1
|
Burda JE, O'Shea TM, Ao Y, Suresh KB, Wang S, Bernstein AM, Chandra A, Deverasetty S, Kawaguchi R, Kim JH, McCallum S, Rogers A, Wahane S, Sofroniew MV. Divergent transcriptional regulation of astrocyte reactivity across disorders. Nature 2022; 606:557-564. [PMID: 35614216 PMCID: PMC10027402 DOI: 10.1038/s41586-022-04739-5] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/07/2022] [Indexed: 01/30/2023]
Abstract
Astrocytes respond to injury and disease in the central nervous system with reactive changes that influence the outcome of the disorder1-4. These changes include differentially expressed genes (DEGs) whose contextual diversity and regulation are poorly understood. Here we combined biological and informatic analyses, including RNA sequencing, protein detection, assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) and conditional gene deletion, to predict transcriptional regulators that differentially control more than 12,000 DEGs that are potentially associated with astrocyte reactivity across diverse central nervous system disorders in mice and humans. DEGs associated with astrocyte reactivity exhibited pronounced heterogeneity across disorders. Transcriptional regulators also exhibited disorder-specific differences, but a core group of 61 transcriptional regulators was identified as common across multiple disorders in both species. We show experimentally that DEG diversity is determined by combinatorial, context-specific interactions between transcriptional regulators. Notably, the same reactivity transcriptional regulators can regulate markedly different DEG cohorts in different disorders; changes in the access of transcriptional regulators to DNA-binding motifs differ markedly across disorders; and DEG changes can crucially require multiple reactivity transcriptional regulators. We show that, by modulating reactivity, transcriptional regulators can substantially alter disorder outcome, implicating them as therapeutic targets. We provide searchable resources of disorder-related reactive astrocyte DEGs and their predicted transcriptional regulators. Our findings show that transcriptional changes associated with astrocyte reactivity are highly heterogeneous and are customized from vast numbers of potential DEGs through context-specific combinatorial transcriptional-regulator interactions.
Collapse
Affiliation(s)
- Joshua E Burda
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Timothy M O'Shea
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Yan Ao
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Keshav B Suresh
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shinong Wang
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Alexander M Bernstein
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Ashu Chandra
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA, USA
| | - Sandeep Deverasetty
- Department of Psychiatry, University of California Los Angeles, Los Angeles, CA, USA
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Riki Kawaguchi
- Department of Psychiatry, University of California Los Angeles, Los Angeles, CA, USA
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Jae H Kim
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Sarah McCallum
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alexandra Rogers
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Shalaka Wahane
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Aloor R, Zhang C, Bandyopadhyay M, Dasgupta S. Impact of nuclear factor-κB on restoration of neuron growth and differentiation in hippocampus of degenerative brain. J Neurosci Res 2015; 93:1471-5. [PMID: 25586448 DOI: 10.1002/jnr.23547] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 11/30/2014] [Indexed: 01/29/2023]
Abstract
The mode of action of nuclear factor-κB (NF-κB) has been extensively observed in different aspects of cell growth and proliferation. The transcription factor regulates various genes controlling inflammation and anti-inflammatory responses in different tissues. Thus, NF-κB signal gains a therapeutic prospect. The activation of NF-κB requires nuclear localization of its p65 subunit. Research also indicates an impact of phosphorylated p65 on the transcription of genes during cell growth and the immune response. Following the trends in investigations over decades, different observations suggest that NF-κB activation and phosphorylation of p65 regulate neuronal plasticity. Also, inhibition of NF-κB activation is a well-demonstrated way to attenuate inflammation. In addition to anti-inflammatory drugs, recent researches unwind a way to regulate regeneration and repair tissue damage. Thus, keeping a critical view on NF-κB signals, we propose the importance of natural or synthetic NF-κB activators for neurogenesis.
Collapse
Affiliation(s)
- Rohit Aloor
- Department of Microbiology, Immunology, Saint James School of Medicine, Anguilla, British West Indies
| | - Christine Zhang
- Department of Microbiology, Immunology, Saint James School of Medicine, Anguilla, British West Indies
| | | | - Subhajit Dasgupta
- Department of Microbiology, Immunology, Saint James School of Medicine, Anguilla, British West Indies.,Department of Biochemistry, Saint James School of Medicine, Anguilla, British West Indies
| |
Collapse
|
3
|
Saavedra A, Baltazar G, Duarte EP. Driving GDNF expression: the green and the red traffic lights. Prog Neurobiol 2008; 86:186-215. [PMID: 18824211 DOI: 10.1016/j.pneurobio.2008.09.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 06/18/2008] [Accepted: 09/03/2008] [Indexed: 01/28/2023]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is widely recognized as a potent survival factor for dopaminergic neurons of the nigrostriatal pathway that degenerate in Parkinson's disease (PD). In animal models of PD, GDNF delivery to the striatum or the substantia nigra protects dopaminergic neurons against subsequent toxin-induced injury and rescues previously damaged neurons, promoting recovery of the motor function. Thus, GDNF was proposed as a potential therapy to PD aimed at slowing down, halting or reversing neurodegeneration, an issue addressed in previous reviews. However, the use of GDNF as a therapeutic agent for PD is hampered by the difficulty in delivering it to the brain. Another potential strategy is to stimulate the endogenous expression of GDNF, but in order to do that we need to understand how GDNF expression is regulated. The aim of this review is to do a comprehensive analysis of the state of the art on the control of endogenous GDNF expression in the nervous system, focusing mainly on the nigrostriatal pathway. We address the control of GDNF expression during development, in the adult brain and after injury, and how damaged neurons signal glial cells to up-regulate GDNF. Pharmacological agents or natural molecules that increase GDNF expression and show neuroprotective activity in animal models of PD are reviewed. We also provide an integrated overview of the signalling pathways linking receptors for these molecules to the induction of GDNF gene, which might also become targets for neuroprotective therapies in PD.
Collapse
Affiliation(s)
- Ana Saavedra
- Department of Cell Biology, Immunology and Neurosciences, Faculty of Medicine, University of Barcelona, Carrer Casanova 143, 08036 Barcelona, Spain.
| | | | | |
Collapse
|
4
|
Abstract
Many eukaryotic proteins share a sequence designated as the zona pellucida (ZP) domain. This structural element, present in extracellular proteins from a wide variety of organisms, from nematodes to mammals, consists of approximately 260 amino acids with eight conserved cysteine (Cys) residues and is located close to the C terminus of the polypeptide. ZP domain proteins are often glycosylated, modular structures consisting of multiple types of domains. Predictions can be made about some of the structural features of the ZP domain and ZP domain proteins. The functions of ZP domain proteins vary tremendously, from serving as structural components of egg coats, appendicularian mucous houses, and nematode dauer larvae, to serving as mechanotransducers in flies and receptors in mammals and nonmammals. Generally, ZP domain proteins are present in filaments and/or matrices, which is consistent with the role of the domain in protein polymerization. A general mechanism for assembly of ZP domain proteins has been presented. It is likely that the ZP domain plays a common role despite its presence in proteins of widely diverse functions.
Collapse
Affiliation(s)
- Luca Jovine
- Brookdale Department of Molecular, Cell, and Developmental Biology, Mount Sinai School of Medicine, New York, New York 10029-6574, USA.
| | | | | | | |
Collapse
|
5
|
Xiang H, Tian Y, Sun Y. Effect of morphine and naloxone on release of the excitatory amino acids of spinal astrocytes induced by TNF-α. ACTA ACUST UNITED AC 2005; 25:91-3. [PMID: 15934319 DOI: 10.1007/bf02831397] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The effect of morphine and naloxone on release of the excitatory amino acids (EAAs) of spinal astrocytes induced by TNF-a was studied. Astrocytes were purified from 2- to 3-day old SD rats and divided into 8 groups: group 1 (without any stimulatants); group 2 (10 ng/ml TNF-alpha); group3 (10 ng/ml TNF-alpha + 0.5 micromol/L morphine); group 4 (10 ng/ml TNF-alpha + 1.0 micromol/L morphine); group 5 (10 ng/ml TNF-alpha + 2. 0 micromol/L morphine); group 6 (10 ng/ml TNF-alpha + 0.5 micromol/L naloxone); group 7 (10 ng/ml TNF-alpha + 1.0 0Lmol/L naloxone); group 8 (10 ng/ml TNF-alpha + 2. 0 micromol/L naloxone). In group 2, 3, 4 and 5, 0, 0.5, 1.0 or 2.0 micromol/L morphine was added to the cells cultured with serum-free Neurobasal/B27 medium containing 10 ng/ml TNF-alpha respectively, while in group 6, 7 and 8, 0.5, 1.0 or 2.0 micromol/L naloxone was added respectively to the cells cultured with serum-free Neurobasal/B27 medium containing 10 ng/ml TNF-alpha. After 30 min incubation, high-pressure liquid chromatography (HPLC) was used to measure the levels of EAAs in all cultured cells. The results showed the level of EAAs in group 2 was significant higher than in group 1 (P < 0.01). As compared with group 2, the levels of EAAs in group 3, 4 and 5 were decreased with the difference being significant between group 5 and group 2 (P < 0.01) or between group 4 and group 2 (P < 0.05). The levels of EAAs in group 6, 7 and group 8 was significantly lower than in group 2 (P < 0.05 or P < 0.01). It was concluded that TNF-alpha could promote the release of glutamate and aspartate from astrocytes, and morphine and naloxone might reduce the release of EAAs in cultured spinal astrocytes induced by TNF-alpha.
Collapse
Affiliation(s)
- Hongbing Xiang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | | | | |
Collapse
|
6
|
Zhai Q, Luo Y, Zhang Y, Berman MA, Dorf ME. Low nuclear levels of nuclear factor-kappa B are essential for KC self-induction in astrocytes: requirements for shuttling and phosphorylation. Glia 2005; 48:327-36. [PMID: 15390109 DOI: 10.1002/glia.20087] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Stimulation with the chemokine KC induces an autocrine response in mouse astrocytes. A requirement for NF-kappa B was established for KC self-induction. NF-kappa B inhibitors, p65 antisense oligonucleotides, or dominant-negative Ikappa Balpha inhibited this autocrine response. Mutation of a specific kappa B site in the KC promoter also blocked KC self-induction. Chromatin immunoprecipitation and in vivo footprinting confirmed the direct binding of NF-kappa B to the KC promoter. However, neither NF-kappa B nuclear translocation, increased Ikappa B degradation, nor upregulation of NF-kappa B DNA binding activity was observed after KC stimulation. Reporter gene assays demonstrated KC-upregulated NF-kappa B transcriptional activity, and this effect was inhibited by dominant-negative IkappaBalpha. Accumulation of NF-kappaB was noted within the nucleus in the presence of nuclear export inhibitor leptomycin B, demonstrating constitutive shuttling of NF-kappa B between the cytoplasm and nucleus. Blocking NF-kappa B shuttling inhibited KC transcription. KC induced p65 phosphorylation, which was critical for NF-kappa B activation as determined with the Gal-4-p65 fusion protein and mutation of p65 phosphorylation sites. In conclusion, low-level nuclear NF-kappa B is essential for KC self-induction, and this effect is mediated by shuttling and phosphorylation of NF-kappa B. The results outline a novel mechanism for NF-kappa B participation in transcription regulation.
Collapse
Affiliation(s)
- Qiwei Zhai
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
7
|
Catley MC, Cambridge LM, Nasuhara Y, Ito K, Chivers JE, Beaton A, Holden NS, Bergmann MW, Barnes PJ, Newton R. Inhibitors of protein kinase C (PKC) prevent activated transcription: role of events downstream of NF-kappaB DNA binding. J Biol Chem 2004; 279:18457-66. [PMID: 14976190 DOI: 10.1074/jbc.m400765200] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In pulmonary A549 cells, the protein kinase C (PKC) inhibitor, Ro 31-8220, and the phosphotidylcholine-specific phospholipase C inhibitor, D609, prevent NF-kappaB-dependent transcription, yet NF-kappaB DNA binding is unaffected (Bergmann, M., Hart, L., Lindsay, M., Barnes, P. J., and Newton, R. (1998) J. Biol. Chem. 273, 6607-6610). We now show that this effect also occurs in BEAS-2B bronchial epithelial cells as well as with other PKC inhibitors (Gö 6976, GF109203X, and calphostin C) in A549 cells. Similarly, phorbol ester, a diacylglycerol mimetic, activates NF-kappaB-dependent transcription and potentiates tumor necrosis factor alpha (TNFalpha)-induced NF-kappaB-dependent transcription, yet unlike TNFalpha, poorly activates IkappaB kinase (IKK) activity, IkappaBalpha degradation, or NF-kappaB DNA binding in both A549 and BEAS-2B cells. As phorbol ester-induced NF-kappaB-dependent transcription was relatively insensitive to the proteasome inhibitor, MG-132, PKC may affect NF-kappaB-dependent transcription via mechanisms other than the core IKK-IkappaB pathway. This is supported by Gal4 one hybrid analysis of p65/RelA transactivation, which was potentiated by TNFalpha and phorbol ester and was inhibited by Ro 31-8220 and D609. Additionally, a number of PKC isoforms, particularly the novel isoform PKCepsilon, induced p65/RelA transactivation. Phosphorylation of p65/RelA and cAMP-responsive element-binding protein (CREB)-binding protein (CBP) was increased by TNFalpha treatment and, in the case of CBP, was prevented by Ro 31-8220 or D609. However, p65/RelA-CBP interactions were unaffected by either compound. As this effect was not limited to NF-kappaB, but was a more general feature of inducible gene transcription, we suggest PKC isoforms may provide a point of intervention in diseases such as inflammation, or cancer, where activated gene expression is prominent.
Collapse
Affiliation(s)
- Matthew C Catley
- Department of Thoracic Medicine, National Heart and Lung Institute, Imperial College, Dovehouse Street, London SW3 6LY, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Sathe SS, Sizemore N, Li X, Vithalani K, Commane M, Swiatkowski SM, Stark GR. Mutant human cells with constitutive activation of NF-kappaB. Proc Natl Acad Sci U S A 2004; 101:192-7. [PMID: 14691254 PMCID: PMC314161 DOI: 10.1073/pnas.0306812101] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
We have used a genetic approach to generate eight different mutant human cell lines in which NF-kappaB is constitutively activated. These independent clones have different phenotypes and belong to several different genetic complementation groups. In one clone inhibitor of kappaB(IkappaB) kinase is constitutively active, but in the seven others it is not, despite the fact that IkappaB is degraded in all eight clones. Thus, IkappaB kinase-independent mechanisms of IkappaB degradation and NF-kappaB activation are predominant in these mutants. Biochemical analyses of the mutants revealed that they fall into at least five different categories, differing in the sets of upstream kinases that are activated, confirming multiple mechanisms of NF-kappaB activation. By introducing a retroviral cDNA library into the Ras C6 cell line, with constitutively active NF-kappaB, followed by selection for functional complementation, we isolated a cDNA encoding a C-terminal fragment of enolase 1 and identified it as negative regulator of NF-kappaB.
Collapse
Affiliation(s)
- Swati S Sathe
- Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Xu J, Chalimoniuk M, Shu Y, Simonyi A, Sun AY, Gonzalez FA, Weisman GA, Wood WG, Sun GY. Prostaglandin E2 production in astrocytes: regulation by cytokines, extracellular ATP, and oxidative agents. Prostaglandins Leukot Essent Fatty Acids 2003; 69:437-48. [PMID: 14623498 DOI: 10.1016/j.plefa.2003.08.016] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Upregulation and activation of phospholipases A2 (PLA2) and cyclooxygenases (COX) leading to prostaglandin E2(PGE2) production have been implicated in a number of neurodegenerative diseases. In this study, we investigated PGE2 production in primary rat astrocytes in response to agents that activate PLA2 including pro-inflammatory cytokines (IL-1beta, TNFalpha and IFNgamma), the P2 nucleotide receptor agonist ATP, and oxidants (H2O2 and menadione). Exposure of astrocytes to cytokines resulted in a time-dependent increase in PGE2 production that was marked by increased expression of secretory sPLA2 and COX-2, but not COX-1 and cytosolic cPLA2. Although astrocytes responded to ATP or phorbol ester (PMA) with increased cPLA2 phosphorylation and arachidonic acid release, ATP or PMA only caused a small increase in levels of PGE2. However, when astrocytes were first treated with cytokines, further exposure to ATP or PMA, but not H2O2 or menadione, markedly increased PGE2 production. These results suggest that ATP release during neuronal excitation or injury can enhance the inflammatory effects of cytokines on PGE2 production and may contribute to chronic inflammation seen in Alzheimer's disease.
Collapse
Affiliation(s)
- J Xu
- Department of Biochemistry, University of Missouri, Columbia, MO 65212, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
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
|
11
|
Yalcin A, Koulich E, Mohamed S, Liu L, D'Mello SR. Apoptosis in cerebellar granule neurons is associated with reduced interaction between CREB-binding protein and NF-kappaB. J Neurochem 2003; 84:397-408. [PMID: 12559002 DOI: 10.1046/j.1471-4159.2003.01540.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cerebellar granule neurons undergo apoptosis when switched from medium containing depolarizing levels of potassium (high K+ medium, HK) to medium containing low K+ (LK). NF-kappaB, a ubiquitously expressed transcription factor, is involved in the survival-promoting effects of HK. However, neither the expression nor the intracellular localization of the five NF-kappaB proteins, or of IkappaB-alpha and IkappaB-beta, are altered in neurons primed to undergo apoptosis by LK, suggesting that uncommon mechanisms regulate NF-kappaB activity in granule neurons. In this study, we show that p65 interacts with the transcriptional co-activator, CREB-binding protein (CBP), in healthy neurons. The decrease in NF-kappaB transcriptional activity caused by LK treatment is accompanied by a reduction in the interaction between p65 and CBP, an alteration that is accompanied by hyperphosporylation of CBP. LK-induced CBP hyperphosphorylation can be mimicked by inhibitors of protein phosphatase (PP) 2A and PP2A-like phosphatases such as okadaic acid and cantharidin, which also causes a reduction in p65-CBP association. In addition, treatment with these inhibitors induces cell death. Treatment with high concentrations of the broad-spectrum kinase inhibitor staurosporine prevents LK-mediated CBP hyperphosphorylation and inhibits cell death. In vitro kinase assays using glutathione-S-transferase (GST)-CBP fusion proteins map the LK-regulated site of phosphorylation to a region spanning residues 1662-1840 of CBP. Our results are consistent with possibility that LK-induced apoptosis is triggered by CBP hyperphosphorylation, an alteration that causes the dissociation of CBP and NF-kappaB.
Collapse
Affiliation(s)
- Asligul Yalcin
- Department of Molecular and Cell Biology, University of Texas at Dallas, Richardson 75083, USA
| | | | | | | | | |
Collapse
|
12
|
Sizemore N, Lerner N, Dombrowski N, Sakurai H, Stark GR. Distinct roles of the Ikappa B kinase alpha and beta subunits in liberating nuclear factor kappa B (NF-kappa B) from Ikappa B and in phosphorylating the p65 subunit of NF-kappa B. J Biol Chem 2002; 277:3863-9. [PMID: 11733537 DOI: 10.1074/jbc.m110572200] [Citation(s) in RCA: 330] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phosphatidylinositol 3'-kinase (PI3K) and the serine/threonine kinase AKT have critical roles in phosphorylating and transactivating the p65 subunit of nuclear factor kappaB (NF-kappaB) in response to the pro-inflammatory cytokines interleukin-1 (IL-1) and tumor necrosis factor (TNF). Mouse embryo fibroblasts (MEFs) lacking either the alpha or beta subunit of IkappaB kinase (IKK) were deficient in NF-kappaB-dependent transcription following treatment with IL-1 or TNF. However, in contrast to IKKbeta-null MEFs, IKKalpha-null MEFs were not substantially defective in the cytokine-stimulated degradation of Ikappabetaalpha or in the nuclear translocation of NF-kappaB. The IKK complexes from IKKalpha- or IKKbeta-null MEFs were both deficient in PI3K-mediated phosphorylation of the transactivation domain of the p65 subunit of NF-kappaB in response to IL-1 and TNF, and constitutively activated forms of PI3K or AKT did not potentiate cytokine-stimulated activation of NF-kappaB in either IKKalpha- or IKKbeta-null MEFs. Collectively, these data indicate that, in contrast to IKKbeta, which is required for both NF-kappaB liberation and p65 phosphorylation, IKKalpha is required solely for the cytokine-induced phosphorylation and activation of the p65 subunit of NF-kappaB that are mediated by the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Nywana Sizemore
- Department of Molecular Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | | | |
Collapse
|
13
|
Kang JL, Pack IS, Hong SM, Lee HS, Castranova V. Silica induces nuclear factor-kappa B activation through tyrosine phosphorylation of I kappa B-alpha in RAW264.7 macrophages. Toxicol Appl Pharmacol 2000; 169:59-65. [PMID: 11076697 DOI: 10.1006/taap.2000.9039] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It was previously reported that protein tyrosine kinase (PTK) but not protein kinase C or A plays an important role in silica-induced activation of NF-kappa B in macrophages. The question is raised whether PTK stimulation and NF-kappa B activation in silica-stimulated macrophages are directly connected through tyrosine phosphorylation of I kappa B-alpha. Results indicate that stimulation of macrophages with silica led to NF-kappaB activation through tyrosine phosphorylation without serine phosphorylation. Specific inhibitors of protein tyrosine kinase, such as genistein and tyrophostin AG126, prevented tyrosine phosphorylation of I kappa B-alpha in response to silica. I kappa B-alpha protein levels remained relatively unchanged for up to 60 min after silica stimulation. Moreover, inhibition of proteasome proteolytic activity did not affect NF-kappa B activation by silica. Antioxidants, such as superoxide dismutase (SOD), N-acetylcysteine (NAC), and pyrrolidine dithiocarbamate (PDTC), blocked tyrosine phosphorylation of I kappa B-alpha induced by silica, suggesting reactive oxygen species (ROS) may be important regulatory molecules in NF-kappa B activation through tyrosine phosphorylation of I kappa B-alpha. The results suggest that tyrosine phosphorylation of I kappa B-alpha represents a proteasome proteolytic activity-independent mechanism for NF-kappa B activation that directly couples NF-kappa B to cellular tyrosine kinase in silica-stimulated macrophages. This proposed mechanism of NF-kappa B activation induced by silica could be used as a target for development of antiinflammatory and antifibrosis drugs.
Collapse
Affiliation(s)
- J L Kang
- Department of Physiology, College of Medicine, Ewha Woman's University, Seoul, #158-056, Korea.
| | | | | | | | | |
Collapse
|
14
|
Wang D, Westerheide SD, Hanson JL, Baldwin AS. Tumor necrosis factor alpha-induced phosphorylation of RelA/p65 on Ser529 is controlled by casein kinase II. J Biol Chem 2000; 275:32592-7. [PMID: 10938077 DOI: 10.1074/jbc.m001358200] [Citation(s) in RCA: 365] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nuclear factor kappaB (NF-kappaB)/Rel transcription factors are key regulators of a variety of genes involved in immune and inflammatory responses, growth, differentiation, apoptosis, and development. In unstimulated cells, NF-kappaB/Rel proteins are sequestered in the cytoplasm by IkappaB inhibitor proteins. Many extracellular stimuli, such as tumor necrosis factor alpha (TNFalpha), cause rapid phosphorylation of IkappaB at N-terminal serine residues leading to ubiquitination and degradation of the inhibitor. Subsequently, NF-kappaB proteins translocate to the nucleus and activate gene expression through kappaB response elements. TNFalpha, as well as certain other stimuli, also induces the phosphorylation of the NF-kappaB proteins. Previously, we have shown that TNFalpha induces RelA/p65 phosphorylation at serine 529 and that this inducible phosphorylation increases NF-kappaB transcriptional activity on an exogenously supplied reporter (). In this report, we demonstrate that casein kinase II (CKII) interacts with p65 in vivo and can phosphorylate p65 at serine 529 in vitro. A CKII inhibitor (PD144795) inhibited TNFalpha-induced p65 phosphorylation in vivo. Furthermore, our results indicate that the association between IkappaBalpha and p65 inhibits p65 phosphorylation by CKII and that degradation of IkappaBalpha allows CKII to phosphorylate p65 to increase NF-kappaB transactivation potential. These data may explain the ability of CKII to modulate cell growth and demonstrate a mechanism whereby CKII can function in an inducible manner.
Collapse
Affiliation(s)
- D Wang
- Department of Biology, Curriculum in Genetics and Molecular Biology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599-7295, USA
| | | | | | | |
Collapse
|
15
|
Vanden Berghe W, Vermeulen L, De Wilde G, De Bosscher K, Boone E, Haegeman G. Signal transduction by tumor necrosis factor and gene regulation of the inflammatory cytokine interleukin-6. Biochem Pharmacol 2000; 60:1185-95. [PMID: 11007957 DOI: 10.1016/s0006-2952(00)00412-3] [Citation(s) in RCA: 229] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Interleukin (IL)-6 is a multifunctional cytokine that can be induced by a plethora of chemical or physiological compounds, including the inflammatory cytokines tumor necrosis factor (TNF) and IL-1. The molecule TNF has a trimeric configuration and thus binds to membrane-bound, cellular receptors to initiate cell death mechanisms and signaling pathways leading to gene induction. Previously, we showed that induced clustering of the intracellular domains of the p55 TNF receptor, or of their respective 'death domains' only, is sufficient to activate the nuclear factor kappa B (NF-kappa B) and several mitogen-activated protein kinase (MAPK) pathways. NF-kappa B is the exclusive transcription factor for induction of the IL-6 gene in response to TNF and functions as the final trigger to activate a multiprotein complex, a so-called 'enhanceosome', at the level of the IL-6 promoter. Furthermore, the enhanceosome displays histone acetylation activity, which turned out to be essential for IL-6 gene activation via NF-kappa B. However, activation of NF-kappa B alone is not sufficient for IL-6 gene induction in response to TNF, as inhibition of the coactivated extracellular signal-regulated kinase and p38 MAPK pathways blocks TNF-mediated gene expression. Nevertheless, the transactivating NF-kappa B subunit p65 is not a direct target of MAPK phosphorylation. Thus, we postulated that other components of the enhanceosome complex are sensitive to MAPK cascades and found that MAPK activity is unequivocally linked to the histone acetylation capacity of the enhanceosome to stimulate gene expression in response to TNF. In contrast, glucocorticoid repression of TNF-driven IL-6 gene expression does not depend on abrogation of histone acetyltransferase activity, but originates from interference of the liganded glucocorticoid receptor with the contacts between NF-kappa B p65 and the promoter configuration around the TATA box.
Collapse
Affiliation(s)
- W Vanden Berghe
- Department of Molecular Biology, Flanders Interuniversity Institute for Biotechnology, 9000 Ghent, Belgium
| | | | | | | | | | | |
Collapse
|
16
|
Jobin C, Sartor RB. The I kappa B/NF-kappa B system: a key determinant of mucosalinflammation and protection. Am J Physiol Cell Physiol 2000; 278:C451-62. [PMID: 10712233 DOI: 10.1152/ajpcell.2000.278.3.c451] [Citation(s) in RCA: 320] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The ubiquitous transcription factor NF-kappa B is a central regulator of the transcriptional activation of a number of genes involved in cell adhesion, immune and proinflammatory responses, apoptosis, differentiation, and growth. Induction of these genes in intestinal epithelial cells (IECs) by activated NF-kappa B profoundly influences mucosal inflammation and repair. NF-kappa B activation requires the removal of I kappa B from NF-kappa B by inducible proteolysis, which liberates this transcription factor for migration to the nucleus, where it binds to kappa B-regulatory elements and induces transcription. I kappa B alpha degradation is incomplete and delayed in IECs, resulting in buffered responses to luminal stimuli. The stimulatory environment partially determines whether the effect of NF-kappa B is protective or deleterious for the host. kappa B-dependent proinflammatory gene expression, particularly chemokines, major histocompatibility complex class II antigens, and adhesion molecules may be extremely important in early protective responses to mucosal pathogens but, when dysregulated, could lead to the development of chronic inflammation, as seen in inflammatory bowel diseases. The key role of NF-kappa B in regulating expression of a number of proinflammatory genes makes this protein an attractive target for selective therapeutic intervention.
Collapse
Affiliation(s)
- C Jobin
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599,
| | | |
Collapse
|
17
|
Carlotti F, Chapman R, Dower SK, Qwarnstrom EE. Activation of nuclear factor kappaB in single living cells. Dependence of nuclear translocation and anti-apoptotic function on EGFPRELA concentration. J Biol Chem 1999; 274:37941-9. [PMID: 10608861 DOI: 10.1074/jbc.274.53.37941] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have studied the dynamics of nuclear translocation during nuclear factor kappaB activation by using a p65(RELA)-enhanced green fluorescent protein (EGFP) fusion construct. Quantitation of expression levels indicates that EGFPRELA can be detected at physiological concentrations of about 60,000 molecules per cell. Stimulation of transfected fibroblasts with interleukin (IL)-1beta caused nuclear translocation of EGFPRELA, typically resulting in a 30-fold increase in nuclear protein at maximum induction and a concomitant 20% decrease in cytoplasmic levels. The response of individual cells to IL-1beta was graded, and the kinetics of nuclear translocation were dependent on the dose of IL-1beta and the level of EGFPRELA expression. The rate of nuclear uptake was saturable, and the time lag for uptake increased at higher EGFPRELA expression levels. Furthermore, nuclear translocation was reduced at less than saturating doses of IL-1beta suggesting that the pathway is limited by incoming signals. The response to IL-1beta was biphasic, demonstrating a decline in nuclear import rate at expression levels above three to four times endogenous. This correlated with the anti-apoptotic function of EGFPRELA which was more prominent at low expression levels and demonstrated successively less protection at higher levels. In comparison, transfection of p50 had no effect on the level of apoptosis and demonstrated some toxicity in combination with EGFPRELA.
Collapse
Affiliation(s)
- F Carlotti
- Division of Molecular Medicine, The Medical School, University of Sheffield, Sheffield S10 2JF, United Kingdom
| | | | | | | |
Collapse
|
18
|
Rothwarf DM, Karin M. The NF- B Activation Pathway:A Paradigm in Information Transfer from Membrane to Nucleus. Sci Signal 1999. [DOI: 10.1126/scisignal.51999re1] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
19
|
Rothwarf DM, Karin M. The NF-kappa B activation pathway: a paradigm in information transfer from membrane to nucleus. SCIENCE'S STKE : SIGNAL TRANSDUCTION KNOWLEDGE ENVIRONMENT 1999; 1999:RE1. [PMID: 11865184 DOI: 10.1126/stke.1999.5.re1] [Citation(s) in RCA: 281] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Nuclear factor kappa B (NF-kappaB)/Rel proteins are dimeric, sequence-specific transcription factors involved in the activation of an exceptionally large number of genes in response to inflammation, viral and bacterial infections, and other stressful situations requiring rapid reprogramming of gene expression. In unstimulated cells, NF-kappaB is sequestered in an inactive form in the cytoplasm bound to inhibitory IkappaB proteins. Stimulation leads to the rapid phosphorylation, ubiquitinylation, and ultimately proteolytic degradation of IkappaB, which frees NF-kappaB to translocate to the nucleus and activate the transcription of its target genes. The multisubunit IkappaB kinase (IKK) responsible for the inducible phosphorylation of IkappaB appears to be the initial point of convergence for most stimuli that activate NF-kappaB. IKK contains two catalytic subunits, IKKalpha and IKKbeta, both of which phosphorylate IkappaB at sites phosphorylated in vivo. Gene knockout studies indicate that IKKbeta is primarily responsible for the activation of NF-kappaB in response to proinflammatory stimuli, whereas IKKalpha is essential for keratinocyte differentiation. The activity of IKK is regulated by phosphorylation. IKK contains a regulatory subunit, IKKgamma, which is critical for activation of IKK and is postulated to serve as a recognition site for upstream activators. When phosphorylated, the IKK recognition site on IkappaBalpha serves as a specific recognition site for the kappa-TrCP-like component of a Skp1-Cullin-F-box-type E3 ubiquitin-protein ligase. A variety of other signaling events, including phosphorylation of NF-kappaB, phosphorylation of IKK, new synthesis of IkappaBs, and the processing of NF-kappaB precursors provide mechanisms of modulating the amount and duration of NF-kappaB activity.
Collapse
Affiliation(s)
- D M Rothwarf
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, University of California, San Diego, CA 92093-0636, USA.
| | | |
Collapse
|
20
|
Pahlevan AA, Wright DJM, Andrews C, George KM, Small PLC, Foxwell BM. The Inhibitory Action of Mycobacterium ulcerans Soluble Factor on Monocyte/T Cell Cytokine Production and NF-κB Function. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.7.3928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Abstract
Buruli ulcer is a chronic and progressive necrotizing ulcer for which there is no medical treatment. Historically, a soluble toxin (factor) derived from the causative Mycobacterium ulcerans was found to induce the massive necrosis of skin and s.c. tissue seen in this condition. However, the persistence of the disease is thought to be caused by a lack of any immune response. We therefore investigated whether the factor was related to immunosuppression. A protocol to partially purify the factor was developed, and its effects on immune competent cells were tested. The factor produced >95% inhibition of LPS-induced release of TNF and IL-10 from human monocytes and caused a loss of adherence of these cells without cell death. The factor also blocked the production of IL-2 from activated T lymphocytes. The factor had no effect on TNF-induced cytotoxicity, but abrogated TNF-induced NF-κB activation. Surprisingly, a synergy was observed between the factor and phorbol ester-directed NF-κB activation. The factor had no effect on IL-1- or LPS-induced NF-κB activity, indicating selective activity of the factor. The factor did not inhibit the degradation of IκBα induced by TNF, indicating that the target for its activity lies within an undefined part of the TNF signaling mechanism. The data indicate that the localized immunosuppression associated with Buruli ulcer relates to the activity of the released factor, and this may provide a target for future therapeutic strategies for this intractable disease.
Collapse
Affiliation(s)
- Ali A. Pahlevan
- *Infectious Diseases and Microbiology, Charing Cross Campus, Imperial College, London, United Kingdom
- ‡Cytokine Biology and Signal Transduction Laboratory, Kennedy Institute of Rheumatology, London, United Kingdom
| | - David J. M. Wright
- *Infectious Diseases and Microbiology, Charing Cross Campus, Imperial College, London, United Kingdom
| | - Caroline Andrews
- ‡Cytokine Biology and Signal Transduction Laboratory, Kennedy Institute of Rheumatology, London, United Kingdom
| | - Kathleen M. George
- †Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840; and
| | - Pamela L. C. Small
- †Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840; and
| | - Brian M. Foxwell
- ‡Cytokine Biology and Signal Transduction Laboratory, Kennedy Institute of Rheumatology, London, United Kingdom
| |
Collapse
|
21
|
Raju U, Gumin GJ, Tofilon PJ. NF kappa B activity and target gene expression in the rat brain after one and two exposures to ionizing radiation. RADIATION ONCOLOGY INVESTIGATIONS 1999; 7:145-52. [PMID: 10406055 DOI: 10.1002/(sici)1520-6823(1999)7:3<145::aid-roi2>3.0.co;2-r] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The central nervous system injury that can result after radiotherapy has been suggested to involve induced gene expression and cytokine production. We have previously shown that irradiation of primary cultures of rat astrocytes results in the activation of NF kappa B. To determine whether such an effect also occurs in vivo, NF kappa B activity was analyzed in the cerebral cortex of the rat brain after whole body irradiation. After a single dose of 15 Gy, NF kappa B activity was increased by 2 h postirradiation, returning to unirradiated levels by 8 hours. The increase was dose-dependent beginning at 2 Gy and continuing to at least 22.5 Gy. NF kappa B activity in the irradiated cortex was not accompanied by I kappa B alpha degradation. When 7.5 Gy was delivered 24 h before the 15 Gy, the increase in NF kappa B activity after 15 Gy was significantly reduced. These results suggest that an initial exposure to radiation induced a refractory period in the brain during which the susceptibility of NF kappa B to activation by subsequent irradiation was significantly reduced. This period of reduced sensitivity to radiation was also apparent for the induction of the NF kappa B-regulated cytokines IL-1 beta, IL-6, and TNF alpha.
Collapse
Affiliation(s)
- U Raju
- Department of Experimental Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston 77030, USA
| | | | | |
Collapse
|
22
|
Kemler I, Fontana A. Role of I?B? and I?B? in the biphasic nuclear translocation of NF-?B in TNF?-stimulated astrocytes and in neuroblastoma cells. Glia 1999. [DOI: 10.1002/(sici)1098-1136(199905)26:3<212::aid-glia3>3.0.co;2-f] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
23
|
Tong W, Shah D, Xu J, Diehl JA, Hans A, Hannink M, Sun GY. Involvement of lipid mediators on cytokine signaling and induction of secretory phospholipase A2 in immortalized astrocytes (DITNC). J Mol Neurosci 1999; 12:89-99. [PMID: 10527453 DOI: 10.1007/bf02736923] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Our previous studies demonstrated the ability of proinflammatory cytokines, such as tumor necrosis factor-alpha (TNF-alpha) and interleukin 1beta (IL-1beta), to stimulate NFkappaB/DNA binding and synthesis of secretory phospholipase A2 (sPLA2) in immortalized astrocytes (DITNC). In this study, we examined possible involvement of lipid mediators in the cytokine action. Using [14C]serine to label sphingomyelin and ceramide in these cells, subsequent exposure of cells to cytokines did not result in alteration of sphingomyelin/ceramide ratio. Furthermore, neither exogenous sphingomyelinase nor cell-permeable ceramides could stimulate NFkappaB/DNA binding. On the other hand, C-2 ceramide (0.3 microM) as well as other lipid mediators, such as lysophosphatidylcholine and arachidonic acid, were able to elicit a small increase in sPLA2 and potentiate the induction of sPLA2 by TNF-alpha. When DITNC cells were prelabeled with [32P]Pi, an increase in labeled phosphatidic acid (PA) was observed on treatment of cells with IL-1beta (200 U/mL). However, despite the ability of phorbol myristate acetate (PMA) to stimulate phospholipase D (PLD) and synthesis of phosphatidylethanol (PEt) in these cells, PLD activity was not affected by IL-1beta. With the [32P]labeled cells, however, PA-phosphohydrolase inhibitors, such as chlorpromazine and propranolol, could elicit large increases in labeled PA, indicating active PA metabolism in these cells. Cytokines also caused an increase in levels of diacylglycerol (DG) in these cells, although the source of this lipid pool is presently not understood. Taken together, these results provide evidence for the participation of PA and DG in cytokine signaling activity. Furthermore, although cytokines did not cause the release of ceramide, lipid mediators, such as lysophospholipids, and AA could modulate cytokine-mediated induction of sPLA2 in astrocytes.
Collapse
Affiliation(s)
- W Tong
- Biochemistry Department, University of Missouri, Columbia 65212, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Walton M, Connor B, Lawlor P, Young D, Sirimanne E, Gluckman P, Cole G, Dragunow M. Neuronal death and survival in two models of hypoxic-ischemic brain damage. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 1999; 29:137-68. [PMID: 10209230 DOI: 10.1016/s0165-0173(98)00053-8] [Citation(s) in RCA: 126] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Two unilateral hypoxic-ischemia (HI) models (moderate and severe) in immature rat brain have been used to investigate the role of various transcription factors and related proteins in delayed neuronal death and survival. The moderate HI model results in an apoptotic-like neuronal death in selectively vulnerable regions of the brain while the more severe HI injury consistently produces widespread necrosis resulting in infarction, with some necrosis resistant cell populations showing evidence of an apoptotic type death. In susceptible regions undergoing an apoptotic-like death there was not only a prolonged induction of the immediate early genes, c-jun, c-fos and nur77, but also of possible target genes amyloid precursor protein (APP751) and CPP32. In contrast, increased levels of BDNF, phosphorylated CREB and PGHS-2 were found in cells resistant to the moderate HI insult suggesting that these proteins either alone or in combination may be of importance in the process of neuroprotection. An additional feature of both the moderate and severe brain insults was the rapid activation and/or proliferation of glial cells (microglia and astrocytes) in and around the site of damage. The glial response following HI was associated with an upregulation of both the CCAAT-enhancer binding protein alpha (microglia only) and NFkappaB transcription factors.
Collapse
Affiliation(s)
- M Walton
- Department of Pharmacology, Faculty of Medicine and Health Science, University of Auckland, Auckland, New Zealand
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Bonizzi G, Piette J, Schoonbroodt S, Merville MP, Bours V. Role of the protein kinase C lambda/iota isoform in nuclear factor-kappaB activation by interleukin-1beta or tumor necrosis factor-alpha: cell type specificities. Biochem Pharmacol 1999; 57:713-20. [PMID: 10037458 DOI: 10.1016/s0006-2952(98)00353-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
It has previously been reported that distinct signaling pathways can lead to nuclear factor (NF)-kappaB activation following stimulation of different cell types with inflammatory cytokines. As the role of atypical protein kinase C (PKC) isoforms in NF-kappaB activation remains a matter of controversy, we investigated whether this role might be cell type-dependent. Immunoblots detected atypical PKC expression in all the analyzed cell lines. The PKC inhibitor calphostin C inhibited NF-kappaB activation by tumor necrosis factor (TNF)-alpha or interleukin (IL)-1beta in Jurkat or NIH3T3 cells but not in MCF7 A/Z cells. Cell transfections with a PKC lambda/iota dominant negative mutant abolished TNF-alpha-induced NF-kappaB-dependent transcription in NIH3T3 and Jurkat cells but not in MCF7 A/Z cells. Similarly, the same mutant blocked NF-kappaB-dependent transactivation after IL-1beta stimulation of NIH3T3 cells, but was ineffective after IL-1beta treatment of MCF7 A/Z cells. In MCF7 A/Z cells, however, the PKC lambda/iota dominant negative mutant could abolish transactivation of an AP-1-dependent reporter plasmid after stimulation with TNF-alpha but not with IL-1beta. These data thus confirm that transduction pathways for NF-kappaB activation after cell stimulation with TNF-alpha or IL-1beta are cell-type specific and that atypical PKC isoforms participate in this pathway in NIH3T3 and Jurkat cells.
Collapse
Affiliation(s)
- G Bonizzi
- Laboratory of Medical Chemistry/Medical Oncology, University of Liège, Belgium
| | | | | | | | | |
Collapse
|
26
|
Li W, Xia J, Sun GY. Cytokine induction of iNOS and sPLA2 in immortalized astrocytes (DITNC): response to genistein and pyrrolidine dithiocarbamate. J Interferon Cytokine Res 1999; 19:121-7. [PMID: 10090397 DOI: 10.1089/107999099314261] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Using an immortalized astrocyte cell line (DITNC), we showed that lipopolysaccharide (LPS), tumor necrosis factor-alpha (TNF-alpha), and interleukin-1beta (IL-1beta) but not interferon-alpha (IFN-alpha) could individually induce secretory phospholipase A2 (sPLA2) mRNA and enzymatic activity. However, induction of inducible nitric oxide synthase (iNOS) mRNA and NO production by cytokines required the presence of IFN-gamma. Using a three-cytokine mixture (TNF-alpha, IL-1beta, and IFN-gamma) that could maximally induce both iNOS and sPLA2, the increase in these mRNA species reached a maximum by 4-8 h, followed by a decline up to 48 h. L-N6-(1-Iminoethyl)lysine acetate (L-NIL) inhibited cytokine-induced NO production with IC50 of 25 microM, but this compound did not affect iNOS mRNA. Furthermore, L-NIL exerted no effect on sPLA2 mRNA or sPLA2 activity. Pyrrolidine dithiocarbamate (PDTC), an inhibitor for NF-kappaB, was more effective in inhibiting iNOS mRNA and NO production than for sPLA2. Surprisingly, genistein inhibited both NO production and sPLA2 activity with IC50 of 72 microM and 88 microM, respectively. On the other hand, daidzein, a genistein analog lacking tyrosine kinase inhibitor activity, was not effective in inhibition of NO production at 250 microM. These results demonstrate distinct pathways for induction of iNOS and sPLA2 in DITNC cells by cytokines and shed new insight on transcriptional regulation for these two mRNA species.
Collapse
Affiliation(s)
- W Li
- Department of Biochemistry, University of Missouri-Columbia, 65212, USA
| | | | | |
Collapse
|
27
|
Wang D, Baldwin AS. Activation of nuclear factor-kappaB-dependent transcription by tumor necrosis factor-alpha is mediated through phosphorylation of RelA/p65 on serine 529. J Biol Chem 1998; 273:29411-6. [PMID: 9792644 DOI: 10.1074/jbc.273.45.29411] [Citation(s) in RCA: 291] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nuclear factor-kappaB (NF-kappaB) is an essential transcription factor in the control of expression of genes involved in immune and inflammatory responses. In unstimulated cells, NF-kappaB complexes are sequestered in the cytoplasm through interactions with IkappaBalpha and other IkappaB proteins. Extracellular stimuli that activate NF-kappaB, such as tumor necrosis factor alpha (TNFalpha), cause rapid phosphorylation of IkappaBalpha at serines 32 and 36. The inducible phosphorylation of IkappaBalpha is followed by its ubiquitination and degradation, allowing NF-kappaB complexes to translocate into the nucleus and to activate gene expression. Previously, it has been shown that TNFalpha as well as other stimuli also lead to the phosphorylation of the RelA/p65 subunit of NF-kappaB. In this report, we demonstrate that the TNFalpha-induced phosphorylation of the RelA/p65 subunit occurs on serine 529, which is in the C-terminal (TA1) transactivation domain. Accordingly, the TNFalpha-induced phosphorylation of Rel/p65 increases NF-kappaB transcriptional activity but does not affect nuclear translocation or DNA binding affinity.
Collapse
Affiliation(s)
- D Wang
- Lineberger Comprehensive Cancer Center and Department of Biology, CB 7295, University of North Carolina, Chapel Hill, North Carolina 27599-7295, USA.p6
| | | |
Collapse
|
28
|
Craxton A, Shu G, Graves JD, Saklatvala J, Krebs EG, Clark EA. p38 MAPK Is Required for CD40-Induced Gene Expression and Proliferation in B Lymphocytes. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.7.3225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
We have investigated the activation of the p38 MAPK pathway in response to CD40 engagement in multiple B cell lines and in human tonsillar B cells to define the role of p38 MAPK in proliferation, NF-κB activation and gene expression. Cross-linking CD40 rapidly stimulates both p38 MAPK and its downstream effector, MAPKAPK-2. Inhibition of p38 MAPK activity in vivo with the specific cell-permeable inhibitor, SB203580, under conditions that completely prevented MAPKAPK-2 activation, strongly perturbed CD40-induced tonsillar B cell proliferation while potentiating the B cell receptor (BCR)-driven proliferative response. SB203580 also significantly reduced expression of a reporter gene driven by a minimal promoter containing four NF-κB elements, indicating a requirement for the p38 MAPK pathway in CD40-induced NF-κB activation. However, CD40-mediated NF-κB binding was not affected by SB203580, suggesting that NF-κB may not be a direct target for the CD40-induced p38 MAPK pathway. In addition, SB203580 selectively reduced CD40-induced CD54/ICAM-1 expression, whereas CD40-dependent expression of CD40 and CD95/Fas and four newly defined CD40-responsive genes cIAP2, TRAF1, TRAF4/CART and DR3 were unaffected. Our observations show that the p38 MAPK pathway is required for CD40-induced proliferation and that CD40 induces gene expression via both p38 MAPK-dependent and -independent pathways.
Collapse
Affiliation(s)
| | | | | | | | - Edwin G. Krebs
- †Pharmacology, University of Washington, Seattle, WA 98195; and
| | | |
Collapse
|
29
|
Sachdev S, Hannink M. Loss of IkappaB alpha-mediated control over nuclear import and DNA binding enables oncogenic activation of c-Rel. Mol Cell Biol 1998; 18:5445-56. [PMID: 9710628 PMCID: PMC109129 DOI: 10.1128/mcb.18.9.5445] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The IkappaB alpha protein is able both to inhibit nuclear import of Rel/NF-kappaB proteins and to mediate the export of Rel/NF-kappaB proteins from the nucleus. We now demonstrate that the c-Rel-IkappaB alpha complex is stably retained in the cytoplasm in the presence of leptomycin B, a specific inhibitor of Crm1-mediated nuclear export. In contrast, leptomycin B treatment results in the rapid and complete relocalization of the v-Rel-IkappaB alpha complex from the cytoplasm to the nucleus. IkappaB alpha also mediates the rapid nuclear shuttling of v-Rel in an interspecies heterokaryon assay. Thus, continuous nuclear export is required for cytoplasmic retention of the v-Rel-IkappaB alpha complex. Furthermore, although IkappaB alpha is able to mask the c-Rel-derived nuclear localization sequence (NLS), IkappaB alpha is unable to mask the v-Rel-derived NLS in the context of the v-Rel-IkappaB alpha complex. Taken together, our results demonstrate that IkappaB alpha is unable to inhibit nuclear import of v-Rel. We have identified two amino acid differences between c-Rel and v-Rel (Y286S and L302P) which link the failure of IkappaB alpha to inhibit nuclear import and DNA binding of a mutant c-Rel protein to oncogenesis. Our results support a model in which loss of IkappaB alpha-mediated control over c-Rel leads to oncogenic activation of c-Rel.
Collapse
Affiliation(s)
- S Sachdev
- Biochemistry Department, University of Missouri-Columbia, Columbia, Missouri 65212, USA
| | | |
Collapse
|
30
|
Abstract
Inflammatory responses are a major component of secondary injury and play a central role in mediating the pathogenesis of acute and chronic spinal cord injury (SCI). The nuclear factor-kappaB (NF-kappaB) family of transcription factors is required for the transcriptional activation of a variety of genes regulating inflammatory, proliferative, and cell death responses of cells. In this study we examined the temporal and cellular expression of activated NF-kappaB after traumatic SCI. We used a contusion model (N.Y.U. Impactor) to initiate the early biochemical and molecular changes that occur after traumatic injury to reproduce the pathological events associated with acute inflammation after SCI. The activation and cellular distribution of activated NF-kappaB was evaluated by using a monoclonal antibody that selectively recognizes activated p65 in a NF-kappaB dimer. Immunohistochemical and Western blot analyses demonstrated that NF-kappaB activation occurred as early as 0.5 hr postinjury and persisted for at least 72 hr. Using electrophoretic mobility shift assays (EMSA), we demonstrate that NF-kappaB is activated after SCI. In our immunohistochemical, Western, and EMSA experiments there are detectable levels of activated NF-kappaB in our control animals. Using double-staining protocols, we detected activated NF-kappaB in macrophages/microglia, endothelial cells, and neurons within the injured spinal cord. Colocalization of activated NF-kappaB with the NF-kappaB-dependent gene product, inducible nitric oxide synthase (iNOS), suggests functional implications for this transcription factor in the pathogenesis of acute spinal cord injury. Although there is considerable evidence for the involvement of an inflammatory reaction after traumatic SCI, this is the first evidence for the activation of NF-kappaB after trauma. Strategies directed at blocking the initiation of this cascade may prove beneficial as a therapeutic approach for the treatment of acute SCI.
Collapse
|
31
|
Luque I, Gélinas C. Distinct domains of IkappaBalpha regulate c-Rel in the cytoplasm and in the nucleus. Mol Cell Biol 1998; 18:1213-24. [PMID: 9488436 PMCID: PMC108834 DOI: 10.1128/mcb.18.3.1213] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/1997] [Accepted: 12/05/1997] [Indexed: 02/06/2023] Open
Abstract
IkappaBalpha is a critical regulator of Rel/NF-KB-mediated gene activation. It controls the induction of NF-KB factors by retaining them in the cytoplasm and also functions in the nucleus to terminate the induction process. In this study, we show that IkappaBalpha regulates the transcriptional activity of c-Rel in the nuclear compartment. We also demonstrate that discrete functional domains of IkappaBalpha are responsible for the cytoplasmic and nuclear regulation of c-Rel. We show that the determinants for the cytoplasmic regulation of c-Rel reside in the N-terminal and central ankyrin regions of IkappaBalpha and that the N-terminal domain of IkappaBalpha is required to mask the c-Rel nuclear localization signal. Importantly, IkappaBalpha sequences necessary to regulate c-Rel in the nucleus map to its central ankyrin domain and to a few negatively charged amino acids that immediately follow in the C-terminal IkappaBalpha PEST domain. The mapping of the IkappaBalpha determinants that control the cytoplasmic and nuclear activities of c-Rel to specific regions of the molecule suggests that IkappaBalpha inhibitors could be designed to antagonize Rel/NF-kappaB activity in different subcellular compartments or at defined stages of activation.
Collapse
Affiliation(s)
- I Luque
- Center for Advanced Biotechnology and Medicine, University of Medicine and Dentistry of New Jersey, Piscataway 08854-5638, USA
| | | |
Collapse
|
32
|
Abstract
Nuclear Factor-Kappa B (NF-kappa B) was first identified by Sen and Baltimore (1986, Cell 46, 705-716) as a constitutively active transcription factor binding the kappa light chain immunoglobulin enhancer in B cells. Shortly afterwards, the same researchers found NF-kappa B to be present in other cell types in an inactive cytoplasmic form which upon cellular stimulation could be induced to translocate to the nucleus and bind DNA. Subsequently, it has been demonstrated that NF-kappa B performs a critical role as a regulator of the immune system, the response to stress, apoptosis, viral replication and is involved in many diseases, leading to it becoming one of the most intensively studied transcription factors of the last decade. The pivotal role played by NF-kappa B is illustrated not only by the great diversity of genes that it regulates, but also by the large variety of stimuli leading to its activation. This article will address how NF-kappa B, a ubiquitously expressed transcription factor composed of dimers formed from five subunits, differentially regulates the expression of such a diverse array of genes with different functions, in different cell types and at different times. Recent research indicates that this behavioral diversity arises from a delicately balanced network of protein: protein interactions: NF-kappa B activity is determined not only through its regulated nuclear localization but is also dependent on the cellular context in which it is found.
Collapse
Affiliation(s)
- N D Perkins
- Department of Biochemistry, University of Dundee, U.K
| |
Collapse
|
33
|
Wu JM, Sun GY. Effects of IL-1 beta on receptor-mediated poly-phosphoinositide signaling pathway in immortalized astrocytes (DITNC). Neurochem Res 1997; 22:1309-15. [PMID: 9342736 DOI: 10.1023/a:1021949417127] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Astrocytes are known to play multi-functional roles in support of many homeostatic mechanisms in the central nervous system including host defense mechanisms. Despite the ability of cytokines to alter gene expression and cellular activity, their effect on receptor-mediated poly-phosphoinositide (poly-PI) signaling pathway has not been examined in detail. In this study, an immortalized astrocyte cell line (DITNC) was used to test the effect of IL-1 beta exposure on the poly-PI signaling pathway. Similar to primary astrocytes, DITNC cells exhibit P2-purinergic receptor response to ATP and UTP leading to transient increases in inositol 1,4,5-trisphosphate [Ins(1,4,5)P3] and intracellular calcium concentration, [Ca2+]i. Upon exposure of DITNC cells to IL-1 beta (100 U/ml) for 24 hrs, an increased response to the poly-PI agonists was observed. The increase in ATP-mediated Ins(1,4,5)P3 release could not be attributed to a shift in the ATP dose or an alteration of the time profile for the release of Ins(1,4,5)P3. Since the increase in response required a lag time of 4 hr after IL-1 beta exposure, it is unlikely that this effect was due to a direct interaction of IL-1 beta with the purinergic receptor. On the other hand, an increase in ATP response could be observed in DITNC cells exposed to conditioned medium obtained after IL-1 beta treatment. It can be concluded that exposure of astrocytes to cytokines may lead to an increase in receptor-mediated poly-PI signaling activity and this may involve compounds secreted into the culture medium, e.g., the secretory phospholipase A2.
Collapse
Affiliation(s)
- J M Wu
- Biochemistry Department, University of Missouri, Columbia 65212, USA
| | | |
Collapse
|
34
|
Raju U, Lu R, Noel F, Gumin GJ, Tofilon PJ. Failure of a second X-ray dose to activate nuclear factor kappaB in normal rat astrocytes. J Biol Chem 1997; 272:24624-30. [PMID: 9305931 DOI: 10.1074/jbc.272.39.24624] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Induced gene expression and subsequent cytokine production have been implicated in the normal tissue injury response to radiotherapy. However, studies of radiation-induced gene expression have used single radiation doses rather than the fractionated exposures typical of the clinical situation. To study the effects of multiple radiation doses on gene expression, we investigated nuclear factor kappaB (NFkappaB) DNA binding activity in primary astrocyte cultures after one and two exposures to x-rays. After a single dose of x-rays (3.8-15 gray (Gy)), NFkappaB binding activity in astrocytes increased in a dose-dependent manner, reaching a maximum by 2-4 h and returning to control levels by 8 h after irradiation. In split-dose experiments, when an interval of 24 h was used between two doses of 7.5 Gy, the second 7.5-Gy exposure failed to induce NFkappaB activation. The period of desensitization induced by the first radiation exposure was dose-dependent, persisting approximately 72 h after 7.5 Gy compared with 24 h after 1.5 Gy. No changes in IkappaBalpha protein levels were detected. However, the presence of a transcription inhibitor prevented the desensitizing effect of the initial irradiation. Irradiation also prevented NFkappaB activation in astrocytes by a subsequent exposure to H2O2, but it had no effect on the activation induced by tumor necrosis factor-alpha. These data indicate that an initial x-ray exposure can desensitize astrocytes to the NFkappaB-activating effects of a subsequent radiation exposure. Furthermore, they suggest that this desensitization depends on gene transcription and may have some specificity for NFkappaB activation mediated by reactive oxygen species.
Collapse
Affiliation(s)
- U Raju
- Department of Experimental Radiation Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
35
|
Abstract
AbstractActivated neutrophils have the ability to upregulate the expression of many genes, in particular those encoding cytokines and chemokines, and to subsequently release the corresponding proteins. Although little is known to date concerning the regulation of gene transcription in neutrophils, it is noteworthy that many of these genes depend on the activation of transcription factors, such as NF-κB, for inducible expression. We therefore investigated whether NF-κB/Rel proteins are expressed in human neutrophils, as well as their fate on cell activation. We now report that dimers consisting of p50 NFκB1, p65 RelA, and/or c-Rel are present in neutrophils and that the greater part of these protein complexes is physically associated with cytoplasmic IκB-α in resting cells. Following neutrophil stimulation with proinflammatory agonists (such as lipopolysaccharide [LPS], tumor necrosis factor-α [TNF-α], and fMet-Leu-Phe) that induce the production of cytokines and chemokines in these cells, NF-κB/Rel proteins translocated to nuclear fractions, resulting in a transient induction of NF-κB DNA binding activity, as determined in gel mobility shift assays. The onset of both processes was found to be closely paralleled by, and dependent on, IκB-α degradation. Proinflammatory neutrophil stimuli also promoted the accumulation of IκB-α mRNA transcripts, resulting in the reexpression of the IκB-α protein. To our knowledge, this constitutes the first indication that NF-κB activation may underlie the action of proinflammatory stimuli towards human neutrophil gene expression and, as such, adds a new facet to our understanding of neutrophil biology.
Collapse
|
36
|
Park SK, Lin HL, Murphy S. Nitric oxide regulates nitric oxide synthase-2 gene expression by inhibiting NF-kappaB binding to DNA. Biochem J 1997; 322 ( Pt 2):609-13. [PMID: 9065784 PMCID: PMC1218233 DOI: 10.1042/bj3220609] [Citation(s) in RCA: 134] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Treatment of astroglial cells with interleukin 1beta and interferon gamma transcriptionally activates the nitric oxide synthase (NOS)-2 gene. The duration of mRNA expression is brief because of transcript instability. In addition, NO donors reduce the expression of NOS-2 mRNA dramatically by reducing the rate of transcription. In this study we observed that the NO donor, spermine NONOate did not inhibit the activation and translocation of NF-kappaB, a key transcription factor in the induction of NOS-2, but inhibited formation of the NF-kappaB-DNA complex. This effect was reversed by methaemoglobin (acting as an NO trap) and by the reducing agent dithiothreitol. Formation of the interferon-regulatory factor-DNA complex was unaffected by NO. These results suggest that NO can modulate its own production by interfering with NF-kappaB interaction with the promoter region of the NOS gene, a negative feedback effect that may be important for limiting NO production in vivo.
Collapse
Affiliation(s)
- S K Park
- Department of Pharmacology, University of Iowa College of Medicine, Iowa City 52242, U.S.A
| | | | | |
Collapse
|
37
|
Raj GV, Safak M, MacDonald GH, Khalili K. Transcriptional regulation of human polyomavirus JC: evidence for a functional interaction between RelA (p65) and the Y-box-binding protein, YB-1. J Virol 1996; 70:5944-53. [PMID: 8709216 PMCID: PMC190614 DOI: 10.1128/jvi.70.9.5944-5953.1996] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The transcriptional control region of the human neurotropic polyomavirus JC virus contains a consensus NF-kappa B site which has been shown to enhance both basal and extracellular stimulus-induced levels of transcription of JC promoters. Here, we show that the expression of JC late promoter constructs containing the NF-kappa B site is decreased by cotransfection with the NF-kappa B/rel subunits, p50 and p52, but enhanced by the p65 subunit. However, JC promoter constructs lacking the NF-kappa B site were activated by p52 and p50 and repressed by p65. This antithetical response of the JC promoter mapped specifically to the D domain, which is a target site for the cellular transcription factor, YB-1. Band shift studies indicated that YB-1 and p65 modulate each other's binding to DNA: YB-1 augments the affinity of p65 for the NF-kappa B site, while p65 reduces the binding of YB-1 to the D domain. Results from coimmunoprecipitation followed by Western blot (immunoblot) analysis suggest an in vivo interaction between p65 and YB-1 in glial cells. Functionally, YB-1 appears to act synergistically with p65 to control transcription from the NF-kappa B site. A converse pattern is seen with the D domain, in which YB-1 acts synergistically with p50 and p52 to regulate transcription. p50 and p52 may function as transcriptional activators on the D domain by removing the repressive effect of p65 on YB-1 binding to the D domain. On the basis of these data, we propose a model in which NF-kappa B/rel subunits functionally interact with consensus NF-kappa B sites or YB-1-binding sites, with disparate effects on eukaryotic gene expression.
Collapse
Affiliation(s)
- G V Raj
- Department of Biochemistry and Molecular Biology, Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | |
Collapse
|
38
|
Abstract
Interleukin-1 (IL-1) is primarily an inflammatory cytokine, although it is capable of mediating a wide variety of effects on many different cell types. Nearly every known signal transduction pathway has been reported to be activated in response to IL-1. However, the significance of many of these signaling events is unclear, due to the use of different and sometimes unique cell lines in studying IL-1-initiated signal transduction. Complicating matters further is the lack of association in many studies between identified IL-1-induced signals and subsequent biological responses. In this article, we review what is known about IL-1 receptor signaling and, whenever possible, correlate signaling events to biological responses.
Collapse
|
39
|
Abstract
The transcription factor NF-kappa B has attracted widespread attention among researchers in many fields based on the following: its unusual and rapid regulation, the wide range of genes that it controls, its central role in immunological processes, the complexity of its subunits, and its apparent involvement in several diseases. A primary level of control for NF-kappa B is through interactions with an inhibitor protein called I kappa B. Recent evidence confirms the existence of multiple forms of I kappa B that appear to regulate NF-kappa B by distinct mechanisms. NF-kappa B can be activated by exposure of cells to LPS or inflammatory cytokines such as TNF or IL-1, viral infection or expression of certain viral gene products, UV irradiation, B or T cell activation, and by other physiological and nonphysiological stimuli. Activation of NF-kappa B to move into the nucleus is controlled by the targeted phosphorylation and subsequent degradation of I kappa B. Exciting new research has elaborated several important and unexpected findings that explain mechanisms involved in the activation of NF-kappa B. In the nucleus, NF-kappa B dimers bind to target DNA elements and activate transcription of genes encoding proteins involved with immune or inflammation responses and with cell growth control. Recent data provide evidence that NF-kappa B is constitutively active in several cell types, potentially playing unexpected roles in regulation of gene expression. In addition to advances in describing the mechanisms of NF-kappa B activation, excitement in NF-kappa B research has been generated by the first report of a crystal structure for one form of NF-kappa B, the first gene knockout studies for different forms of NF-kB and of I kappa B, and the implications for therapies of diseases thought to involve the inappropriate activation of NF-kappa B.
Collapse
Affiliation(s)
- A S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill 27599, USA
| |
Collapse
|
40
|
Sancéau J, Kaisho T, Hirano T, Wietzerbin J. Triggering of the human interleukin-6 gene by interferon-gamma and tumor necrosis factor-alpha in monocytic cells involves cooperation between interferon regulatory factor-1, NF kappa B, and Sp1 transcription factors. J Biol Chem 1995; 270:27920-31. [PMID: 7499267 DOI: 10.1074/jbc.270.46.27920] [Citation(s) in RCA: 167] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
We investigated the molecular basis of the synergistic induction by interferon-gamma (IFN-gamma)/tumor necrosis factor-alpha (TNF-alpha) of human interleukin-6 (IL-6) gene in THP-1 monocytic cells, and compared it with the basis of this induction by lipopolysaccharide (LPS). Functional studies with IL-6 promoter demonstrated that three regions are the targets of the IFN-gamma and/or TNF-alpha action, whereas only one of these regions seemed to be implicated in LPS activation. The three regions concerned are: 1) a region between -73 and -36, which is the minimal element inducible by LPS or TNF-alpha; 2) an element located between -181 and -73, which appeared to regulate the response to IFN-gamma and TNF-alpha negatively; and 3) a distal element upstream of -224, which was inducible by IFN-gamma alone. LPS signaling was found to involve NF kappa B activation by the p50/p65 heterodimers. Synergistic induction of the IL-6 gene by IFN-gamma and TNF-alpha, in monocytic cells, involved cooperation between the IRF-1 and NF kappa B p65 homodimers with concomitant removal of the negative effect of the retinoblastoma control element present in the IL-6 promoter. This removal occurred by activation of the constitutive Sp1 factor, whose increased binding activity and phosphorylation were mediated by IFN-gamma.
Collapse
Affiliation(s)
- J Sancéau
- INSERM, U365, Interferons et Cytokines, Institut Curie, Section de Recherches, Paris, France
| | | | | | | |
Collapse
|
41
|
Guerrini L, Blasi F, Denis-Donini S. Synaptic activation of NF-kappa B by glutamate in cerebellar granule neurons in vitro. Proc Natl Acad Sci U S A 1995; 92:9077-81. [PMID: 7568076 PMCID: PMC40927 DOI: 10.1073/pnas.92.20.9077] [Citation(s) in RCA: 225] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Neuronal proliferation, migration, and differentiation are regulated by the sequential expression of particular genes at specific stages of development. Such processes rely on differential gene expression modulated through second-messenger systems. Early postnatal mouse cerebellar granule cells migrate into the internal granular layer and acquire differentiated properties. The neurotransmitter glutamate has been shown to play an important role in this developmental process. We show here by immunohistochemistry that the RelA subunit of the transcription factor NF-kappa B is present in several areas of the mouse brain. Moreover, immunofluorescence microscopy and electrophoretic mobility-shift assay demonstrate that in cerebellar granule cell cultures derived from 3- to 7-day-old mice, glutamate specifically activates the transcription factor NF-kappa B, as shown by binding of nuclear extract proteins to a synthetic oligonucleotide reproducing the kappa B site of human immunodeficiency virus. The use of different antagonists of the glutamate recpetors indicates that the effect of glutamate occurs mainly via N-methyl-D-aspartate (NMDA)-receptor activation, possibly as a result of an increase in intracellular Ca2+. The synaptic specificity of the effect is strongly suggested by the observation that glutamate failed to activate NF-kappa B in astrocytes, while cytokines, such as interleukin 1 alpha and tumor necrosis factor alpha, did so. The effect of glutamate appears to be developmentally regulated. Indeed, NF-kappa B is found in an inducible form in the cytoplasm of neurons of 3- to 7-day-old mice but is constitutively activated in the nuclei of neurons derived from older pups (8-10 days postnatal). Overall, these observations suggest the existence of a new pathway of trans-synaptic regulation of gene expression.
Collapse
Affiliation(s)
- L Guerrini
- Department of Genetics and Microbial Biology, University of Milan, Italy
| | | | | |
Collapse
|
42
|
Finco TS, Baldwin AS. Mechanistic aspects of NF-kappa B regulation: the emerging role of phosphorylation and proteolysis. Immunity 1995; 3:263-72. [PMID: 7552992 DOI: 10.1016/1074-7613(95)90112-4] [Citation(s) in RCA: 320] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- T S Finco
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill 27599, USA
| | | |
Collapse
|
43
|
Tong W, Hu ZY, Sun GY. Stimulation of group II phospholipase A2 mRNA expression and release in an immortalized astrocyte cell line (DITNC) by LPS, TNF alpha, and IL-1 beta. Interactive effects. MOLECULAR AND CHEMICAL NEUROPATHOLOGY 1995; 25:1-17. [PMID: 7546015 DOI: 10.1007/bf02815083] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Astrocytes are immunoactive cells in brain and have been implicated in the defense mechanism in response to external injury. Previous studies using cultured glial cells indicated the ability of astrocytes to respond to bacteria endotoxin and cytokines, resulting in the release of phospholipase A2. In this study, we examined the interactive effects of lipopolysaccharides (LPS), interleukin 1 beta (IL-1 beta) and tumor necrosis factor (TNF alpha) to stimulate phospholipase A2 (PLA2) in an immortalized astrocyte cell line (DITNC) with many properties of type I astrocytes. Northern blot analysis using oligonucleotide probes derived from the cDNA encoding the rat spleen group II PLA2 indicated the ability of DITNC cells to respond to all three factors in the induction of gene expression and the release of PLA2. After an initial lag time of 2 h, PLA2 release was proportional to time, reaching a plateau by 12 h. This event occurred at a time period preceding any signs of cell death. Cycloheximide at 1.25 microM completely inhibited cytokine-induced PLA2 release. When suboptimal amounts of TNF alpha were added to the DITNC culture together with IL-1 beta or LPS, a synergistic increase in the induction of PLA2 release could be observed. On the other hand, combination of IL-1 beta and LPS resulted only in an additive increase in PLA2 release. Antibodies to IL-1 beta and TNF alpha completely neutralized the effects of these two agents on PLA2 release. However, neither antibody was able to inhibit the PLA2 release induced by LPS, suggesting that the effect of LPS was not complicated by the release of IL-1 beta or TNF alpha. Taken together, results show that the immortalized astrocyte cell line (DITNC) can be used for studies to elucidate the molecular mechanism underlying the cytokine signaling cascade and subsequent induction of PLA2 synthesis.
Collapse
Affiliation(s)
- W Tong
- Biochemistry Department, University of Missouri, Columbia 65212, USA
| | | | | |
Collapse
|