI: 10.1038/sj.onc.1210728] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Addressing the puzzling role of amidated gastrin(17) (G17) and the gastrin/CCKB/CCK2 receptor in colorectal carcinogenesis, we analysed potential candidate genes involved in G17-dependent NF-kappaB inhibition and apoptosis. The colorectal carcinoma cell line Colo320 overexpressing the wild-type CCK2 receptor (Colo320wt) underwent G17-induced apoptosis along with suppressed NF-kappaB activation and decreased expression of the antiapoptotic NF-kappaB target genes cIAP1 and cIAP2, whereas G17 was without effect on Colo320 cells expressing a CCK2 receptor bearing a loss of function mutation (Colo320mut). Gene microarray analysis revealed an elevated expression of the stress response gene IEX-1 in G17-treated Colo320wt but not Colo320mut cells. Quantitative real-time PCR and conventional RT-PCR confirmed this G17-dependent increase of IEX-1 expression in Colo320wt cells. If these cells were subjected to IEX-1 knockdown by small interfering RNA transfection, the apoptosis-inducing effect of G17 was abolished. Moreover, tumor necrosis factor alpha (TNFalpha)- or 5-FU-induced apoptosis that is greatly enhanced by G17 treatment in Colo320wt cells was prevented if IEX-1 expression was repressed. Under these conditions of blocked IEX-1 expression, the NF-kappaB activity remained unaffected by G17, in particular in Colo320wt cells co-treated with TNFalpha and also the suppressive effect of G17 on cIAP1 and cIAP2 expression was not observed anymore if IEX-1 expression was blocked. Conversely, IEX-1 overexpression in Colo320mut cells caused an increase of basal and TNFalpha- or 5-FU-induced apoptosis, an effect not further triggered by G17 treatment. Using a xenograft tumor model in severe combined immune deficiency mice, we could show that experimental systemic hypergastrinemia induced by the administration of omeprazole led to enhanced apoptosis as well as to a marked increase of IEX-1 expression in Colo320wt tumors, but not in Colo320mut tumors. These observations indicate that the proapoptotic effect of G17 on human colon cancer cells expressing the wild-type CCK2 receptor is mediated by IEX-1, which modulates NF-kappaB-dependent antiapoptotic protection and thereby exerts tumor-suppressive potential.
Collapse
Affiliation(s)
- S Sebens Müerköster
- Laboratory of Molecular Gastroenterology and Hepatology, 1st Department of Medicine, University Hospital of Schleswig-Holstein, Kiel, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1835
|
Hutti JE, Turk BE, Asara JM, Ma A, Cantley LC, Abbott DW. IkappaB kinase beta phosphorylates the K63 deubiquitinase A20 to cause feedback inhibition of the NF-kappaB pathway. Mol Cell Biol 2007; 27:7451-61. [PMID: 17709380 PMCID: PMC2169042 DOI: 10.1128/mcb.01101-07] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Misregulation of NF-kappaB signaling leads to infectious, inflammatory, or autoimmune disorders. IkappaB kinase beta (IKKbeta) is an essential activator of NF-kappaB and is known to phosphorylate the NF-kappaB inhibitor, IkappaBalpha, allowing it to undergo ubiquitin-mediated proteasomal degradation. However, beyond IkappaBalpha, few additional IKKbeta substrates have been identified. Here we utilize a peptide library and bioinformatic approach to predict likely substrates of IKKbeta. This approach predicted Ser381 of the K63 deubiquitinase A20 as a likely site of IKKbeta phosphorylation. While A20 is a known negative regulator of innate immune signaling pathways, the mechanisms regulating the activity of A20 are poorly understood. We show that IKKbeta phosphorylates A20 in vitro and in vivo at serine 381, and we further show that this phosphorylation event increases the ability of A20 to inhibit the NF-kappaB signaling pathway. Phosphorylation of A20 by IKKbeta thus represents part of a novel feedback loop that regulates the duration of NF-kappaB signaling following activation of innate immune signaling pathways.
Collapse
Affiliation(s)
- Jessica E Hutti
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
1836
|
Matteucci E, Ridolfi E, Maroni P, Bendinelli P, Desiderio MA. c-Src/Histone Deacetylase 3 Interaction Is Crucial for Hepatocyte Growth Factor–Dependent Decrease of CXCR4 Expression in Highly Invasive Breast Tumor Cells. Mol Cancer Res 2007; 5:833-45. [PMID: 17699109 DOI: 10.1158/1541-7786.mcr-07-0054] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hepatocyte growth factor (HGF), a cytokine of tumor microenvironment, exerts opposite effects on CXCR4 expression in MCF-7 (low invasive) and MDA-MB231 (highly invasive) breast carcinoma cells, and here, we show that completely different molecular mechanisms downstream of c-Src activation were involved. As experimental models, we used cells transfected with two CXCR4 promoter constructs and treated with HGF or cotransfected with c-Src wild-type (Srcwt) expression vector; phospho-c-Src formation was enhanced in both cell lines. In MCF-7 cells, consistent with activations of CXCR4Luc constructs after HGF treatment and Srcwt expression, Ets1 and nuclear factor-kappaB (NF-kappaB) transcription factors were activated. In contrast, in MDA-MB231 cells, CXCR4Luc construct, Ets1 and NF-kappaB activities decreased. The divergence point seemed to be downstream of HGF/c-Src and consisted in the interaction between c-Src and the substrate histone deacetylase 3 (HDAC3). Only in MDA-MB231 cells, HDAC3 level was enhanced in membranes and nuclei 30 min after HGF and colocalized/coimmunoprecipitated with phospho-c-Src and phosphotyrosine. Thus, the CXCR4 induction by HGF in MCF-7 cells required NF-kappaB and Ets1 activations, downstream of phosphoinositide-3-kinase/Akt, whereas in HGF-treated MDA-MB231 cells, HDAC3 activation via c-Src probably caused a reduction of transcription factor activities, such as that of NF-kappaB. These results indicate possible roles of HGF in invasive growth of breast carcinomas. By enhancing CXCR4 in low invasive tumor cells, HGF probably favors their homing to secondary sites, whereas by suppressing CXCR4 in highly invasive cells, HGF might participate to retain them in the metastatic sites.
Collapse
Affiliation(s)
- Emanuela Matteucci
- Institute of General Pathology, School of Medicine, University of Milan, via Luigi Mangiagalli, 31-20133 Milan, Italy
| | | | | | | | | |
Collapse
|
1837
|
Souvannavong V, Saidji N, Chaby R. Lipopolysaccharide from Salmonella enterica activates NF-kappaB through both classical and alternative pathways in primary B Lymphocytes. Infect Immun 2007; 75:4998-5003. [PMID: 17698569 PMCID: PMC2044549 DOI: 10.1128/iai.00545-07] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Lipopolysaccharides (LPS) are potent polyclonal B-lymphocyte activators. Recently, we have shown that LPS inhibits both spontaneous and drug-induced apoptosis in mature B lymphocytes, through cytosolic retention of Bax, a proapoptotic protein of the Bcl-2 family, by preventing its translocation to mitochondria. Research within the last few years has revealed that members of the NF-kappaB transcription factor regulate cell viability by activating genes involved in mitochondrion-dependent apoptosis. In this report, we examined the effect of sustained LPS stimulation on cytosolic and nuclear proteins of the IkappaB/NF-kappaB family to determine which NF-kappaB pathway, canonical (classical) or noncanonical (alternative), is activated by this agent in mature B cells. Immunoblotting analyses showed that LPS induced a time-dependent degradation of the NF-kappaB inhibitors IkappaBbeta and IkappaBepsilon (preferentially to isoform IkappaBalpha), via IkappaB kinase beta. In addition, we observed that LPS triggered the processing of NF-kappaB p105 to p50 and that of NF-kappaB p100 to p52 in parallel with nuclear translocation of active p50 and p52, as NF-kappaBp50/RelA and NF-kappaBp52/RelB heterodimers, respectively. These results suggest that sustained stimulation with LPS can activate NF-kappaB through both classical and alternative pathways.
Collapse
Affiliation(s)
- Vongthip Souvannavong
- CNRS, Institut de Biochimie Biophysique Moléculaire et Cellulaire, UMR 8619, Université Paris-Sud, Bat. 430, 91405 Orsay cedex, France.
| | | | | |
Collapse
|
1838
|
Abstract
Nuclear factor-kappaB (NF-kappaB) is a transcriptional regulator of genes involved in immunity, inflammatory response, cell fate, and function. Recent attention has focused on the pathophysiological role of NF-kappaB in the diseased liver. In vivo studies using rodent models of liver disease and cell-targeted perturbation of NF-kappaB activity have revealed complex and multicellular functions in hepatic inflammation, fibrosis, and the development of hepatocellular carcinoma - a process we have termed the "inflammation-fibrosis-cancer axis". This review summarizes the current state of knowledge and provides insight into the vast complexity of the hepatic NF-kappaB signaling system, which should provide a rich source of new therapeutic targets.
Collapse
Affiliation(s)
- Ahmed M Elsharkawy
- Liver Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | |
Collapse
|
1839
|
Radisky DC, Bissell MJ. NF-kappaB links oestrogen receptor signalling and EMT. Nat Cell Biol 2007; 9:361-3. [PMID: 17401385 DOI: 10.1038/ncb0407-361] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
1840
|
|
1841
|
Wu ZH, Miyamoto S. Many faces of NF-kappaB signaling induced by genotoxic stress. J Mol Med (Berl) 2007; 85:1187-202. [PMID: 17607554 DOI: 10.1007/s00109-007-0227-9] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2007] [Revised: 04/20/2007] [Accepted: 05/22/2007] [Indexed: 12/22/2022]
Abstract
The nuclear factor-kappaB (NF-kappaB) family of dimeric transcription factors plays pivotal roles in physiologic and pathologic processes, including immune and inflammatory responses and development and progression of various human cancers. Inactive NF-kappaB dimers normally exist in the cytoplasm in association with inhibitor proteins belonging to the inhibitor of NF-kappaB (IkappaB) family of related proteins. Activation of NF-kappaB involves its release from IkappaB and subsequent nuclear translocation to induce expression of target genes. Intense research effort has revealed many distinct signaling pathways and mechanisms of NF-kappaB activation induced by immune and inflammatory stimuli. These aspects of NF-kappaB biology have been amply reviewed in the literature. However, those that involve DNA-damaging agents are less well understood, and multiple conflicting pathways and mechanisms have been described in the literature. In this review, we summarize the proposed mechanisms of NF-kappaB activation by various DNA-damaging agents, discuss the significance of such activation in the context of cancer treatment, and highlight some of the critical questions that remain to be addressed in future studies.
Collapse
Affiliation(s)
- Zhao-Hui Wu
- Department of Pharmacology, University of Wisconsin-Madison, WI 53706, USA
| | | |
Collapse
|
1842
|
Abstract
Crosstalk between the NF-kappaB and p53 transcription factors can play a pivotal role in determining the cellular response to stress; in a recent issue of Molecular Cell, Huang et al. (2007) show that IkappaB kinase alpha (IKKalpha) can determine which pathway is dominant.
Collapse
|
1843
|
Abstract
Cytokines are pleiotropic substances that are known to participate in inflammatory and immune responses as well as cell differentiation and proliferation. Interleukin-6 (IL-6) is a key cytokine with pro-inflammatory function. Wound healing is a complex cascade of physiologic events comprising inflammation, proliferation and remodeling, and proceeds with the integrated actions of different cells, cytokines, and the extracellular matrix. Aberrant wound healing results in keloid formation which causes disfigured appearance, discomfort, psychological stress, and patient frustration. In this review, the role of IL-6 signaling pathway in the pathogenesis of keloid is assessed and its potential as a therapeutic target is addressed. The existing data suggest that IL-6 mediated inflammation is a key player and may be considered as a common causative factor for development of keloid. Furthermore, in a recent comprehensive study, we confirmed the functional role of IL-6 signaling in keloid pathogenesis. Accordingly, inhibitory strategies of IL-6 signaling pathway by targeting the IL-6 receptors, its downstream effecters, or other molecules influencing this pathway appear to have considerable potential as new therapeutic or preventive challenges for keloid. Hopefully, several IL-6 blocking agents including a humanized antibody to IL-6 receptor have been developed and successfully used in clinical trials of inflammatory diseases. It is likely that these agents may prove worthy in the treatment or prevention of keloid as well. Future in-depth exploration of such challenges will shed light on their efficacy and safety for clinical application in keloid.
Collapse
Affiliation(s)
- Mohammad Ghazizadeh
- Department of Molecular Pathology, Institute of Gerontology, Postgraduate School of Medicine, Nippon Medical School.
| |
Collapse
|
1844
|
González-Ramos R, Donnez J, Defrère S, Leclercq I, Squifflet J, Lousse JC, Van Langendonckt A. Nuclear factor-kappa B is constitutively activated in peritoneal endometriosis. ACTA ACUST UNITED AC 2007; 13:503-9. [PMID: 17483545 DOI: 10.1093/molehr/gam033] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Red (active), black and white endometriotic lesions are characteristic of peritoneal endometriosis. The transcription factor nuclear factor-kappa B (NF-kappaB) activates proinflammatory, proliferative and antiapoptotic genes in many cell types. To determine whether NF-kappaB is activated in peritoneal endometriosis in women, and further ascertain the differential inflammatory status of endometriotic implants, NF-kappaB activation and intercellular adhesion molecule (ICAM)-1 expression were investigated in peritoneal endometriotic lesions according to their type. Furthermore, p65 and p50 subunits of active NF-kappaB dimers were evaluated in endometriotic lesions to gain some insight into NF-kappaB-implicated pathways. Thirty-six biopsies of peritoneal endometriotic lesions were analyzed. Constitutive NF-kappaB activation, involving p65- and p50-containing dimers, was demonstrated in peritoneal endometriotic lesions by electrophoretic mobility shift assays and supershift analyses, as well as NF-kappaB (p65) DNA-binding activity immunodetection assays. NF-kappaB activation and ICAM-1 expression (evaluated by immunoblotting) were significantly higher in red lesions than black lesions, whereas IkappaBalpha (NF-kappaB inhibitory protein) expression was constant, as shown by western blot analysis. This is the first study to demonstrate constitutive NF-kappaB activation in peritoneal endometriosis in women. NF-kappaB activation and ICAM-1 expression in red lesions confirm the more extensive inflammatory pattern of these lesions compared with black lesions. The involvement of p50/p65 dimers in NF-kappaB activation suggests implication of the classic NF-kappaB activation pathway, making it an attractive therapeutic target in endometriosis.
Collapse
Affiliation(s)
- Reinaldo González-Ramos
- Department of Gynecology, Université Catholique de Louvain, Cliniques Universitaires St Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
1845
|
Abstract
The modulation of apoptosis has emerged as an important weapon in the pathogenic arsenal of multiple intracellular protozoan parasites. Cryptosporidium parvum, Leishmania spp., Trypanosoma cruzi, Theileria spp., Toxoplasma gondii and Plasmodium spp. have all been shown to inhibit the apoptotic response of their host cell. While the pathogen mediators responsible for this modulation are unknown, the parasites are interacting with multiple apoptotic regulatory systems to render their host cell refractory to apoptosis during critical phases of intracellular infection, including parasite invasion, establishment and replication. Additionally, emerging evidence suggests that the parasite life cycle stage impacts the modulation of apoptosis and possibly parasite differentiation. Dissection of the host-pathogen interactions involved in modulating apoptosis reveals a dynamic and complex interaction that recent studies are beginning to unravel.
Collapse
Affiliation(s)
- John C Carmen
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | | |
Collapse
|