51
|
Abstract
Complexes formed from the nuclear factor kappaB (NF-kappaB) family of transcription factors are ubiquitously expressed and are induced by a diverse array of stimuli. This results in their becoming activated in a wide variety of different settings. While the functions of NF-kappaB in many of these contexts have been the subject of intense research and are now well established, it is also clear that there is great diversity in the effects and consequences of NF-kappaB activation. NF-kappaB subunits do not necessarily regulate the same genes, in an identical manner, in all of the different circumstances in which they are induced. This review will discuss the different functions of NF-kappaB, the pathways that modulate NF-kappaB subunit activity and, in contrast to its more commonly thought of role as a promoter of cancer cell growth and survival, the ability of NF-kappaB, under some circumstances, to behave as a tumor suppressor.
Collapse
Affiliation(s)
- N D Perkins
- Division of Gene Regulation and Expression, School of Life Sciences, University of Dundee, MSI/WTB Complex, Dow Street, Dundee, Scotland DD1 5EH, UK.
| | | |
Collapse
|
52
|
Rice J, Courter DL, Giachelli CM, Scatena M. Molecular mediators of alphavbeta3-induced endothelial cell survival. J Vasc Res 2006; 43:422-36. [PMID: 16888388 DOI: 10.1159/000094884] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2005] [Accepted: 05/18/2006] [Indexed: 12/19/2022] Open
Abstract
The alphavbeta3 integrin interaction with the extracellular matrix (ECM) plays an essential role in inhibiting apoptosis in endothelial cells. We have recently shown that alphavbeta3 ligation on rat aortic endothelial cells (RAECs) specifically activates the transcription factor nuclear factor kappaB (NF-kappaB) and promotes cell survival. Inhibiting NF-kappaB nuclear translocation abolished the protective effect of alphavbeta3 ligands. Here, we report that ligation of alphavbeta3 by its ligand, osteopontin (OPN), induces the phosphorylation and activation of inhibitory kappa B kinase beta IKKbeta and promotes the specific degradation of inhibitory kappa Balpha (IkappaBalpha) in RAECs. Overexpression of a dominant negative (DN) IKKbeta protein prevents IkappaBalpha phosphorylation, NF-kappaB activation, and inhibits the protective effects of OPN. The NF-kappaB-inducing kinase (NIK) has been shown to be one of the upstream kinases involved in IKK activation. OPN-mediated NF-kappaB activity is increased upon NIK wild-type (WT) overexpression and blocked following NIK DN overexpression. In addition, NIK-/-mouse embryonic fibroblasts (MEFs) plated on OPN display reduced NF-kappaB activity and decreased IkappaBalpha phosphorylation compared to NIK+/+MEFs. Finally, functional inhibition of integrin beta3-dependent NF-kappaB signaling decreases OPN-induced IkappaBalpha, IKKbeta and NIK phosphorylation. These studies for the first time show that the alphavbeta3-NF-kappaB-dependent endothelial survival pathway is dependent on IkappaBalpha, IKKbeta, and NIK.
Collapse
Affiliation(s)
- Julie Rice
- Department of Pathology, University of Washington, Seattle 98195-1720, USA
| | | | | | | |
Collapse
|
53
|
Leu TH, Charoenfuprasert S, Yen CK, Fan CW, Maa MC. Lipopolysaccharide-induced c-Src expression plays a role in nitric oxide and TNFalpha secretion in macrophages. Mol Immunol 2006; 43:308-16. [PMID: 15869794 DOI: 10.1016/j.molimm.2005.03.015] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2005] [Indexed: 11/26/2022]
Abstract
As tyrosine kinases are indispensable in lipopolysaccharide (LPS)-induced macrophage activation, the myeloid-specific Src members (i.e. Lyn, Fgr and Hck) are speculated to play important roles in this process. However, the normal LPS responsiveness in lyn(-/-)fgr(-/-)hck(-/-) macrophages implicates the presence of an elusive, compensating tyrosine kinase(s). In this study, we demonstrate the upregulation of c-Src in Raw264.7 and peritoneal macrophages (PEMs) by LPS, which is inhibited by PP2 (an inhibitor for Src family kinases), pyrrolidinedithiocarbamate (PDTC; NF-kappaB inhibitor) and LY294002 (PI3K inhibitor). And this LPS-mediated c-Src induction is also observed in macrophages recovered from LPS-challenged rats. Intriguingly, PP2 attenuates the ability of PEMs to elicit COX-2 expression and nitric oxide production in response to LPS. Similar results are also observed when macrophages recovered from rats receiving either LPS alone or LPS and PP2 both are compared. Furthermore, administration of PP2 in Raw264.7 and animal models of sepsis greatly suppresses TNFalpha secretion and serum TNFalpha level, respectively. Therefore, we conclude that c-Src, with its LPS induction, has an unperceived role in transmitting LPS signaling in macrophages.
Collapse
Affiliation(s)
- Tzeng-Horng Leu
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | | | | | | | | |
Collapse
|
54
|
Syeda F, Grosjean J, Houliston RA, Keogh RJ, Carter TD, Paleolog E, Wheeler-Jones CPD. Cyclooxygenase-2 induction and prostacyclin release by protease-activated receptors in endothelial cells require cooperation between mitogen-activated protein kinase and NF-kappaB pathways. J Biol Chem 2006; 281:11792-804. [PMID: 16467309 DOI: 10.1074/jbc.m509292200] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The functional significance of protease-activated receptors (PARs) in endothelial cells is largely undefined, and the intracellular consequences of their activation are poorly understood. Here, we show that the serine protease thrombin, a PAR-1-selective peptide (TFLLRN), and SLIGKV (PAR-2-selective peptide) induce cyclooxygenase-2 (COX-2) protein and mRNA expression in human endothelial cells without modifying COX-1 expression. COX-2 induction was accompanied by sustained production of 6-keto-PGF1alpha, the stable hydrolysis product of prostacyclin, and this was inhibited by indomethacin and the COX-2-selective inhibitor NS398. PAR-1 and PAR-2 stimulation rapidly activated both ERK1/2 and p38MAPK, and pharmacological blockade of MEK with either PD98059 or U0126 or of p38MAPK by SB203580 or SB202190 strongly inhibited thrombin- and SLIGKV-induced COX-2 expression and 6-keto-PGF1alpha formation. Thrombin and peptide agonists of PAR-1 and PAR-2 increased luciferase activity in human umbilical vein endothelial cells infected with an NF-kappaB-dependent luciferase reporter adenovirus, and this, as well as PAR-induced 6-keto-PGF1alpha synthesis, was inhibited by co-infection with adenovirus encoding wild-type or mutated (Y42F) IkappaBalpha. Thrombin- and SLIGKV-induced COX-2 expression and 6-keto-PGF1alpha generation were markedly attenuated by the NF-kappaB inhibitor PG490 and partially inhibited by the proteasome pathway inhibitor MG-132. Activation of PAR-1 or PAR-2 promoted nuclear translocation and phosphorylation of p65-NF-kappaB, and thrombin-induced but not PAR-2-induced p65-NF-kappaB phosphorylation was reduced by inhibition of MEK or p38MAPK. Activation of PAR-4 by AYPGKF increased phosphorylation of ERK1/2 and p38MAPK without modifying NF-kappaB activation or COX-2 induction. Our data show that PAR-1 and PAR-2, but not PAR-4, are coupled with COX-2 expression and sustained endothelial production of vasculoprotective prostacyclin by mechanisms that depend on ERK1/2, p38MAPK, and IkappaBalpha-dependent NF-kappaB activation.
Collapse
Affiliation(s)
- Farisa Syeda
- Department of Veterinary Basic Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
55
|
Abu-Amer Y, Faccio R. Therapeutic approaches in bone pathogeneses: targeting the IKK/NF-κB axis. ACTA ACUST UNITED AC 2006. [DOI: 10.2217/17460816.1.1.133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
56
|
Kang JL, Jung HJ, Lee K, Kim HR. Src Tyrosine Kinases Mediate Crystalline Silica-Induced NF-κB Activation through Tyrosine Phosphorylation of IκB-α and p65 NF-κB in RAW 264.7 Macrophages. Toxicol Sci 2006; 90:470-7. [PMID: 16431847 DOI: 10.1093/toxsci/kfj096] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Protein tyrosine kinases (PTKs) and mitogen-activated protein kinases (MAPKs) have been demonstrated to play a crucial role in the signaling pathways induced by silica. In the present study, we investigated whether Src family TKs play a role in crystalline silica-induced NF-kappaB activation and whether NF-kappaB activation requires Src TK-dependent MAPK activity in RAW 264.7 cells, a mouse peritoneal macrophage cell line. Selective Src TK inhibitors, damnacanthal or PP1, inhibited silica-induced NF-kappaB activation in a dose-dependent manner. Furthermore, these kinase inhibitors suppressed silica-induced tyrosine phosphorylation of IkappaB-alpha and p65 NF-kappaB. Within a similar time frame, c-Src and Lck were physically associated with IkappaB-alpha and with p65 NF-kappaB. Silica stimulated the phosphorylation of extracellular signal-regulated kinase 1 and 2 (ERK1/2), but not p38 MAPK and c-Jun NH(2)-terminal kinase 1 and 2 (JNK1/2). Damnacanthal or PP1 substantially blocked the silica-induced activation of ERK1/2. Moreover, PD98059, an inhibitor of ERK1/2, or SB203580, an inhibitor of p38 MAPK, failed to inhibit silica-induced NF-kappaB activation. These results suggest that c-Src and Lck act for silica-induced NF-kappaB activation by mediating the tyrosine phosphorylations of IkappaB-alpha and p65 NF-kappaB. However, the Src TK-dependent activation of ERK1/2 may not be involved in the silica signaling pathway leading to NF-kappaB activation.
Collapse
Affiliation(s)
- Jihee Lee Kang
- Department of Physiology, Division of Cell Biology, Ewha Medical Research Center, Ewha Womans University College of Medicine, Seoul, Korea.
| | | | | | | |
Collapse
|
57
|
Morimoto H, Ozaki A, Okamura H, Yoshida K, Kitamura S, Haneji T. Okadaic acid induces tyrosine phosphorylation of IkappaBalpha that mediated by PKR pathway in human osteoblastic MG63 cells. Mol Cell Biochem 2006; 276:211-7. [PMID: 16132703 DOI: 10.1007/s11010-005-4440-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2004] [Accepted: 03/23/2005] [Indexed: 10/25/2022]
Abstract
Treatment of human osteosarcoma cell line MG 63 cells with okadaic acid stimulated phosphorylation of IkappaBalpha, as judged from the results of Western blot analysis and a lambda protein phosphatase dephosphorylation assay. The stimulated phosphorylation of IkappaBalpha was both time- and dose-dependent. The phosphorylation sites of IkappaBalpha were taken to be tyrosine residues because the anti-phospho-tyrosine antibody bound to the samples immunoprecipitated with the anti-IkappaBalpha antibody. In the cells treated with 100 nM okadaic acid consequential translocation of NF-kappaB p65 from the cytosol to the nucleus occurred. Double-stranded RNA-dependent protein kinase (PKR) is a player in the cellular antiviral response and is involved in transcriptional stimulation through activation of NF-kappaB. We investigated the functional relationship between PKR and IkappaBalpha phosphorylation by constructing MG 63 PKR K/R cells that produced a catalytically inactive mutant PKR. NF-kappaB p65 was detected in the nucleus of these cells, even in the unstimulated cells. Although IkappaBalpha was degraded phosphorylation of eIF-2 alpha, a substrate of PKR, did not occur in the mutant cells treated with okadaic acid. Our results suggest that okadaic acid-induced tyrosine phosphorylation of IkappaBalpha was mediated by PKR kinase activity, thus indicating the involvement of this kinase in the control mechanism governing the activation of NF-kappaB.
Collapse
Affiliation(s)
- Hiroyuki Morimoto
- Department of Oral and Maxillofacial Anatomy, Institute of Health Biosciences, The University of Tokushima Graduate School, 3-18-15, Kuramoto, Tokushima 770-8504, Japan.
| | | | | | | | | | | |
Collapse
|
58
|
Jalal DI, Kone BC. Src activation of NF-kappaB augments IL-1beta-induced nitric oxide production in mesangial cells. J Am Soc Nephrol 2005; 17:99-106. [PMID: 16338964 DOI: 10.1681/asn.2005070693] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
NF-kappaB is a critical transcription factor that is involved in glomerulonephritis and inflammatory host responses and a critical transactivator of the inducible nitric oxide (NO) synthase gene in mesangial cells. The Src protein tyrosine kinases (SFK) are involved in several signaling pathways and have been proposed to mediate cytokine activation of NF-kappaB in a few cell types. However, the specific involvement of SFK in IL-1beta induction of NO production has not been clearly established. Accordingly, pharmacologic and molecular tools were used to clarify this issue in cultured murine mesangial cells. The SFK antagonist 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo(3,4-d)pyrimidine (PP2) dramatically inhibited IL-1beta-mediated induction of endogenous NO production as measured by the Griess reaction, as well as the induction of NF-kappaB p50/p65 DNA-binding activity in gel shift assays and the activity of an NF-kappaB-responsive promoter-reporter construct transiently transfected into the cells. Immunoprecipitation and immunoblotting with anti-IkappaBalpha and anti-phosphotyrosine antibodies revealed that PP2 also inhibited IL-1beta-stimulated tyrosine phosphorylation of IkappaBalpha, a requisite step in NF-kappaB activation in this signaling cascade. In agreement with the pharmacologic inhibition studies, siRNA directed against c-Src specifically limited c-Src protein expression and inhibited IL-1beta-mediated induction of NF-kappaB DNA-binding activity, whereas control siRNA had no effect. Conversely, overexpression of constitutively active c-Src augmented basal and IL-1beta-mediated induction of NF-kappaB DNA-binding activity and NO production. Thus, SFK play a key role in IL-1beta-induced NO production in mesangial cells and do so via tyrosine phosphorylation of IkappaBalpha and consequent NF-kappaB activation.
Collapse
Affiliation(s)
- Diane I Jalal
- Department of Internal Medicine, The University of Texas Medical School at Houston, 6431 Fannin, MSB 1.150, Houston, TX 77030, USA
| | | |
Collapse
|
59
|
Hirayama T, Dai S, Abbas S, Yamanaka Y, Abu-Amer Y. Inhibition of inflammatory bone erosion by constitutively active STAT-6 through blockade of JNK and NF-kappaB activation. ACTA ACUST UNITED AC 2005; 52:2719-29. [PMID: 16142755 DOI: 10.1002/art.21286] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE NF-kappaB and JNK signaling pathways play key roles in the pathogenesis of inflammatory arthritis. Both factors are also activated in response to osteoclastogenic factors, such as RANKL and tumor necrosis factor alpha. Inflammatory arthritis and bone erosion subside in the presence of antiinflammatory cytokines such as interleukin-4 (IL-4). We have previously shown that IL-4 inhibits osteoclastogenesis in vitro through inhibition of NF-kappaB and JNK activation in a STAT-6-dependent manner. This study was undertaken to investigate the potential of constitutively active STAT-6 to arrest the activation of NF-kappaB and JNK and to subsequently ameliorate the bone erosion associated with inflammatory arthritis in mice. METHODS Inflammatory arthritis was induced in wild-type and STAT-6-null mice by intraperitoneal injection of arthritis-eliciting serum derived from K/BxN mice. Bone erosion was assessed in the joints by histologic and immunostaining techniques. Cell-permeable Tat-STAT-6 fusion proteins were administered intraperitoneally. Cells were isolated from bone marrow and from joints for the JNK assay, the DNA-binding assays (electrophoretic mobility shift assays), and for in vitro osteoclastogenesis. RESULTS Activation of NF-kappaB and JNK in vivo was increased in extracts of cells retrieved from the joints of arthritic mice. Cell-permeable, constitutively active STAT-6 (i.e., STAT-6-VT) was effective in blocking NF-kappaB and JNK activation in RANKL-treated osteoclast progenitors. More importantly, STAT-6-VT protein significantly inhibited the in vivo activation of NF-kappaB and JNK, attenuated osteoclast recruitment in the inflamed joints, and decreased bone destruction. CONCLUSION Our findings indicate that the administration of STAT-6-VT presents a novel approach to the alleviation of bone erosion in inflammatory arthritis.
Collapse
Affiliation(s)
- Teruhisa Hirayama
- Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|
60
|
Kang JL, Lee HW, Kim HJ, Lee HS, Castranova V, Lim CM, Koh Y. Inhibition of SRC tyrosine kinases suppresses activation of nuclear factor-kappaB, and serine and tyrosine phosphorylation of IkappaB-alpha in lipopolysaccharide-stimulated raw 264.7 macrophages. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2005; 68:1643-62. [PMID: 16195219 DOI: 10.1080/15287390500192114] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Involvement of protein tyrosine kinases (PTK) in lipopolysaccharide (LPS)-induced nuclear factor-kappa B (NF-kappaB) activation has been demonstrated. Studies investigated the role of PTK and the underlying mechanisms by which PTK play a role in LPS induction of pathways leading to NF-kappaB activation in macrophages. Inhibitors of PTK-genistein, herbimycin A, or AG126-blocked LPS-induced NF-kappaB activation. Genistein also blocked pervanadate-induced NF-kappaB activation. Furthermore, Src TK selective inhibitors-damnacanthal or PP1-blocked LPS-induced NF-kappaB activation over a range of nanomolar concentrations. Genistein, damnacanthal, or PP1 blocked the LPS-induced serine phosphorylation, the degradation of IkappaB-alpha, and the consequent translocation of the p65 subunit of NF-kappaB to the nucleus. In addition to serine phosphorylation of IkappaB-alpha, LPS-induced NF-kappaB activation also required tyrosine phosphorylation of IkappaB-alpha. These TK inhibitors blocked substantially LPS induction of tyrosine phosphorylation of IkappaB-alpha. Furthermore, cSrc and Lck were physically associated with IkappaB-alpha. These results suggest that the LPS-induced NF-kappaB pathways are dependent on both serine and tyrosine phosphorylation of IkappaB-alpha, and that Src TK, such as cSrc and Lck, are key components of the LPS signaling pathway through at least two different mechanisms associated with NF-kappaB activation.
Collapse
Affiliation(s)
- Jihee Lee Kang
- Department of Physiology, College of Medicine, Division of Cell Biology, Ewha Medical Research Center, Ewha Womans University, Seoul, Korea.
| | | | | | | | | | | | | |
Collapse
|
61
|
Brown M, McGuinness M, Wright T, Ren X, Wang Y, Boivin GP, Hahn H, Feldman AM, Jones WK. Cardiac-specific blockade of NF-κB in cardiac pathophysiology: differences between acute and chronic stimuli in vivo. Am J Physiol Heart Circ Physiol 2005; 289:H466-76. [PMID: 15695559 DOI: 10.1152/ajpheart.00170.2004] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of NF-κB in cardiac physiology and pathophysiology has been difficult to delineate due to the inability to specifically block NF-κB signaling in the heart. Cardiac-specific transgenic models have recently been developed that repress NF-κB activation by preventing phosphorylation at specific serine residues of the inhibitory κB (IκB) protein isoform IκBα. However, these models are unable to completely block NF-κB because of a second signaling pathway that regulates NF-κB function via Tyr42 phosphorylation of IκBα. We report the development of transgenic (3M) mouse lines that express the mutant IκBα(S32A,S36A,Y42F)in a cardiac-specific manner. NF-κB activation in cardiomyopathic TNF-1.6 mice is completely blocked by the 3M transgene but only partially blocked (70–80%) by the previously described double-mutant 2M [IκBα(S32A,S36A)] transgene, which demonstrates the action of two proximal pathways for NF-κB activation in TNF-α-induced cardiomyopathy. In contrast, after acute stimuli including administration of TNF-α and ischemia-reperfusion (I/R), NF-κB activation is blocked in both 2M and 3M transgenic mice. This result suggests that phosphorylation of the regulatory Ser32 and Ser36 predominantly mediates NF-κB activation in these situations. We show that infarct size after I/R is reduced by 70% in 3M transgenic mice, which conclusively demonstrates that NF-κB is involved in I/R injury. In summary, we have engineered novel transgenic mice that allow us to distinguish two major proximal pathways for NF-κB activation. Our results demonstrate that the serine and tyrosine phosphorylation pathways are differentially activated during different pathophysiological processes (cardiomyopathy and I/R injury) and that NF-κB contributes to infarct development after I/R.
Collapse
Affiliation(s)
- Maria Brown
- Dept. of Pharmacology and Cell Biophysics, 231 Albert Sabin Way, ML0575, Univ. of Cincinnati, Cincinnati, OH 45267-0575, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Peng Y, Power MR, Li B, Lin TJ. Inhibition of IKK down-regulates antigen + IgE-induced TNF production by mast cells: a role for the IKK-IκB-NF-κB pathway in IgE-dependent mast cell activation. J Leukoc Biol 2005; 77:975-83. [PMID: 15784689 DOI: 10.1189/jlb.0204115] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Mast cells (MC) are major effector cells for allergic diseases. Cross-linking of immunoglobulin E (IgE) and its high-affinity receptor, FcepsilonRI, by antigen initiates a cascade of signaling events leading to nuclear factor (NF)-kappaB activation and tumor necrosis factor (TNF) production. Here, we demonstrated that inhibition of inhibitor of kappaB (IkappaB) kinase (IKK) by a peptide IKK inhibitor or by four individual chemical IKK inhibitors including 15-deoxy-prostaglandin J(2), BMS-345541, SC-514, or sulindac significantly blocked IgE + trinitrophenyl (TNP)-induced TNF production by mouse bone marrow-derived MC (BMMC). Moreover, IgE + TNP induced a rapid phosphorylation of IKKalpha but not IKKbeta in BMMC. IgE + TNP-induced phosphorylation of IKKalpha was accompanied with phosphorylation and degradation of IkappaBalpha, subsequent NF-kappaB activation, and TNF production. Inhibition of IKK by sulindac decreased IKKalpha phosphorylation, IkappaBalpha phosphorylation and degradation, NF-kappaB activation, and TNF production by BMMC. It is interesting that IgE + TNP stimulation also induced a prominent synthesis of IKKalpha and IkappaBalpha. Inhibition of NF-kappaB activity by pyrrolidine dithiocarbomate (PDTC) blocked IgE + TNP-induced IkappaBalpha synthesis. NF-kappaB activity and TNF production were also inhibited when PDTC was used even after IgE + TNP stimulation, suggesting a potential role for the newly synthesized IkappaBalpha in MC activation. In addition, IgE + TNP-induced IKKalpha and IkappaBalpha phosphorylation was inhibited by a protein kinase C (PKC) inhibitor Ro 31-8220. Taken together, our results support a role for the IKK-IkappaB-NF-kappaB pathway, which likely involves PKC in IgE-dependent TNF production by MC. Thus, IKK may serve as a new target for the regulation of MC function in allergy.
Collapse
Affiliation(s)
- Yongde Peng
- Department of Microbiology and Immunology, Dalhousie University, Isaac Walton Killam Health Centre, Halifax, Nova Scotia, Canada
| | | | | | | |
Collapse
|
63
|
Kabe Y, Ando K, Hirao S, Yoshida M, Handa H. Redox regulation of NF-kappaB activation: distinct redox regulation between the cytoplasm and the nucleus. Antioxid Redox Signal 2005; 7:395-403. [PMID: 15706086 DOI: 10.1089/ars.2005.7.395] [Citation(s) in RCA: 430] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Reduction/oxidation (redox) regulation mediates numerous cellular responses and contributes to several physiological diseases. The transcription factor nuclear factor kappaB (NF-kappaB) is known to be a redox-sensitive factor. NF-kappaB plays a central role in immune responses and inflammation, through regulation of the gene expression of a large number of cytokines and other immune response genes. NF-kappaB is trapped in the cytoplasm in stimulated cells and translocates into the nucleus in response to several stimuli, including oxidative stress. Reactive oxygen species enhance the signal transduction pathways for NF-kappaB activation in the cytoplasm and translocation into the nucleus. In contrast, the DNA binding activity of oxidized NF-kappaB is significantly diminished, and that activity is restored by reducing enzymes, such as thioredoxin or redox factor 1. This review describes the signal transduction pathways for NF-kappaB activation and redox regulation of NF-kB activation in the cytoplasm and nucleus.
Collapse
Affiliation(s)
- Yasuaki Kabe
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | | | | | | | | |
Collapse
|
64
|
van Vliet C, Bukczynska PE, Puryer MA, Sadek CM, Shields BJ, Tremblay ML, Tiganis T. Selective regulation of tumor necrosis factor-induced Erk signaling by Src family kinases and the T cell protein tyrosine phosphatase. Nat Immunol 2005; 6:253-60. [PMID: 15696169 DOI: 10.1038/ni1169] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2004] [Accepted: 01/18/2005] [Indexed: 12/27/2022]
Abstract
The proinflammatory cytokine tumor necrosis factor (TNF) modulates cellular responses through the mitogen-activated protein kinase (MAPK) and nuclear factor-kappaB (NF-kappaB) signaling pathways, but the molecular mechanisms underlying MAPK activation are unknown. T cell protein tyrosine phosphatase (TCPTP) is essential for hematopoietic development and negatively regulates inflammatory responses. Using TCPTP-deficient fibroblasts, we show here that TCPTP regulates TNF-induced MAPK but not NF-kappaB signaling. TCPTP interacted with the adaptor protein TRAF2, and dephosphorylated and inactivated Src tyrosine kinases to suppress downstream signaling through extracellular signal-regulated kinases and production of interleukin 6. These results link TCPTP and Src tyrosine kinases to the selective regulation of TNF-induced MAPK signaling and identify a previously unknown mechanism for modulating inflammatory responses mediated by TNF.
Collapse
Affiliation(s)
- Catherine van Vliet
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | | | | | | | | | | | | |
Collapse
|
65
|
Haefner B. The transcription factor NF-kappaB as drug target. PROGRESS IN MEDICINAL CHEMISTRY 2005; 43:137-88. [PMID: 15850825 DOI: 10.1016/s0079-6468(05)43005-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Affiliation(s)
- Burkhard Haefner
- Department of Inflammation, Johnson & Johnson Pharmaceutical Research and Development, A Division of Janssen Pharmaceutica, Beerse, Belgium
| |
Collapse
|
66
|
Amoui M, Suhr SM, Baylink DJ, Lau KHW. An osteoclastic protein-tyrosine phosphatase may play a role in differentiation and activity of human monocytic U-937 cell-derived, osteoclast-like cells. Am J Physiol Cell Physiol 2004; 287:C874-84. [PMID: 15355856 DOI: 10.1152/ajpcell.00294.2003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study investigated if an osteoclastic protein-tyrosine phosphatase (PTP), PTP-oc, plays a role in the functional activity and differentiation of osteoclastic cells by determining the effects of overexpression of wild-type (WT)- or phosphatase-deficient (PD)-PTP-oc on bone resorption activity and differentiation of human promyelomonocytic U-937 cells, which could be induced to differentiate into "osteoclast-like" cells by phorbol ester/1,25(OH)(2)D(3) treatment. U-937 cells overexpressing WT- or PD-PTP-oc were produced with a transposon-based vector. The size and depth of resorption pits created by WT-PTP-oc-overexpressing osteoclast-like cells were greater, while those by PD-PTP-oc-overexpressing osteoclast-like cells were less, than those created by control osteoclast-like cells. Overexpression of WT-PTP-oc also enhanced, while overexpression of PD-PTP-oc suppressed, their differentiation into osteoclast-like cells. Overexpression of WT-PTP-oc increased apoptosis and proliferation of U-937 cells, and overexpression of PD-PTP-oc reduced cell proliferation. Cells overexpressing WT-PTP-oc has also led to greater c-Src and NF-kappabeta activation, whereas cells overexpressing PD-PTP-oc resulted in less c-Src and NF-kappabeta activation. c-Src activation and NF-kappabeta activation each correlated with resorption activity and differentiation into osteoclast-like cells. In summary, these results show that 1) PTP-oc regulates both the activity and the differentiation of osteoclast-like cells derived from U-937 cells; 2) PTP-oc enzymatic activity is important to these processes; 3) high PTP-oc enzymatic activity caused an increase in U-937 cell apoptosis and proliferation, leading to no significant changes in the number of viable cells; and 4) some of the PTP-oc actions are mediated in part by the c-Src and/or NF-kappabeta pathways.
Collapse
Affiliation(s)
- Mehran Amoui
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, 11201 Benton St., Loma Linda, CA 92357, USA
| | | | | | | |
Collapse
|
67
|
Satoh A, Gukovskaya AS, Nieto JM, Cheng JH, Gukovsky I, Reeve JR, Shimosegawa T, Pandol SJ. PKC-delta and -epsilon regulate NF-kappaB activation induced by cholecystokinin and TNF-alpha in pancreatic acinar cells. Am J Physiol Gastrointest Liver Physiol 2004; 287:G582-91. [PMID: 15117677 DOI: 10.1152/ajpgi.00087.2004] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although NF-kappaB plays an important role in pancreatitis, mechanisms underlying its activation remain unclear. We investigated the signaling pathways mediating NF-kappaB activation in pancreatic acinar cells induced by high-dose cholecystokinin-8 (CCK-8), which causes pancreatitis in rodent models, and TNF-alpha, which contributes to inflammatory responses of pancreatitis, especially the role of PKC isoforms. We determined subcellular distribution and kinase activities of PKC isoforms and NF-kappaB activation in dispersed rat pancreatic acini. We applied isoform-specific, cell-permeable peptide inhibitors to assess the role of individual PKC isoforms in NF-kappaB activation. Both CCK-8 and TNF-alpha activated the novel isoforms PKC-delta and -epsilon and the atypical isoform PKC-zeta but not the conventional isoform PKC-alpha. Inhibition of the novel PKC isoforms but not the conventional or the atypical isoform resulted in the prevention of NF-kappaB activation induced by CCK-8 and TNF-alpha. NF-kappaB activation by CCK-8 and TNF-alpha required translocation but not tyrosine phosphorylation of PKC-delta. Activation of PKC-delta, PKC-epsilon, and NF-kappaB with CCK-8 involved both phosphatidylinositol-specific PLC and phosphatidylcholine (PC)-specific PLC, whereas with TNF-alpha they only required PC-specific PLC for activation. Results indicate that CCK-8 and TNF-alpha initiate NF-kappaB activation by different PLC pathways that converge at the novel PKCs (delta and epsilon) to mediate NF-kappaB activation in pancreatic acinar cells. These findings suggest a key role for the novel PKCs in pancreatitis.
Collapse
Affiliation(s)
- Akihiko Satoh
- Research Center for Alcoholic Liver and Pancreatic Diseases, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California 90073, USA
| | | | | | | | | | | | | | | |
Collapse
|
68
|
Takada Y, Aggarwal BB. TNF activates Syk protein tyrosine kinase leading to TNF-induced MAPK activation, NF-kappaB activation, and apoptosis. THE JOURNAL OF IMMUNOLOGY 2004; 173:1066-77. [PMID: 15240695 DOI: 10.4049/jimmunol.173.2.1066] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Spleen tyrosine kinase (Syk), a nonreceptor protein kinase initially found to be expressed only in hemopoietic cells, has now been shown to be expressed in nonhemopoietic cells and to mediate signaling of various cytokines. Whether Syk plays any role in TNF signaling was investigated. Treatment of Jurkat T cells with TNF activated Syk kinase but not ZAP70, another member of Syk kinase family, and the optimum activation occurred at 10 s and with 1 nM TNF. TNF also activated Syk in myeloid and epithelial cells. TNF-induced Syk activation was abolished by piceatannol (Syk-selective inhibitor), which led to the suppression of TNF-induced activation of c- JNK, p38 MAPK, and p44/p42 MAPK. Jurkat cells that did not express Syk (JCaM1, JCaM1/lck) showed lack of TNF-induced Syk, JNK, p38 MAPK, and p44/p42 MAPK activation, as well as TNF-induced IkappaBalpha phosphorylation, IkappaBalpha degradation, and NF-kappaB activation. TNF-induced NF-kappaB activation was enhanced by overexpression of Syk by Syk-cDNA and suppressed when Syk expression was down-regulated by expression of Syk-small interfering RNA (siRNA-Syk). The apoptotic effects of TNF were reduced by up-regulation of NF-kappaB by Syk-cDNA, and enhanced by down-regulation of NF-kappaB by siRNA-Syk. Immunoprecipitation of cells with Syk Abs showed TNF-dependent association of Syk with both TNFR1 and TNFR2; this association was enhanced by up-regulation of Syk expression with Syk-cDNA and suppressed by down-regulation of Syk using siRNA-Syk. Overall, our results demonstrate that Syk activation plays an essential role in TNF-induced activation of JNK, p38 MAPK, p44/p42 MAPK, NF-kappaB, and apoptosis.
Collapse
Affiliation(s)
- Yasunari Takada
- Cytokine Research Laboratory, Department of Bioimmunotherapy, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | |
Collapse
|
69
|
Dai S, Hirayama T, Abbas S, Abu-Amer Y. The IkappaB kinase (IKK) inhibitor, NEMO-binding domain peptide, blocks osteoclastogenesis and bone erosion in inflammatory arthritis. J Biol Chem 2004; 279:37219-22. [PMID: 15252035 DOI: 10.1074/jbc.c400258200] [Citation(s) in RCA: 162] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of NF-kappaB leads to expression of ample genes that regulate inflammatory and osteoclastogenic responses. The process is facilitated by induction of IkappaB kinase (IKK) complex that phosphorylates IkappaB and leads to its dissociation from the NF-kappaB complex, thus permitting activation of NF-kappaB. The IKK complex contains primarily IKKalpha, IKKbeta, and the regulatory kinase IKKgamma, also known as NEMO. NEMO regulates the IKK complex activity through its binding to carboxyl-terminal region of IKKalpha and IKKbeta, termed NEMO-binding domain (NBD). In this regard, a cell-permeable NBD peptide has been shown to block association of NEMO with the IKK complex and inhibit activation of NF-kappaB. Given the pivotal role of cytokine-induced NF-kappaB in osteoclastogenesis and inflammatory bone loss, we deduced that cell-permeable TAT-NBD peptide may hinder osteoclastogenesis and bone erosion in inflammatory arthritis. Using NBD peptides, we show that wild type, but not mutant, NBD blocks IKK activation and reduces cytokine-induced promoter and DNA binding activities of NF-kappaB and inhibits cytokine-induced osteoclast formation by osteoclast precursors. Consistent with the key role of NF-kappaB in osteoinflammatory responses in vivo, wild type TAT-NBD administered into mice prior to induction of inflammatory arthritis efficiently block in vivo osteoclastogenesis, inhibits focal bone erosion, and ameliorates inflammatory responses in the joints of arthritic mice. The mutant NBD peptide fails to exert these functions. These results provide strong evidence that IKKs are potent regulators of cytokine-induced osteoclastogenesis and inflammatory arthritis. More importantly, blockade of NEMO assembly with the IKK complex is a viable strategy to avert inflammatory osteolysis.
Collapse
Affiliation(s)
- Simon Dai
- Department of Orthopaedics and Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
70
|
Park HB, Golubovskaya V, Xu L, Yang X, Lee JW, Scully S, Craven RJ, Cance WG. Activated Src increases adhesion, survival and alpha2-integrin expression in human breast cancer cells. Biochem J 2004; 378:559-67. [PMID: 14629195 PMCID: PMC1223979 DOI: 10.1042/bj20031392] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2003] [Revised: 11/14/2003] [Accepted: 11/20/2003] [Indexed: 11/17/2022]
Abstract
Focal adhesion kinase (FAK) is an intracellular kinase that localizes to focal adhesions. FAK is overexpressed in human tumours, and FAK regulates both cellular adhesion and anti-apoptotic survival signalling. Disruption of FAK function by overexpression of the FAK C-terminal domain [FAK-CD, analogous to the FRNK (FAK-related non-kinase) protein] leads to loss of adhesion and apoptosis in tumour cells. We have shown that overexpression of an activated form of the Src tyrosine kinase suppressed the loss of adhesion induced by dominant-negative; adenoviral FAK-CD and decreased the apoptotic response in BT474 and MCF-7 breast cancer cell lines. This adhesion-dependent apoptosis was increased by the Src-family kinase inhibitor PP2 [4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine]. We have also shown that expression of activated Src in breast cancer cells increased the expression of alpha2-integrin and that overexpression of alpha2-integrin suppressed FAK-CD-mediated loss of adhesion. Our results suggest a model in which Src regulates adhesion and survival through enhanced expression of the alpha2-integrin. This provides a mechanism through which Src promotes cellular adhesion and alters the adhesive function of FAK.
Collapse
Affiliation(s)
- Hee Boong Park
- Department of Surgery, Ajou University School of Medicine, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
71
|
Fan C, Li Q, Zhang Y, Liu X, Luo M, Abbott D, Zhou W, Engelhardt JF. IkappaBalpha and IkappaBbeta possess injury context-specific functions that uniquely influence hepatic NF-kappaB induction and inflammation. J Clin Invest 2004; 113:746-55. [PMID: 14991073 PMCID: PMC351311 DOI: 10.1172/jci17337] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2002] [Accepted: 01/06/2004] [Indexed: 11/17/2022] Open
Abstract
IkappaB proteins play an important role in regulating NF-kappaB induction following a diverse range of environmental injuries. Studies evaluating IkappaBbeta knock-in mice (AKBI), in which the IkappaBalpha gene is replaced by the IkappaBbeta cDNA, have uncovered divergent properties of IkappaBalpha and IkappaBbeta that influence their ability to activate hepatic NF-kappaB and subsequent downstream proinflammatory processes in a stimulus-specific manner. While AKBI mice demonstrated identical levels of hepatic NF-kappaB activation in response to endotoxin, a significantly reduced level of hepatic NF-kappaB activation was observed in AKBI mice after liver ischemia/reperfusion (I/R) injury. This reduced level of NF-kappaB activation in AKBI mice after liver I/R also correlated with decreased induction of serum TNF-alpha, reduced hepatic inflammation, and increased survival. In contrast, no differences in any of these indicators were observed between AKBI mice and WT littermates after a lethal injection of LPS. Molecular studies suggest that the specificity of IkappaBalpha, but not IkappaBbeta, to properly regulate NF-kappaB induction during the acute phase of I/R injury is due to injury context-specific activation of c-Src and subsequent tyrosine phosphorylation of IkappaBalpha on Tyr42. These results demonstrate that IkappaBalpha and IkappaBbeta play unique injury context-specific roles in activating NF-kappaB-mediated proinflammatory responses and suggest that strategies aimed at inhibiting IkappaBalpha gene expression may be of potential therapeutic benefit in hepatic I/R injury.
Collapse
Affiliation(s)
- Chenguang Fan
- Molecular Biology Program, College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Clohisy JC, Roy BC, Biondo C, Frazier E, Willis D, Teitelbaum SL, Abu-Amer Y. Direct inhibition of NF-kappa B blocks bone erosion associated with inflammatory arthritis. THE JOURNAL OF IMMUNOLOGY 2004; 171:5547-53. [PMID: 14607962 DOI: 10.4049/jimmunol.171.10.5547] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Inflammatory arthritis is associated with devastating joint tissue destruction and periarticular bone erosion. Although secreted products of infiltrating immune cells perpetuate the inflammatory response, the osteolytic component of this disease is a direct result of localized recruitment and activation of osteoclasts. Given that NF-kappaB plays a central role in both processes, the function of this transcription factor was examined. Using a mouse model of autoreactive Ig transfer that engenders inflammatory arthritis, we show numerous osteoclasts in the articular joint tissue associated with progressive periarticular osteolytic lesions. Moreover, cells retrieved from these joints exhibit heightened NF-kappaB activity. Importantly, direct administration of dominant negative*I-kappaB or tyrosine 42-mutated I-kappaB (Y42F*I-kappaB) proteins into mice before induction of the disease attenuates in vivo activation of the transcription factor. More importantly, these I-kappaB mutant forms significantly inhibit in vivo production of TNF and receptor activator of NF-kappaB ligand, and block joint swelling, osteoclast recruitment, and osteolysis. Thus, NF-kappaB appears to be the centerpiece of inflammatory-osteolytic arthritis and direct inhibition of this transcription factor by unique and novel I-kappaB mutant proteins blocks manifestation of the disease.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- Arthritis, Experimental/prevention & control
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Arthritis, Rheumatoid/prevention & control
- Carrier Proteins/blood
- Carrier Proteins/metabolism
- Cell Movement/genetics
- Cell Movement/immunology
- Hindlimb
- I-kappa B Proteins/administration & dosage
- I-kappa B Proteins/genetics
- I-kappa B Proteins/pharmacology
- Immunization, Passive
- Immunoglobulins/administration & dosage
- Immunoglobulins/blood
- Inflammation Mediators/administration & dosage
- Inflammation Mediators/pharmacology
- Injections, Intraperitoneal
- Membrane Glycoproteins/blood
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred NOD
- Mice, Transgenic
- Mutagenesis, Site-Directed
- NF-kappa B/antagonists & inhibitors
- NF-kappa B/blood
- NF-kappa B/genetics
- NF-kappa B/physiology
- Osteoclasts/pathology
- Osteolysis/metabolism
- Osteolysis/pathology
- Osteolysis/prevention & control
- RANK Ligand
- Receptor Activator of Nuclear Factor-kappa B
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- John C Clohisy
- Department of Orthopaedic Research Laboratory, Barnes-Jewish Hospital at Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | |
Collapse
|
73
|
Clohisy JC, Hirayama T, Frazier E, Han SK, Abu-Amer Y. NF-kB signaling blockade abolishes implant particle-induced osteoclastogenesis. J Orthop Res 2004; 22:13-20. [PMID: 14656654 DOI: 10.1016/s0736-0266(03)00156-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In this study we investigated the effect of NF-kB signaling blockade on polymethylmethacrylate (PMMA) particle-induced osteoclastogenesis in vitro. We first established effective blockade of NF-kB activity as tested by electrophoretic mobility shift assays (EMSA). Particle-induced NF-kB activation in murine osteoclast precursor cells (CSF-1-dependent bone marrow macrophages) was markedly reduced by co-treatment of the cells with the NF-kB inhibitors N-tosyl-L-phenylalanine chloromethyl ketone (TPCK) and Calpain Inhibitor I (CPI). This inhibition of NF-kB activity was associated with blockade of p50 NF-kB subunit nuclear translocation. We then established a direct NF-kB inhibition approach by utilizing a TAT-bound, mutant IkB (TAT:IkB(46-317)), and demonstrated an inhibitory effect evidenced by decreased NF-kB DNA binding activity. Having established that these strategies (TPCK, CPI, TAT: IkB(46-317)) effectively block NF-kB activation, we next investigated the effect of these agents on particle-stimulated osteoclast formation. PMMA particle stimulation of mature osteoclast formation from RANKL-primed osteoclast precursor cells was blocked by all three inhibitors. To further test the efficacy of NF-kB blockade, experiments were performed with the TAT:IkB(46-317) mutant peptide in whole bone marrow cultures that contain supporting stromal cells. Again, this inhibitor efficiently blocked particle-induced osteoclastogenesis. Thus, we have shown that pharmaceutical and molecular blockade of NF-kB activation inhibits PMMA particle-directed osteoclastogenesis in vitro.
Collapse
Affiliation(s)
- John C Clohisy
- Department of Orthopaedic Surgery, Barnes-Jewish Hospital at Washington University School of Medicine, One Barnes-Jewish Hospital Plaza, Suite 11300, West Pavillion, St. Louis, MO 63110, USA.
| | | | | | | | | |
Collapse
|
74
|
Mahabeleshwar GH, Kundu GC. Tyrosine Kinase p56 Regulates Cell Motility and Nuclear Factor κB-mediated Secretion of Urokinase Type Plasminogen Activator through Tyrosine Phosphorylation of IκBα following Hypoxia/Reoxygenation. J Biol Chem 2003; 278:52598-612. [PMID: 14534291 DOI: 10.1074/jbc.m308941200] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nuclear factor kappaB (NFkappaB) plays major role in regulating cellular responses as a result of environmental injuries. The molecular mechanism(s) by which hypoxia/reoxygenation (H/R) regulates p56lck-dependent activation of NFkappaB through tyrosine phosphorylation of IkappaBalpha and modulates the expression of downstream genes that are involved in cell migration in human breast cancer cells are not well defined. In this paper, we investigated the involvement of protein-tyrosine kinase p56lck in the redox-regulated activation of NFkappaB following H/R in highly invasive (MDA-MB-231) and low invasive (MCF-7) breast cancer cells. We demonstrated that H/R induces tyrosine phosphorylation of p56lck, nuclear translocation of NFkappaB, NFkappaB-DNA binding, and transactivation of NFkappaB through tyrosine phosphorylation of IkappaBalpha. Transfection of these cells with wild type Lck but not with mutant Lck F394 followed by H/R induces the tyrosine phosphorylation of inhibitor of nuclear factor kappaB (IkappaBalpha) and transcriptional activation of NFkappaB, and these are inhibited by Lck inhibitors. In vitro kinase assay demonstrated that immunoprecipitated p56lck but not Lyn or Fyn directly phosphorylate IkappaBalpha in presence of H/R. Pervanadate, H2O2, and H/R induce the interaction between Lck and tyrosine-phosphorylated IkappaBalpha, and this interaction is inhibited by Src homology 2 domain inhibitory peptide, suggesting that tyrosine-phosphorylated IkappaBalpha interacts with Src homology 2 domain of Lck. Luciferase reporter gene assay indicated that Lck induces NFkappaB-dependent urokinase type plasminogen activator (uPA) promoter activity in presence of H/R. Furthermore, H/R stimulates the cell motility through secretion of uPA. To our knowledge, this is the first report that p56lck in presence of H/R regulates NFkappaB activation, uPA secretion, and cell motility through tyrosine phosphorylation of IkappaBalpha and further demonstrates an important redox-regulated pathway for NFkappaB activation following H/R injury that is independent of IkappaB kinase/IkappaBalpha-mediated signaling pathways.
Collapse
|
75
|
Abstract
Inflammatory osteolysis induced by implant-derived wear debris is associated with infiltration of various cell-types to the implant-bone interface leading to abundant secretion of pro-inflammatory cytokines and activation of proteinases that together lead to propagation of the localized inflammatory response and periprosthetic bone erosion. Tumor necrosis factor family members are considered to be direct mediators of inflammation and osteolysis. These cytokines exert their osteoclastic effects via activation of the transcription factor NF-kappaB and certain MAP kinases, including c-Jun, Erks and p38, all known to be essential for the development of osteoclasts. We have recently documented that the osteoclastogenic cytokines TNF and RANKL play a pivotal role in the development of inflammatory osteolysis. We have also found that PMMA particles stimulate osteoclastogenesis, at least in part, by induction of RANKL, TNF, and by activation of the transcription factor NF-kappaB. More importantly, our data indicate that inhibitors of the osteoclastogenic factors, TNF and RANKL abrogate particle-induced osteoclastogenesis. In the current study, we investigated if PMMA particles activate MAP kinases, and the potential role of these kinases as mediators of osteolysis. Using kinase assays, we show that in osteoclast precursors, PMMA particles markedly and rapidly activate p38 and ERK MAP kinases. This activation was specific, evident by complete blockade with specific inhibitory compounds. Similarly, we show that PMMA particles activate the JNK pathway, which is known to be involved in inflammatory and osteoclastogenic events. We also show that p38 MAP kinase regulates PMMA-activation of NF-kappaB, thus providing a possible mechanism for particle action in osteoclast precursors. Finally, we provide evidence that specific inhibitors of MAP kinases are capable of inhibiting PMMA-stimulated osteoclastogenesis. These data provide evidence that MAP kinases are potent mediators of particle-induced osteoclastogenesis.
Collapse
Affiliation(s)
- S Abbas
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
76
|
Wu XB, Li Y, Schneider A, Yu W, Rajendren G, Iqbal J, Yamamoto M, Alam M, Brunet LJ, Blair HC, Zaidi M, Abe E. Impaired osteoblastic differentiation, reduced bone formation, and severe osteoporosis in noggin-overexpressing mice. J Clin Invest 2003; 112:924-34. [PMID: 12975477 PMCID: PMC193662 DOI: 10.1172/jci15543] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
We describe the effects of the overexpression of noggin, a bone morphogenetic protein (BMP) inhibitor, on osteoblast differentiation and bone formation. Cells of the osteoblast and chondrocyte lineages, as well as bone marrow macrophages, showed intense beta-gal histo- or cytostaining in adult noggin+/- mice that had a LacZ transgene inserted at the site of noggin deletion. Despite identical BMP levels, however, osteoblasts of 20-month-old C57BL/6J and 4-month-old senescence-accelerated mice (SAM-P6 mice) had noggin expression levels that were approximately fourfold higher than those of 4-month-old C57BL/6J and SAM-R1 (control) mice, respectively. U-33 preosteoblastic cells overexpressing the noggin gene showed defective maturation and, in parallel, a decreased expression of Runx-2, bone sialoprotein, osteocalcin, and RANK-L. Noggin did not inhibit the ligandless signaling and pro-differentiation action of the constitutively activated BMP receptor type 1A, ca-ALK-3. Transgenic mice overexpressing noggin in mature osteocalcin-positive osteoblasts showed dramatic decreases in bone mineral density and bone formation rates with histological evidence of decreased trabecular bone and CFU-osteoblast colonies at 4 and 8 months. Together, the results provide compelling evidence that noggin, expressed in mature osteoblasts, inhibits osteoblast differentiation and bone formation. Thus, the overproduction of noggin during biological aging may result in impaired osteoblast formation and function and hence, net bone loss.
Collapse
Affiliation(s)
- Xue-Bin Wu
- Division of Endocrinology, Diabetes, and Bone Diseases, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1055, New York, New York 10029, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Klein BY, Rojansky N, Ben-Yehuda A, Abou-Atta I, Abedat S, Friedman G. Cell death in cultured human Saos2 osteoblasts exposed to low-density lipoprotein. J Cell Biochem 2003; 90:42-58. [PMID: 12938155 DOI: 10.1002/jcb.10603] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Osteoporosis (OP) and atherosclerotic-cardiovascular diseases (and possibly dementia) constitute emerging age-related co-morbidity states that might share risk factors. Blood-born lipids, like LDL involved in atherosclerosis and apolipoprotein-E4 (ApoE4) involved in dementia, may also be implicated in development of OP. We examined osteoblast cell lines as a culture model for OP by exposure to lipoproteins. ApoE expression in Saos2 and U2OS osteoblasts was confirmed by PCR. ApoE4 did decrease cell counts relatively to ApoE3, especially in Saos2 cells in which it was less selective for cells with higher alkaline phosphatase (ALP, an osteoblast marker) activity than ApoE3. This associates with ApoE4, being a risk factor for both dementia and OP. Saos2, but not U2OS, showed a decrease in cell counts after 48 h exposure to native LDL (NLDL). Both cell lines had decreased cell counts already after 24 h when exposed to oxidized-LDL (OxLDL) for which Saos2 also showed a higher sensitivity than U2OS. Exposure of Saos2 to both, OxLDL at low concentration (5 microg/ml) and NLDL revealed a shrunken size cell fraction of 17-23% on the fluorescence-activated cell sorter (FACS) analysis. Such shrunken cell fraction was not seen when Saos2 cells were exposed to 50 microg/ml of OxLDL or to OxLDL combined with 10 nM dexamethasone (DEX, a stimulator of osteoprogenitor differentiation). DEX treatment has lysed the cells earlier than 24 h post exposure and has selected more resistant cells that did not show apoptotic shrinkage in the FACS analysis done after 24 h. We interpret this as a failure to detect the apoptotic cell fraction due to their lysis prior to the FACS analysis. Western blots performed at different time points (10 min, 30 min, 4 h, 24 h, and 48 h) under OxLDL + DEX revealed a fall in the positive regulator of pp60Src-kinase phosphotyrosine (pY)418 relative to the DEX controls during the first 4 h. This is consistent with DEX osteogenic induction, known to be negatively regulated by c-Src, although the pY418/pY529 ratios (negative/positive kinase regulation) fell only at the 10 min time point. Contrarily the pY418/pY529 ratio increased, relative to untreated controls, under 5 microg/ml and 50 microg/ml of NLDL at the 4 h time point and under 50 microg/ml NLDL only at the 10 min time point, being consistent with the ability of a higher dose of LDL to antagonize osteoblast differentiation. This could be even more acceptable if the NLDL would have become minimally oxidized during its long purification procedure. Under NLDL, the Bcl-2/Bax ratio was pro-apoptotic at 10 min, 30 min, and 4 h only under 50 microg/ml, whereas under OxLDL + DEX it was pro-apoptotic only after 4 h suggesting that additional pathways contribute to cell death. These results indicate that lipid effects on human osteoblast lines in culture may be used as a model to identify molecular targets shared between OP and atherosclerosis for intervention in this co-morbidity.
Collapse
Affiliation(s)
- Benjamin Y Klein
- Laboratory of Experimental Surgery, Hadassah University Hospital, Jerusalem 91120, Israel.
| | | | | | | | | | | |
Collapse
|
78
|
De Bosscher K, Vanden Berghe W, Haegeman G. The interplay between the glucocorticoid receptor and nuclear factor-kappaB or activator protein-1: molecular mechanisms for gene repression. Endocr Rev 2003; 24:488-522. [PMID: 12920152 DOI: 10.1210/er.2002-0006] [Citation(s) in RCA: 629] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The inflammatory response is a highly regulated physiological process that is critically important for homeostasis. A precise physiological control of inflammation allows a timely reaction to invading pathogens or to other insults without causing overreaction liable to damage the host. The cellular signaling pathways identified as important regulators of inflammation are the signal transduction cascades mediated by the nuclear factor-kappaB and the activator protein-1, which can both be modulated by glucocorticoids. Their use in the clinic includes treatment of rheumatoid arthritis, asthma, allograft rejection, and allergic skin diseases. Although glucocorticoids have been widely used since the late 1940s, the molecular mechanisms responsible for their antiinflammatory activity are still under investigation. The various molecular pathways proposed so far are discussed in more detail.
Collapse
Affiliation(s)
- Karolien De Bosscher
- Department of Molecular Biology, Ghent University, K. L. Ledeganckstraat 35, 9000 Gent, Belgium
| | | | | |
Collapse
|
79
|
Khan E, Abu-Amer Y. Activation of peroxisome proliferator-activated receptor-gamma inhibits differentiation of preosteoblasts. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 2003; 142:29-34. [PMID: 12878983 DOI: 10.1016/s0022-2143(03)00058-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) is critical for phenotype determination at early differentiation stages of mesenchymal cells. Activation of this nuclear receptor inhibits gene expression in part by antagonizing the activities of several transcription factors. In this study we examined inhibitory mechanisms of osteoblast differentiation markers by activating PPAR-gamma. Our data indicate that the PPAR-gamma natural ligand 15d-PGJ2 dose-dependently inhibits expression of alkaline phosphatase and mineral deposition by primary stromal cells and by cell lines such as ST2 and MC3T3-E1. We next show that PPAR-gamma nuclear translocation coincides with duration and doses of ligand addition, indicating that 15d-PGJ2-activated PPAR-gamma rapidly translocates to the nuclear component where it exerts its biological effects. Further examination of downstream osteogenic signaling pathways induced by beta-glycerophosphate and ascorbic acid reveals that induction of osteoblast differentiation by these agents involves activation of the transcription factors Cbfa1 and NF-kappaB. The former is critical for osteoblast differentiation. To test whether inhibition of alkaline phosphatase expression and mineral deposition by activated PPAR-gamma reflects attenuation of transcriptional activity, we performed DNA protein-binding assays for NF-kappaB and Cbfa1. Our findings indicate that 15d-PGJ2-induced PPAR-gamma abrogates beta-glycerophosphate-activated Cbfa1 and NF-kappaB. These findings were consistent in primary and stromal cell lines, ST2 and MC3T3-E1. Thus activation of PPAR-gamma by 15d-PGJ2 inhibits DNA-binding activity of the transcription factors Cbfa1 and NF-kappaB, leading to diminished expression of osteoblast/stromal differentiation markers.
Collapse
Affiliation(s)
- Emma Khan
- Department of Orthopedic Research Laboratory, Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | |
Collapse
|
80
|
Abstract
Nuclear factor-kappaB (NF-kappaB) is a transcriptional factor that was originally discovered in the nucleus of B cells that bind to the kappa light chain of the immunoglobulins. Research during 15 years, however, has revealed that NF-kappaB is present in its inactive state in the cytoplasm of almost every cell type. When activated, NF-kappaB translocates to the nucleus, binds the DNA and regulates the expression of over 200 different genes. The product of these genes regulate the immune system, cell proliferation, tumor metastasis, inflammation and viral replication. Several tumor cell types express constitutively activated form of NF-kappaB and it is required for the proliferation of the tumor cells. Numerous studies have shown that Hodgkin's disease cells exhibit constitutive active NF-kappaB. The present review examines the mechanism how NF-kappaB is activated and its relevance to Hodgkin's disease.
Collapse
Affiliation(s)
- Anas Younes
- Department of Lymphoma/Myeloma, The University of Texas, M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 429 Houston, TX 77030, USA.
| | | | | |
Collapse
|
81
|
Abbas S, Abu-Amer Y. Dominant-negative IkappaB facilitates apoptosis of osteoclasts by tumor necrosis factor-alpha. J Biol Chem 2003; 278:20077-82. [PMID: 12637573 DOI: 10.1074/jbc.m208619200] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Osteoclasts are the sole bone-resorbing cells. Heightened activity of these cells under pathological conditions leads to the development of bone loss diseases, such as osteolysis, osteoporosis, and rheumatoid arthritis. We have shown previously that tumor necrosis factor alpha-(TNF) strongly induces osteoclastogenesis of preosteoclasts and do so through activation of the transcription factor, NF-kappaB. Most importantly, recent studies have shown that NF-kappaB is required for the development of osteoclasts. This transcription factor has also been proven as an essential mediator of inflammatory diseases including those related to bone. In this regard, we have shown that various mutated forms of IkappaBalpha are potent inhibitors of osteoclastogenesis. In this study, we examined the direct effect of DN-IkappaB on mature and preosteoclast development in the presence of TNF. Our findings indicate that once committed to the osteoclastogenic pathway, preosteoclasts form giant and hyperactive osteoclasts in response to TNF. However, administration of DN-IkappaB to cultures prior to TNF exposure averts the osteoclastogenic effect of TNF into apoptosis. Screening potential mediators of DN-IkappaB and TNF-induced apoptosis shows that caspase 3, caspase 9, poly(ADP-ribose)polymerase, and Bax are activated, whereas levels of Bcl-XL, cIAP-1, and TRAF6 were reduced. Taken together, these findings suggest that under conditions of NF-kappaB inactivity levels of pro-survival factors are diminished, which in turn facilitates TNF induction of pro-apoptotic factors leading to apoptosis.
Collapse
Affiliation(s)
- Sabiha Abbas
- Department of Orthopaedics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | |
Collapse
|
82
|
Gu Y, Xu YC, Wu RF, Nwariaku FE, Souza RF, Flores SC, Terada LS. p47phox participates in activation of RelA in endothelial cells. J Biol Chem 2003; 278:17210-7. [PMID: 12618429 DOI: 10.1074/jbc.m210314200] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Activation of endothelial cell NF-kappaB by interleukin (IL)-1 constitutes an event critical to the progression of the innate immune response. In this context, oxidants have been associated with NF-kappaB activation, although the molecular source and mechanism of targeting have remained obscure. We found that RelA, essential for NF-kappaB activation by IL-1, was associated with the NADPH oxidase adapter protein p47(phox) in yeast two-hybrid, coprecipitation, and in vitro binding studies. RelA and p47-GFP also colocalized in endothelial cells in focal submembranous dorsoventral protrusions. Overexpression of p47(phox) synergized with IL-1beta in the activation of an artificial kappaB-luciferase reporter and specifically augmented IL-1beta-induced RelA transactivation activity. p47(phox) overexpression also greatly increased IL-1beta-stimulated RelA phosphorylation, whereas it had no effect on I-kappaB degradation or on RelA nuclear translocation or kappaB binding. The tandem SH3 domains of p47(phox) were found to associate with a proline-rich mid-region of RelA (RelA-PR) located between the Rel homology and transactivation domains. The RelA-PR peptide blocked interaction of p47(phox) and RelA, and ectopic expression of RelA-PR abrogated IL-1beta-induced transactivation of the NF-kappaB-dependent E-selectin promoter. Further, suppression of NADPH oxidase function through the inhibitor diphenylene iodonium, the superoxide dismutase mimetic Mn(III) tetrakis(4-benzoic acid)porphyrin (MnTBAP), or expression of a dominant interfering mutant of a separate NADPH oxidase subunit (p67(V204A)) decreased IL-1beta-induced E-selectin promoter activation, suggesting that p47(phox) facilitates NF-kappaB activation through linkage with the NADPH oxidase. IL-1beta rapidly increased tyrosine phosphorylation of IL-1 type I receptor-associated proteins, suggesting that oxidants may operate through inactivation of local protein-tyrosine phosphatases in the proximal IL-1beta signaling pathway leading to RelA activation.
Collapse
Affiliation(s)
- Ying Gu
- Department of Internal Medicine, University of Texas Southwestern, Dallas, Texas 75216, USA
| | | | | | | | | | | | | |
Collapse
|
83
|
Huang WC, Chen JJ, Inoue H, Chen CC. Tyrosine phosphorylation of I-kappa B kinase alpha/beta by protein kinase C-dependent c-Src activation is involved in TNF-alpha-induced cyclooxygenase-2 expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4767-75. [PMID: 12707358 DOI: 10.4049/jimmunol.170.9.4767] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The signaling pathway involved in TNF-alpha-induced cyclooxygenase-2 (COX-2) expression was further studied in human NCI-H292 epithelial cells. A protein kinase C (PKC) inhibitor (staurosporine), tyrosine kinase inhibitors (genistein and herbimycin A), or a Src kinase inhibitor (PP2) attenuated TNF-alpha- or 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced COX-2 promoter activity. TNF-alpha- or TPA-induced I-kappaB kinase (IKK) activation was also blocked by these inhibitors, which reversed I-kappaBalpha degradation. Activation of c-Src and Lyn kinases, two Src family members, was inhibited by the PKC, tyrosine kinase, or Src kinase inhibitors. The dominant-negative c-Src (KM) mutant inhibited induction of COX-2 promoter activity by TNF-alpha or TPA. Overexpression of the constitutively active PKCalpha (PKCalpha A/E) or wild-type c-Src plasmids induced COX-2 promoter activity, and these effects were inhibited by the dominant-negative c-Src (KM), NF-kappaB-inducing kinase (NIK) (KA), or IKKbeta (KM) mutant. The dominant-negative PKCalpha (K/R) or c-Src (KM) mutant failed to block induction of COX-2 promoter activity caused by wild-type NIK overexpression. In coimmunoprecipitation experiments, IKKalpha/beta was found to be associated with c-Src and to be phosphorylated on its tyrosine residues after TNF-alpha or TPA treatment. Two tyrosine residues, Tyr(188) and Tyr(199), near the activation loop of IKKbeta, were identified to be crucial for NF-kappaB activation. Substitution of these residues with phenylalanines attenuated COX-2 promoter activity and c-Src-dependent phosphorylation of IKKbeta induced by TNF-alpha or TPA. These data suggest that, in addition to activating NIK, TNF-alpha also activates PKC-dependent c-Src. These two pathways cross-link between c-Src and NIK and converge at IKKalpha/beta, and go on to activate NF-kappaB, via serine phosphorylation and degradation of IkappaB-alpha, and, finally, to initiate COX-2 expression.
Collapse
Affiliation(s)
- Wei-Chien Huang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | |
Collapse
|
84
|
Abbas S, Zhang YH, Clohisy JC, Abu-Amer Y. Tumor necrosis factor-alpha inhibits pre-osteoblast differentiation through its type-1 receptor. Cytokine 2003; 22:33-41. [PMID: 12946103 DOI: 10.1016/s1043-4666(03)00106-6] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tumor necrosis factor-alpha (TNF) is a pro-inflammatory cytokine with a profound role in many skeletal diseases. The cytokine has been described as a mediator of bone loss in osteolysis and other inflammatory bone diseases. In addition to its known bone resorptive action, TNF reduces bone formation by inhibiting osteoblast differentiation. Using primary and transformed osteoblastic cells, we first document that TNF inhibits expression of alkaline phosphatase and matrix deposition, both considered markers of osteoblast differentiation. The effects are dose- and time-dependent. Core-binding factor A1 (cbfa1) is a transcription factor critical for osteoblast differentiation, and we show here that it is activated by the osteoblast differentiation agent, beta-glycerophosphate. Therefore, we investigated whether the inhibitory effects of TNF were associated with altered activity of this transcription factor. Using retardation assays, we show that TNF significantly inhibits cbfal activation by beta-glycerophosphate, manifested by reduced DNA-binding activity. Next, we turned to determine the signaling pathway by which TNF inhibits osteoblast differentiation. Utilizing animals lacking individual TNF receptors, we document that TNFr1 is required for transmitting the cytokine's inhibitory effect. In the absence of this receptor, TNF failed to impact all osteoblast differentiation markers tested. In summary, TNF blocks expression of osteoblast differentiation markers and inhibits beta-glycerophosphate-induced activation of the osteoblast differentiation factor cbfa1. Importantly, these effects are mediated via a mechanism requiring the TNF type-1 receptor.
Collapse
Affiliation(s)
- Sabiha Abbas
- Department of Orthopaedics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
85
|
Vanden Berghe W, De Bosscher K, Vermeulen L, De Wilde G, Haegeman G. Induction and repression of NF-kappa B-driven inflammatory genes. ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2003:233-78. [PMID: 12355719 DOI: 10.1007/978-3-662-04660-9_14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- W Vanden Berghe
- Unit of Eukaryotic Gene Expression and Signal Transduction, Department of Molecular Biology, University of Gent-VIB, K.L. Ledeganckstraat 35, 9000 Gent, Belgium.
| | | | | | | | | |
Collapse
|
86
|
Huang WC, Chen JJ, Chen CC. c-Src-dependent tyrosine phosphorylation of IKKbeta is involved in tumor necrosis factor-alpha-induced intercellular adhesion molecule-1 expression. J Biol Chem 2003; 278:9944-52. [PMID: 12645577 DOI: 10.1074/jbc.m208521200] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The signaling pathway involved in tumor necrosis factor-alpha (TNF-alpha)-induced intercellular adhesion molecule-1 (ICAM-1) expression was further studied in human A549 epithelial cells. TNF-alpha- or 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced ICAM-1 promoter activity was inhibited by a protein kinase C (PKC) inhibitor (staurosporine), tyrosine kinase inhibitors (genistein and herbimycin A), or an Src-specific tyrosine kinase inhibitor (PP2). TNF-alpha- or TPA-induced IkappaBalpha kinase (IKK) activation was also blocked by these inhibitors, which slightly reversed TNF-alpha-induced but completely reversed TPA-induced IkappaBalpha degradation. c-Src and Lyn, two members of the Src kinase family, were abundantly expressed in A549 cells, and their activation by TNF-alpha or TPA was inhibited by the same inhibitors. Furthermore, the dominant-negative c-Src (KM) mutant inhibited induction of ICAM-1 promoter activity by TNF-alpha or TPA. Overexpression of the constitutively active PKC or wild-type c-Src plasmids induced ICAM-1 promoter activity, this effect being inhibited by the dominant-negative c-Src (KM) or IKKbeta (KM) mutant but not by the nuclear factor-kappaB-inducing kinase (NIK) (KA) mutant. The c-Src (KM) mutant failed to block induction of ICAM-1 promoter activity caused by overexpression of wild-type NIK. In co-immunoprecipitation and immunoblot experiments, IKK was found to be associated with c-Src and to be phosphorylated on tyrosine residues after TNF-alpha or TPA treatment. Two tyrosine residues, Tyr188 and Tyr199, near the activation loop of IKKbeta, were identified as being important for NF-kappaB activation. Substitution of these residues with phenylalanines abolished ICAM-1 promoter activity and c-Src-dependent phosphorylation of IKKbeta induced by TNF-alpha or TPA. These data suggest that, in addition to activating NIK, TNF-alpha also activates PKC-dependent c-Src. These two pathways converge at IKKbeta and go on to activate NF-kappaB, via serine phosphorylation and degradation of IkappaB-alpha, and, finally, to initiate ICAM-1 expression.
Collapse
Affiliation(s)
- Wei-Chien Huang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10018, Taiwan
| | | | | |
Collapse
|
87
|
Huang S, Dudez T, Scerri I, Thomas MA, Giepmans BNG, Suter S, Chanson M. Defective activation of c-Src in cystic fibrosis airway epithelial cells results in loss of tumor necrosis factor-alpha-induced gap junction regulation. J Biol Chem 2003; 278:8326-32. [PMID: 12506110 DOI: 10.1074/jbc.m208264200] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF-alpha) signaling is central to the transmission of the innate immune response and subsequent activation of the adaptive immune system. The functioning of both systems is required for optimal clearance of pathogens from the airways. In cystic fibrosis (CF), dysfunction of the CF transmembrane conductance regulator (CFTR) is associated with recurrent pulmonary infections despite an intense inflammatory and immune response. We reported recently that TNF-alpha decreased gap junction connectivity in non-CF airway cells, a mechanism that was absent in CF cells expressing the DeltaPhe-508 mutant of CFTR. We have now identified the tyrosine kinase c-Src as a possible pathway between the mediators of inflammation and the gap junction protein connexin43 (Cx43). Indeed, TNF-alpha increased the proportion of activated c-Src in non-CF airway cells. Moreover, pharmacological antagonists and expression in non-CF cells of a dominant negative construct of c-Src prevented Cx43 channel closure by TNF-alpha. Finally, gap junction channel closure was prevented by expression of a Cx43 mutant lacking tyrosine phosphorylation sites for c-Src. Additional experiments showed that activation of c-Src was defective in CF airway cells but rescued in CFTR-corrected CF cells. These data suggest that CFTR dysfunction is associated with altered TNF-alpha signaling, resulting in the persistence of gap junction connectivity in CF airway cells. We propose that altered regulation of c-Src may contribute to the dysregulated inflammatory response that is characteristic of the CF phenotype.
Collapse
Affiliation(s)
- Song Huang
- Laboratory of Clinical Investigation III, Department of Pediatrics, Geneva University Hospitals, Switzerland
| | | | | | | | | | | | | |
Collapse
|
88
|
Clohisy JC, Frazier E, Hirayama T, Abu-Amer Y. RANKL is an essential cytokine mediator of polymethylmethacrylate particle-induced osteoclastogenesis. J Orthop Res 2003; 21:202-12. [PMID: 12568950 DOI: 10.1016/s0736-0266(02)00133-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
RANKL is a TNF superfamily member and an essential cytokine mediator of developmental osteoclastogenesis. We examined the role of RANKL in PMMA particle-induced osteoclastogenesis in vitro. In murine whole bone marrow cultures, PMMA particles stimulate a 2.5 fold increase in secreted RANKL, a 5-8 fold increase in osteoclast number and induce the formation of giant multinuclear osteoclasts. RANKL and TNF, potential cytokine mediators of PMMA, had similar osteoclastogenic effects. The RANKL inhibitor OPG was utilized to define the role of RANKL in mediating the PMMA response and was found to inhibit basal and PMMA particle-induced osteoclastogenesis. Additionally, particles stimulate osteoclast formation in RANKL-primed osteoclast precursor cells (devoid of supporting stromal cells) while RANKL untreated osteoclast precursors demonstrate no osteoclastogenic response to particles. Since TNF can potentiate RANKL action and is thought to mediate implant osteolysis we analyzed TNF(-/-) whole bone marrow cultures to elucidate the role of this cytokine. In TNF(-/-) cultures basal osteoclastogenesis remains intact, yet the PMMA effect is blunted. Finally, we show that PMMA, RANKL and TNF all activate the NF-kB and c-jun/AP-1 signaling pathways which are both fundamental to osteoclast formation and are potential sites of signal convergence in RANKL-mediated particle osteoclastogenesis.
Collapse
Affiliation(s)
- John C Clohisy
- Department of Orthopaedic Surgery, Barnes-Jewish Hospital at Washington University, School of Medicine, One Barnes-Jewish Hospital Plaza, 11300 West Pavilion, Campus Box 8233, St. Louis, Missouri 63110, USA.
| | | | | | | |
Collapse
|
89
|
Fan C, Li Q, Ross D, Engelhardt JF. Tyrosine phosphorylation of I kappa B alpha activates NF kappa B through a redox-regulated and c-Src-dependent mechanism following hypoxia/reoxygenation. J Biol Chem 2003; 278:2072-80. [PMID: 12429743 DOI: 10.1074/jbc.m206718200] [Citation(s) in RCA: 141] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
NF kappa B is a critical transcription factor involved in modulating cellular responses to environmental injuries. Tyrosine 42 phosphorylation of I kappa B alpha has been shown to mediate NF kappa B activation following hypoxia/reoxygenation (H/R) or pervanadate treatment. This pathway differs from the canonical proinflammatory pathways, which mediate NF kappa B activation through serine phosphorylation of I kappa B alpha by the IKK complex. In the present study, we investigated the involvement of c-Src in the redox activation of NFkappaB following H/R or pervanadate treatment. Our results demonstrate that pervanadate or H/R treatment leads to tyrosine phosphorylation of I kappa B alpha and NF kappa B transcriptional activation independent of the IKK pathway. In contrast, inhibition of c-Src by pp2 treatment or in c-Src (-/-) knockout cell lines, demonstrated a significant reduction in I kappa B alpha tyrosine phosphorylation and NF kappa B activation following pervanadate or H/R treatment. Overexpression of glutathione peroxidase-1 or catalase, but not Mn-SOD or Cu,Zn-SOD, significantly reduced both NF kappa B activation and tyrosine phosphorylation of I kappa B alpha. In vitro kinase assays further demonstrated that immunoprecipitated c-Src has the capacity to directly phosphorylate GST-I kappa B alpha and that this I kappa B alpha kinase activity is significantly reduced by Gpx-1 overexpression. These results suggest that c-Src-dependent tyrosine phosphorylation of I kappa B alpha and subsequent activation of NF kappa B is controlled by intracellular H(2)O(2) and defines an important redox-regulated pathway for NF kappa B activation following H/R injury that is independent of the IKK complex.
Collapse
Affiliation(s)
- Chenguang Fan
- Molecular Biology Graduate Program, the Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | |
Collapse
|
90
|
Fan C, Yang J, Engelhardt JF. Temporal pattern of NFkappaB activation influences apoptotic cell fate in a stimuli-dependent fashion. J Cell Sci 2002; 115:4843-53. [PMID: 12432072 DOI: 10.1242/jcs.00151] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The transcription factor NFkappaB is a critical immediate early response gene involved in modulating cellular responses and apoptosis following diverse environmental injuries. The activation of NFkappaB is widely accepted to play an anti-apoptotic role in cellular responses to injury. Hence, enhancing NFkappaB activation in the setting of injury has been proposed as one potential therapeutic approach to environmental injuries. To this end, we constructed a recombinant adenoviral vector (Ad.IkappaBalphaAS) expressing antisense IkappaBalpha mRNA that is capable of augmenting NFkappaB activation prior to and following four types of cellular injury [TNF-alpha, UV, hypoxia/reoxygenation (H/R) or pervanadate treatment]. Biochemical and functional analyses of NFkappaB activation pathways for these injuries demonstrated two categories involving either serine (S32/36) phosphorylation (TNF-alpha, UV) or tyrosine (Y42) phosphorylation (H/R or PV) of IkappaBalpha. We hypothesized that activation of NFkappaB prior to injury using antisense IkappaBalpha mRNA would reduce apoptosis. As anticipated, recombinant adenoviral IkappaBalpha phosphorylation mutants (Ad.IkappaBalphaS32/36A or Ad.IkappaBalphaY42F) preferentially reduced NFkappaB activation and enhanced apoptosis following injuries associated with either serine or tyrosine phosphorylation of IkappaBalpha, respectively. These studies demonstrate for the first time that an IkappaBalphaY42F mutant can effectively modulate NFkappaB-mediated apoptosis in an injury-context-dependent manner. Interestingly, constitutive activation of NFkappaB following Ad.IkappaBalphaAS infection reduced apoptosis only following injuries associated with IkappaBalpha Y42, but not S32/36, phosphorylation. These findings demonstrate that the temporal regulation of NFkappaB and the apoptotic consequences of this activation are differentially influenced by the pathway mediating NFkappaB activation. They also provide new insight into the therapeutic potential and limitations of modulating NFkappaB for environmental injuries such as ischemia/reperfusion and pro-inflammatory diseases.
Collapse
Affiliation(s)
- Chenguang Fan
- Molecular Biology Graduate Program, University of Iowa College of Medicine, Iowa City, Iowa, 52242 USA
| | | | | |
Collapse
|
91
|
Ashikawa K, Majumdar S, Banerjee S, Bharti AC, Shishodia S, Aggarwal BB. Piceatannol inhibits TNF-induced NF-kappaB activation and NF-kappaB-mediated gene expression through suppression of IkappaBalpha kinase and p65 phosphorylation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:6490-7. [PMID: 12444159 DOI: 10.4049/jimmunol.169.11.6490] [Citation(s) in RCA: 190] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Piceatannol is an anti-inflammatory, immunomodulatory, and anti-proliferative stilbene that has been shown to interfere with the cytokine signaling pathway. Previously, we have shown that resveratrol suppresses the activation of the nuclear transcription factor NF-kappaB. Piceatannol, previously reported as a selective inhibitor of protein tyrosine kinase Syk, is structurally homologous to resveratrol. Whether piceatannol can also suppress NF-kappaB activation was investigated. The treatment of human myeloid cells with piceatannol suppressed TNF-induced DNA binding activity of NF-kappaB. In contrast, stilbene or rhaponticin (another analog of piceatannol) had no effect, suggesting the critical role of hydroxyl groups. The effect of piceatannol was not restricted to myeloid cells, as TNF-induced NF-kappaB activation was also suppressed in lymphocyte and epithelial cells. Piceatannol also inhibited NF-kappaB activated by H(2)O(2), PMA, LPS, okadaic acid, and ceramide. Piceatannol abrogated the expression of TNF-induced NF-kappaB-dependent reporter gene and of matrix metalloprotease-9, cyclooxygenase-2, and cyclin D1. When examined for the mechanism, we found that piceatannol inhibited TNF-induced IkappaBalpha phosphorylation, p65 phosphorylation, p65 nuclear translocation, and IkappaBalpha kinase activation, but had no significant effect on IkappaBalpha degradation. Piceatannol inhibited NF-kappaB in cells with deleted Syk, indicating the lack of involvement of this kinase. Overall, our results clearly demonstrate that hydroxyl groups of stilbenes are critical and that piceatannol, a tetrahydroxystilbene, suppresses NF-kappaB activation induced by various inflammatory agents through inhibition of IkappaBalpha kinase and p65 phosphorylation.
Collapse
Affiliation(s)
- Kazuhiro Ashikawa
- Cytokine Research Laboratory, Department of Bioimmunotherapy, University of Texas, M. D. Anderson Cancer Center, Houston 77030, USA
| | | | | | | | | | | |
Collapse
|
92
|
Abstract
In this study, a role for cellular Src in muscle cell proliferation and differentiation was investigated. Pharmacological inhibition of Src-class kinases repressed proliferation and promoted differentiation of the C2C12 muscle cell line, even when the cells were cultured under growth-inducing conditions of high serum. Pharmacological inhibition of Src-class kinases also affected cellular components that regulate proliferation and differentiation in muscle; cyclin D1 levels were reduced while, myogenin was increased. Suppression of cyclin D1 and enhancement of myogenin levels also occurred upon expression of a dominant negative Src, corroborating a role for Src kinases in regulating proliferation and differentiation. Inhibition of Src-family kinases also blocked fibroblast growth factor (FGF) induced proliferation but, notably, did not reverse the effect of FGF to inhibit differentiation. Evidence for the Src-class kinase Src in myoblast mitogenesis was obtained by determining the pattern of protein expression and activity for this kinase. Under all conditions examined, Src's expression and enzymatic activity were high in cultures of myoblasts and down-regulated during differentiation. Importantly, Src's activity was rapidly stimulated by mitogen-containing serum and attenuated when myoblasts were switched to low serum-containing differentiation medium. These data indicate that Src is important for maintaining muscle cell proliferation.
Collapse
Affiliation(s)
- William J Rosoff
- Department of Neuroscience, Georgetown University Medical Center, Washington DC 20007, USA
| | | |
Collapse
|
93
|
Ameixa C, Friedland JS. Interleukin-8 secretion from Mycobacterium tuberculosis-infected monocytes is regulated by protein tyrosine kinases but not by ERK1/2 or p38 mitogen-activated protein kinases. Infect Immun 2002; 70:4743-6. [PMID: 12117995 PMCID: PMC128139 DOI: 10.1128/iai.70.8.4743-4746.2002] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Mycobacterium tuberculosis upregulates NF-kappaB binding and interleukin-8 (IL-8) gene expression and secretion in primary human monocytes. Inhibition of tyrosine protein kinases but not of ERK1/2 or p38 mitogen-activated protein kinases downregulates tuberculosis-induced IL-8 secretion. The inhibitor genistein decreased NF-kappaB nuclear translocation and IL-8 gene transcription in addition to acting on posttranscriptional processing.
Collapse
Affiliation(s)
- Clara Ameixa
- Department of Infectious Diseases, Faculty of Medicine, Imperial College of Science, Technology and Medicine, Hammersmith Campus, London, United Kingdom
| | | |
Collapse
|
94
|
Abstract
The vascular endothelial surface is a major target of oxidative stress, but we are only now beginning to understand the molecular sources and physiologic consequences of such oxidative activity. Along with exogenous oxidants, provided by professional phagocytes or circulating enzymes, vascular cells generate oxidants in response to cytokine and growth factor stimulation, and these endogenous oxidants participate in vascular cell signal transduction. Endothelial cells express at least four of the five principal subunits of an NADPH oxidase, and we review evidence that such an oxidase is tightly regulated in both activity and in subcellular targeting. Both of these features are likely to contribute to the signal specificity of unstable oxidants.
Collapse
Affiliation(s)
- Lance S Terada
- Department of Medicine, University of Texas Southwestern, and the Dallas VAMC, Dallas, TX 75216, USA.
| |
Collapse
|
95
|
Clohisy JC, Teitelbaum S, Chen S, Erdmann JM, Abu-Amer Y. Tumor necrosis factor-alpha mediates polymethylmethacrylate particle-induced NF-kappaB activation in osteoclast precursor cells. J Orthop Res 2002; 20:174-81. [PMID: 11918294 DOI: 10.1016/s0736-0266(01)00088-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Tumor necrosis factor-alpha (TNF) is a potent osteoclastogenic cytokine that has a fundamental role in the pathogenesis of implant particle-induced osteolysis. The nuclear transcription factor NF-kappaB mediates TNF signaling and this transcription complex is necessary for osteoclastogenesis. Because polymethylmethacrylate (PMMA) particles cause osteolysis, we reasoned the PMMA would induce NF-kappaB activation. In fact, we find that exposure of osteoclast precursors, in the form of colony stimulating factor-1 (CSF-1) dependent murine bone marrow macrophages, to PMMA particles prompts nuclear translocation and activation of NF-kappaB. Supershift assays confirm the presence of the p50 and p65 NF-kappaB subunits in the activated transcription factor. Particle-induced NF-kappaB activation is equal in both wild type and LPS- hyporesponsive cells indicating that the phenomenon does not represent endotoxin contamination. A soluble, competitive inhibitor of TNF (huTNFr:Fc) dampens particle-directed NF-kappaB activation and this response is also abrogated in TNF-/- osteoclast precursors. Thus, PMMA particle activation of NF-kappaB is a secondary event resulting from enhanced TNF expression and is independent of LPS contamination.
Collapse
Affiliation(s)
- John C Clohisy
- Department of Orthopaedic Surgery, Barnes-Jewish Hospital at Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | | | | | | | |
Collapse
|
96
|
Lee SE, Chung WJ, Kwak HB, Chung CH, Kwack KB, Lee ZH, Kim HH. Tumor necrosis factor-alpha supports the survival of osteoclasts through the activation of Akt and ERK. J Biol Chem 2001; 276:49343-9. [PMID: 11675379 DOI: 10.1074/jbc.m103642200] [Citation(s) in RCA: 202] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Differentiated osteoclasts have a short life span. We tested various cytokines and growth factors for the effects on the survival of purified mature osteoclasts. In the absence of any added factors, osteoclasts exhibited the survival rate of less than 25% after a 24-h incubation. Among the tested factors, tumor necrosis factor-alpha (TNF-alpha) was found to increase the survival rate to approximately 80%. The TNF-alpha-enhanced survival of osteoclasts appeared to be associated with reduction in apoptosis and suppression of caspase activation. The antiapoptotic signaling pathways involved in the TNF-alpha-induced osteoclast survival were investigated. TNF-alpha treatment increased the phosphorylation of Akt in osteoclasts, which was suppressed by a phosphatidylinositol 3-kinase inhibitor LY294002 and an Src family kinase-selective inhibitor PP1. These inhibitors also attenuated the TNF-alpha stimulation of osteoclast survival. In addition an increase in the phosphorylation of ERK was observed upon TNF-alpha stimulation. PD98059, a specific inhibitor of the ERK-activating kinase MEK-1, abolished the TNF-alpha-induced ERK phosphorylation and osteoclast survival, and in these responses the involvement of Grb2 and ceramide was observed. These results suggest that TNF-alpha promotes the survival of osteoclasts by engaging the phosphatidylinositol 3-kinase Akt and MEK/ERK signaling pathways.
Collapse
Affiliation(s)
- S E Lee
- National Research Laboratory for Bone Metabolism, Chosun University, Gwangju 501-759, Korea
| | | | | | | | | | | | | |
Collapse
|
97
|
Wooten MW, Vandenplas ML, Seibenhener ML, Geetha T, Diaz-Meco MT. Nerve growth factor stimulates multisite tyrosine phosphorylation and activation of the atypical protein kinase C's via a src kinase pathway. Mol Cell Biol 2001; 21:8414-27. [PMID: 11713277 PMCID: PMC100005 DOI: 10.1128/mcb.21.24.8414-8427.2001] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Atypical protein kinase C (PKC) isoforms are required for nerve growth factor (NGF)-initiated differentiation of PC12 cells. In the present study, we report that PKC-iota becomes tyrosine phosphorylated in the membrane coincident with activation posttreatment with nerve growth factor. Tyrosine phosphorylation and activation of PKC-iota were inhibited in a dose-dependent manner by both PP2 and K252a, src and TrkA kinase inhibitors. Purified src was observed to phosphorylate and activate PKC-iota in vitro. In PC12 cells deficient in src kinase activity, both NGF-induced tyrosine phosphorylation and activation of PKC-iota were also diminished. Furthermore, we demonstrate activation of src by NGF along with formation of a signal complex including the TrkA receptor, src, and PKC-iota. Recruitment of PKC-iota into the complex was dependent on the tyrosine phosphorylation state of PKC-iota. The association of src and PKC-iota was constitutive but was enhanced by NGF treatment, with the src homology 3 domain interacting with a PXXP sequence within the regulatory domain of PKC-iota (amino acids 98 to 114). Altogether, these findings support a role for src in regulation of PKC-iota. Tyrosine 256, 271, and 325 were identified as major sites phosphorylated by src in the catalytic domain. Y256F and Y271F mutations did not alter src-induced activation of PKC-iota, whereas the Y325F mutation significantly reduced src-induced activation of PKC-iota. The functional relevance of these mutations was tested by determining the ability of each mutant to support TRAF6 activation of NF-kappaB, with significant impairment by the Y325F PKC-iota mutant. Moreover, when the Y352F mutant was expressed in PC12 cells, NGF's ability to promote survival in serum-free media was reduced. In summary, we have identified a novel mechanism for NGF-induced activation of atypical PKC involving tyrosine phosphorylation by c-Src.
Collapse
Affiliation(s)
- M W Wooten
- Department of Biological Sciences, Auburn University, 331 Funchess Hall, Auburn, AL 36849, USA.
| | | | | | | | | |
Collapse
|
98
|
Bierhaus A, Schiekofer S, Schwaninger M, Andrassy M, Humpert PM, Chen J, Hong M, Luther T, Henle T, Klöting I, Morcos M, Hofmann M, Tritschler H, Weigle B, Kasper M, Smith M, Perry G, Schmidt AM, Stern DM, Häring HU, Schleicher E, Nawroth PP. Diabetes-associated sustained activation of the transcription factor nuclear factor-kappaB. Diabetes 2001; 50:2792-808. [PMID: 11723063 DOI: 10.2337/diabetes.50.12.2792] [Citation(s) in RCA: 627] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Activation of the transcription factor nuclear factor-kappaB (NF-kappaB) has been suggested to participate in chronic disorders, such as diabetes and its complications. In contrast to the short and transient activation of NF-kappaB in vitro, we observed a long-lasting sustained activation of NF-kappaB in the absence of decreased IkappaBalpha in mononuclear cells from patients with type 1 diabetes. This was associated with increased transcription of NF-kappaBp65. A comparable increase in NF-kappaBp65 antigen and mRNA was also observed in vascular endothelial cells of diabetic rats. As a mechanism, we propose that binding of ligands such as advanced glycosylation end products (AGEs), members of the S100 family, or amyloid-beta peptide (Abeta) to the transmembrane receptor for AGE (RAGE) results in protein synthesis-dependent sustained activation of NF-kappaB both in vitro and in vivo. Infusion of AGE-albumin into mice bearing a beta-globin reporter transgene under control of NF-kappaB also resulted in prolonged expression of the reporter transgene. In vitro studies showed that RAGE-expressing cells induced sustained translocation of NF-kappaB (p50/p65) from the cytoplasm into the nucleus for >1 week. Sustained NF-kappaB activation by ligands of RAGE was mediated by initial degradation of IkappaB proteins followed by new synthesis of NF-kappaBp65 mRNA and protein in the presence of newly synthesized IkappaBalpha and IkappaBbeta. These data demonstrate that ligands of RAGE can induce sustained activation of NF-kappaB as a result of increased levels of de novo synthesized NF-kappaBp65 overriding endogenous negative feedback mechanisms and thus might contribute to the persistent NF-kappaB activation observed in hyperglycemia and possibly other chronic diseases.
Collapse
MESH Headings
- Adult
- Amyloid beta-Peptides/metabolism
- Amyloid beta-Peptides/pharmacology
- Animals
- Cell Nucleus/metabolism
- Cytoplasm/metabolism
- DNA/metabolism
- Diabetes Mellitus, Type 1/metabolism
- Endothelium, Vascular/metabolism
- Feedback
- Female
- Glycation End Products, Advanced/metabolism
- Glycation End Products, Advanced/pharmacology
- Humans
- I-kappa B Proteins/metabolism
- Immunohistochemistry
- Leukocytes, Mononuclear/metabolism
- Male
- Mice
- Mice, Transgenic
- Middle Aged
- NF-kappa B/analysis
- NF-kappa B/genetics
- NF-kappa B/metabolism
- NF-kappa B/physiology
- RNA, Messenger/biosynthesis
- Rats
- Rats, Sprague-Dawley
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/metabolism
- S100 Proteins/metabolism
- Serum Albumin, Bovine/pharmacology
- Transcription Factor RelA
Collapse
Affiliation(s)
- A Bierhaus
- Department of Medicine I and Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Majeed M, Caveggion E, Lowell CA, Berton G. Role of Src kinases and Syk in Fcγ receptor‐mediated phagocytosis and phagosome‐lysosome fusion. J Leukoc Biol 2001. [DOI: 10.1189/jlb.70.5.801] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Meytham Majeed
- Department of Pathology, Section of General Pathology, University of Verona, Verona, Italy, and
| | - Elena Caveggion
- Department of Pathology, Section of General Pathology, University of Verona, Verona, Italy, and
| | - Clifford A. Lowell
- Department of Laboratory Medicine, University of California San Francisco, San Francisco
| | - Giorgio Berton
- Department of Pathology, Section of General Pathology, University of Verona, Verona, Italy, and
| |
Collapse
|
100
|
Ferlito M, Romanenko OG, Ashton S, Squadrito F, Halushka PV, Cook JA. Effect of cross‐tolerance between endotoxin and TNF‐α or IL‐1β on cellular signaling and mediator production. J Leukoc Biol 2001. [DOI: 10.1189/jlb.70.5.821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Marcella Ferlito
- Departments of Physiology and Neuroscience and Charleston
- Institute of Pharmacology, Medical University of Messina, Italy
| | | | - Sarah Ashton
- Departments of Physiology and Neuroscience and Charleston
| | | | - Perry V. Halushka
- Pharmacology and Medicine, Medical University of South Carolina, Charleston, and
| | - James A. Cook
- Departments of Physiology and Neuroscience and Charleston
| |
Collapse
|