1
|
Ehle C, Iyer-Bierhoff A, Wu Y, Xing S, Kiehntopf M, Mosig AS, Godmann M, Heinzel T. Downregulation of HNF4A enables transcriptomic reprogramming during the hepatic acute-phase response. Commun Biol 2024; 7:589. [PMID: 38755249 PMCID: PMC11099168 DOI: 10.1038/s42003-024-06288-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 05/03/2024] [Indexed: 05/18/2024] Open
Abstract
The hepatic acute-phase response is characterized by a massive upregulation of serum proteins, such as haptoglobin and serum amyloid A, at the expense of liver homeostatic functions. Although the transcription factor hepatocyte nuclear factor 4 alpha (HNF4A) has a well-established role in safeguarding liver function and its cistrome spans around 50% of liver-specific genes, its role in the acute-phase response has received little attention so far. We demonstrate that HNF4A binds to and represses acute-phase genes under basal conditions. The reprogramming of hepatic transcription during inflammation necessitates loss of HNF4A function to allow expression of acute-phase genes while liver homeostatic genes are repressed. In a pre-clinical liver organoid model overexpression of HNF4A maintained liver functionality in spite of inflammation-induced cell damage. Conversely, HNF4A overexpression potently impaired the acute-phase response by retaining chromatin at regulatory regions of acute-phase genes inaccessible to transcription. Taken together, our data extend the understanding of dual HNF4A action as transcriptional activator and repressor, establishing HNF4A as gatekeeper for the hepatic acute-phase response.
Collapse
Affiliation(s)
- Charlotte Ehle
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena, 07745, Jena, Germany
| | - Aishwarya Iyer-Bierhoff
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena, 07745, Jena, Germany
| | - Yunchen Wu
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena, 07745, Jena, Germany
- Marshall Laboratory of Biomedical Engineering, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Shaojun Xing
- Marshall Laboratory of Biomedical Engineering, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Michael Kiehntopf
- Department of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, 07747, Jena, Germany
| | - Alexander S Mosig
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
| | - Maren Godmann
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena, 07745, Jena, Germany
| | - Thorsten Heinzel
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena, 07745, Jena, Germany.
| |
Collapse
|
2
|
Paraiso IL, Tran TQ, Magana AA, Kundu P, Choi J, Maier CS, Bobe G, Raber J, Kioussi C, Stevens JF. Xanthohumol ameliorates Diet-Induced Liver Dysfunction via Farnesoid X Receptor-Dependent and Independent Signaling. Front Pharmacol 2021; 12:643857. [PMID: 33959012 PMCID: PMC8093804 DOI: 10.3389/fphar.2021.643857] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/22/2021] [Indexed: 12/20/2022] Open
Abstract
The farnesoid X receptor (FXR) plays a critical role in the regulation of lipid and bile acid (BA) homeostasis. Hepatic FXR loss results in lipid and BA accumulation, and progression from hepatic steatosis to nonalcoholic steatohepatitis (NASH). This study aimed to evaluate the effects of xanthohumol (XN), a hop-derived compound mitigating metabolic syndrome, on liver damage induced by diet and FXR deficiency in mice. Wild-type (WT) and liver-specific FXR-null mice (FXRLiver−/−) were fed a high-fat diet (HFD) containing XN or the vehicle formation followed by histological characterization, lipid, BA and gene profiling. HFD supplemented with XN resulted in amelioration of hepatic steatosis and decreased BA concentrations in FXRLiver−/− mice, the effect being stronger in male mice. XN induced the constitutive androstane receptor (CAR), pregnane X receptor (PXR) and glucocorticoid receptor (GR) gene expression in the liver of FXRLiver−/− mice. These findings suggest that activation of BA detoxification pathways represents the predominant mechanism for controlling hydrophobic BA concentrations in FXRLiver−/− mice. Collectively, these data indicated sex-dependent relationship between FXR, lipids and BAs, and suggest that XN ameliorates HFD-induced liver dysfunction via FXR-dependent and independent signaling.
Collapse
Affiliation(s)
- Ines L Paraiso
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States.,Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| | - Thai Q Tran
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| | - Armando Alcazar Magana
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States.,Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States.,Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Payel Kundu
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| | - Jaewoo Choi
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Claudia S Maier
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Gerd Bobe
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States.,Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, United States
| | - Jacob Raber
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States.,Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States.,Department of Neurology, Psychiatry and Radiation Medicine, Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Portland, OR, United States
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| | - Jan F Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States.,Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| |
Collapse
|
3
|
Gao Z, Ruan J. Computational modeling of in vivo and in vitro protein-DNA interactions by multiple instance learning. Bioinformatics 2018; 33:2097-2105. [PMID: 28334224 DOI: 10.1093/bioinformatics/btx115] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 02/27/2017] [Indexed: 12/25/2022] Open
Abstract
Motivation The study of transcriptional regulation is still difficult yet fundamental in molecular biology research. While the development of both in vivo and in vitro profiling techniques have significantly enhanced our knowledge of transcription factor (TF)-DNA interactions, computational models of TF-DNA interactions are relatively simple and may not reveal sufficient biological insight. In particular, supervised learning based models for TF-DNA interactions attempt to map sequence-level features ( k -mers) to binding event but usually ignore the location of k -mers, which can cause data fragmentation and consequently inferior model performance. Results Here, we propose a novel algorithm based on the so-called multiple-instance learning (MIL) paradigm. MIL breaks each DNA sequence into multiple overlapping subsequences and models each subsequence separately, therefore implicitly takes into consideration binding site locations, resulting in both higher accuracy and better interpretability of the models. The result from both in vivo and in vitro TF-DNA interaction data show that our approach significantly outperform conventional single-instance learning based algorithms. Importantly, the models learned from in vitro data using our approach can predict in vivo binding with very good accuracy. In addition, the location information obtained by our method provides additional insight for motif finding results from ChIP-Seq data. Finally, our approach can be easily combined with other state-of-the-art TF-DNA interaction modeling methods. Availability and Implementation http://www.cs.utsa.edu/∼jruan/MIL/. Contact jianhua.ruan@utsa.edu. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Zhen Gao
- Department of Computer Science, University of Texas at San Antonio, San Antonio, TX, USA
| | - Jianhua Ruan
- Department of Computer Science, University of Texas at San Antonio, San Antonio, TX, USA
| |
Collapse
|
4
|
Lerrer B, Gertler AA, Cohen HY. The complex role of SIRT6 in carcinogenesis. Carcinogenesis 2015; 37:108-18. [PMID: 26717993 DOI: 10.1093/carcin/bgv167] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 11/25/2015] [Indexed: 12/28/2022] Open
Abstract
SIRT6, a member of the mammalian sirtuins family, functions as a mono-ADP-ribosyl transferase and NAD(+)-dependent deacylase of both acetyl groups and long-chain fatty acyl groups. SIRT6 regulates diverse cellular functions such as transcription, genome stability, telomere integrity, DNA repair, inflammation and metabolic related diseases such as diabetes, obesity and cancer. In this review, we will discuss the implication of SIRT6 in the biology of cancer and the relevance to organism homeostasis and lifespan.
Collapse
Affiliation(s)
- Batia Lerrer
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Asaf A Gertler
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Haim Y Cohen
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| |
Collapse
|
5
|
Yamaguchi J, Tanaka T, Eto N, Nangaku M. Inflammation and hypoxia linked to renal injury by CCAAT/enhancer-binding protein δ. Kidney Int 2015; 88:262-75. [PMID: 25692954 PMCID: PMC4526770 DOI: 10.1038/ki.2015.21] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Revised: 12/08/2014] [Accepted: 12/11/2014] [Indexed: 12/11/2022]
Abstract
Tubulointerstitial hypoxia plays a critical role in the pathogenesis of kidney injury, and hypoxia-inducible factor (HIF)-1 is a master regulator of cellular adaptation to hypoxia. Aside from oxygen molecules, factors that modify HIF-1 expression and functional operation remain obscure. Therefore, we sought to identify novel HIF-1-regulating genes in kidney. A short-hairpin RNA library consisting of 150 hypoxia-inducible genes was derived from a microarray analysis of the rat renal artery stenosis model screened for the effect on HIF-1 response. We report that CCAAT/enhancer-binding protein δ (CEBPD), a transcription factor and inflammatory response gene, is a novel HIF-1 regulator in kidney. CEBPD was induced in the nuclei of tubular epithelial cells in both acute and chronic hypoxic kidneys. In turn, CEBPD induction augmented HIF-1α expression and its transcriptional activity. Mechanistically, CEBPD directly bound to the HIF-1α promoter and enhanced its transcription. Notably, CEBPD was rapidly induced by inflammatory cytokines, such as IL-1β in a nuclear factor-κB-dependent manner, which not only increased HIF-1α expression during hypoxia, but was also indispensable for the non-hypoxic induction of HIF-1α. Thus our study provides novel insight into HIF-1 regulation in tubular epithelial cells and offers a potential hypoxia and inflammation link relevant in both acute and chronic kidney diseases.
Collapse
Affiliation(s)
- Junna Yamaguchi
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tetsuhiro Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | | | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Abstract
Serum amyloid A (SAA), a protein originally of interest primarily to investigators focusing on AA amyloidogenesis, has become a subject of interest to a very broad research community. SAA is still a major amyloid research topic because AA amyloid, for which SAA is the precursor, is the prototypic model of in vivo amyloidogenesis and much that has been learned with this model has been applicable to much more common clinical types of amyloid. However, SAA has also become a subject of considerable interest to those studying (i) the synthesis and regulation of acute phase proteins, of which SAA is a prime example, (ii) the role that SAA plays in tissue injury and inflammation, a situation in which the plasma concentration of SAA may increase a 1000-fold, (iii) the influence that SAA has on HDL structure and function, because during inflammation the majority of SAA is an apolipoprotein of HDL, (iv) the influence that SAA may have on HDL's role in reverse cholesterol transport, and therefore, (v) SAA's potential role in atherogenesis. However, no physiological role for SAA, among many proposed, has been widely accepted. None the less from an evolutionary perspective SAA must have a critical physiological function conferring survival-value because SAA genes have existed for at least 500 million years and SAA's amino acid sequence has been substantially conserved. An examination of the published literature over the last 40 years reveals a great deal of conflicting data and interpretation. Using SAA's conserved amino acid sequence and the physiological effects it has while in its native structure, namely an HDL apolipoprotein, we argue that much of the confounding data and interpretation relates to experimental pitfalls not appreciated when working with SAA, a failure to appreciate the value of physiologic studies done in the 1970-1990 and a current major focus on putative roles of SAA in atherogenesis and chronic disease. When viewed from an evolutionary perspective, published data suggest that acute-phase SAA is part of a systemic response to injury to recycle and reuse cholesterol from destroyed and damaged cells. This is accomplished through SAA's targeted delivery of HDL to macrophages, and its suppression of ACAT, the enhancement of neutral cholesterol esterase and ABC transporters in macrophages. The recycling of cholesterol during serious injury, when dietary intake is restricted and there is an immediate and critical requirement of cholesterol in the generation of myriads of cells involved in inflammation and repair responses, is likely SAA's important survival role. Data implicating SAA in atherogenesis are not relevant to its evolutionary role. Furthermore, in apoE(-/-) mice, domains near the N- and C- termini of SAA inhibit the initiation and progression of aortic lipid lesions illustrating the conflicting nature of these two sets of data.
Collapse
|
7
|
The transcription factor C/EBP delta has anti-apoptotic and anti-inflammatory roles in pancreatic beta cells. PLoS One 2012; 7:e31062. [PMID: 22347430 PMCID: PMC3275575 DOI: 10.1371/journal.pone.0031062] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 01/01/2012] [Indexed: 12/31/2022] Open
Abstract
In the course of Type 1 diabetes pro-inflammatory cytokines (e.g., IL-1β, IFN-γ and TNF-α) produced by islet-infiltrating immune cells modify expression of key gene networks in β-cells, leading to local inflammation and β-cell apoptosis. Most known cytokine-induced transcription factors have pro-apoptotic effects, and little is known regarding “protective” transcription factors. To this end, we presently evaluated the role of the transcription factor CCAAT/enhancer binding protein delta (C/EBPδ) on β-cell apoptosis and production of inflammatory mediators in the rat insulinoma INS-1E cells, in purified primary rat β-cells and in human islets. C/EBPδ is expressed and up-regulated in response to the cytokines IL-1β and IFN-γ in rat β-cells and human islets. Small interfering RNA-mediated C/EBPδ silencing exacerbated IL-1β+IFN-γ-induced caspase 9 and 3 cleavage and apoptosis in these cells. C/EBPδ deficiency increased the up-regulation of the transcription factor CHOP in response to cytokines, enhancing expression of the pro-apoptotic Bcl-2 family member BIM. Interfering with C/EBPδ and CHOP or C/EBPδ and BIM in double knockdown approaches abrogated the exacerbating effects of C/EBPδ deficiency on cytokine-induced β-cell apoptosis, while C/EBPδ overexpression inhibited BIM expression and partially protected β-cells against IL-1β+IFN-γ-induced apoptosis. Furthermore, C/EBPδ silencing boosted cytokine-induced production of the chemokines CXCL1, 9, 10 and CCL20 in β-cells by hampering IRF-1 up-regulation and increasing STAT1 activation in response to cytokines. These observations identify a novel function of C/EBPδ as a modulatory transcription factor that inhibits the pro-apoptotic and pro-inflammatory gene networks activated by cytokines in pancreatic β-cells.
Collapse
|
8
|
Williams PJ, Nishu K, Rahman MM. HDAC inhibitor trichostatin A suppresses osteoclastogenesis by upregulating the expression of C/EBP-β and MKP-1. Ann N Y Acad Sci 2012; 1240:18-25. [PMID: 22172035 DOI: 10.1111/j.1749-6632.2011.06286.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Histone deacetylases (HDACs) remove the acetyl groups from the lysine residues of histone tails, leading to the formation of a condensed and transcriptionally silenced chromatin. HDAC inhibitors (HDACi) block this action and can result in hyperacetylation of histones, leading to a less compact and more transcriptionally active chromatin and thereby, gene expression. Previously, we have shown that HDACi inhibit osteoclast differentiation. However, which genes are transcriptionally activated following hyperacetylation of histones, and lead to the suppression of osteoclastogenesis, has yet to be elucidated. In this study, we show that an HDACi, trichostatin A (TSA), inhibits the receptor activator of the nuclear factor-κB (NF-κB) ligand (RANKL)-stimulated TNF-α production, NF-κB activation, and bone resorbing pit formation, and downregulates c-Fos and NFATc1 in RAW 264.7 cells. Interestingly, expression of antiosteoclastogenic factors CCAAT enhancer binding protein (C/EBP)-β and mitogen-activated protein kinase phosphatase (MKP)-1 was significantly upregulated in TSA-treated, RANKL-stimulated RAW 264.7 cells. These findings suggest that TSA upregulates the expression of C/EBP-β and MKP-1, which may downregulate pro-osteoclastogenic factors and signaling molecules, ultimately suppressing osteoclastogenesis.
Collapse
Affiliation(s)
- Paul J Williams
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | | | | |
Collapse
|
9
|
Khanjani S, Terzidou V, Lee YS, Thornton S, Johnson MR, Bennett PR. Synergistic Regulation of Human Oxytocin Receptor Promoter by CCAAT/ Enhancer-Binding Protein and RELA1. Biol Reprod 2011; 85:1083-8. [DOI: 10.1095/biolreprod.111.092304] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
10
|
Zhou X, Wang L, Feng H, Guo Q, Dai H. Acute phase response in Chinese soft-shelled turtle (Trionyx sinensis) with Aeromonas hydrophila infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2011; 35:441-451. [PMID: 21095204 DOI: 10.1016/j.dci.2010.11.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 11/12/2010] [Accepted: 11/14/2010] [Indexed: 05/30/2023]
Abstract
Chinese soft-shelled turtle (Trionyx sinensis) is an important culture reptile. However, little is known about its acute phase response (APR) caused by bacteria. Serum amyloid A (SAA) is a major acute phase protein (APP). In this study, a turtle SAA homologue was identified and described in reptiles. The full-length cDNA of turtle SAA was 554 bp and contained a 381 bp open reading frame (ORF) coding for a protein of 127 aa. Similar to other known SAA genes, the turtle SAA gene contained three exons and two introns. The promoter region of turtle SAA gene contained the consensus binding sites for nuclear factor (NF)-κB and c-Rel. The turtle SAA amino acid sequence shared the highest identity to avian SAA sequences. Meantime, we present the first systematic study with expression levels of five genes encoding APPs in immune response caused by Aeromonas hydrophila infection. After infection, turtle SAA mRNA was induced in liver at 8h, then increased more than 1200-fold at 2d; in spleen and kidney, the SAA mRNAs were also induced during 8h-7d, but the level was far lower than that in the liver. The complement 3 (C3), fibrinogen-gamma chain (Fb-G) and cathepsin L (CathL) mRNAs were increased in liver at 2d, whereas the albumin (ALB) mRNA was significantly decreased during 8h-7d. Our studies suggest that the APR in turtle with A. hydrophila infection is similar to that in mammals, and SAA is a major indicator of bacterial infection, especially at early stage, in reptiles. Additionally, the different expression patterns of five APP genes observed in present studies could provide clues for understanding the innate immune mechanisms in the APR of reptiles.
Collapse
Affiliation(s)
- Xiuxia Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | | | | | | | | |
Collapse
|
11
|
Mukhopadhyay S, Mukherjee S, Ray BK, Ray A, Stone WL, Das SK. Antioxidant liposomes protect against CEES-induced lung injury by decreasing SAF-1/MAZ-mediated inflammation in the guinea pig lung. J Biochem Mol Toxicol 2010; 24:187-94. [DOI: 10.1002/jbt.20329] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
12
|
Ejarque-Ortiz A, Gresa-Arribas N, Straccia M, Mancera P, Solà C, Tusell JM, Serratosa J, Saura J. CCAAT/enhancer binding protein delta in microglial activation. J Neurosci Res 2010; 88:1113-23. [PMID: 19908286 DOI: 10.1002/jnr.22272] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The transcription factor CCAAT/enhancer binding protein delta (C/EBP delta) regulates transcription of genes that play important roles in glial activation. Previous studies have shown the astroglial expression of C/EBP delta but the microglial expression of C/EBP delta remains virtually unexplored, with the exception of two microarray studies. In this report, using murine primary cultures and BV2 cells we clearly demonstrate that C/EBP delta is expressed by microglia and it is upregulated in microglial activation. Lipopolysaccharide upregulates C/EBP delta both in microglia and in astrocytes. This effect is time-dependent, with a maximum effect at 3 hr at mRNA level and at 4-8 hr at protein level, and concentration-dependent, with a maximum effect at 100 ng/mL. The lipopolysaccharide-induced C/EBP delta upregulation in BV2 microglia is mimicked by agonists of the toll-like receptors 2, 3 and 9 and can be prevented by an inhibitor of extracellular signal-regulated kinase activation. C/EBP delta from activated BV2 microglia binds to the cyclooxygenase-2 promoter and forms complexes with C/EBP beta isoforms. These results point to C/EBP delta as a putative key regulator of proinflammatory gene expression in microglial activation.
Collapse
Affiliation(s)
- Aroa Ejarque-Ortiz
- Department of Cerebral Ischaemia and Neurodegeneration, IIBB, CSIC, IDIBAPS, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Toll-like receptor signaling in bony fish. Vet Immunol Immunopathol 2010; 134:139-50. [DOI: 10.1016/j.vetimm.2009.09.021] [Citation(s) in RCA: 326] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Revised: 09/18/2009] [Accepted: 09/22/2009] [Indexed: 01/08/2023]
|
14
|
Feingold KR, Shigenaga JK, Patzek SM, Chui LG, Moser A, Grunfeld C. Endotoxin, zymosan, and cytokines decrease the expression of the transcription factor, carbohydrate response element binding protein, and its target genes. Innate Immun 2010; 17:174-82. [PMID: 20100709 DOI: 10.1177/1753425909357578] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Carbohydrate response element binding protein (ChREBP) is a recently discovered transcription factor whose levels and activity are increased by glucose leading to the activation of target genes, which include acetyl-CoA carboxylase, fatty acid synthase, and liver-type pyruvate kinase. Here, we demonstrate that lipopolysaccharide (LPS) treatment causes a marked decrease in ChREBP mRNA and protein levels in the liver of mice fed a normal chow diet or in mice fasted for 24 h and then re-fed a high carbohydrate diet. This decrease occurs rapidly and is a sensitive response (half-maximal dose 0.1 μg/mouse). The decrease in ChREBP is accompanied by a decrease in the expression of ChREBP target genes. Zymosan and turpentine treatment also decrease hepatic ChREBP levels and the expression of its target genes. Additionally, tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1β) decrease liver ChREBP expression both in vivo and in Hep3B cells in culture. Finally, LPS decreased ChREBP expression in muscle and adipose tissue. These studies demonstrate that ChREBP is down-regulated during the acute phase response resulting in alterations in the expression of ChREBP regulated target genes. Thus, ChREBP joins a growing list of transcription factors that are regulated during the acute phase response.
Collapse
Affiliation(s)
- Kenneth R Feingold
- Metabolism Section, Department of Veterans Affairs Medical Center, University of California-San Francisco, 4150 Clement Street, San Francisco, CA 94121, USA.
| | | | | | | | | | | |
Collapse
|
15
|
Sullivan CP, Seidl SE, Rich CB, Raymondjean M, Schreiber BM. Secretory phospholipase A2, group IIA is a novel serum amyloid A target gene: activation of smooth muscle cell expression by an interleukin-1 receptor-independent mechanism. J Biol Chem 2009; 285:565-75. [PMID: 19850938 DOI: 10.1074/jbc.m109.070565] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Atherosclerosis is a multifactorial vascular disease characterized by formation of inflammatory lesions. Elevated circulating acute phase proteins indicate disease risk. Serum amyloid A (SAA) is one such marker but its function remains unclear. To determine the role of SAA on aortic smooth muscle cell gene expression, a preliminary screen of a number of genes was performed and a strong up-regulation of expression of secretory phospholipase A(2), group IIA (sPLA(2)) was identified. The SAA-induced increase in sPLA(2) was validated by real time PCR, Western blot analysis, and enzyme activity assays. Demonstrating that SAA increased expression of sPLA(2) heteronuclear RNA and that inhibiting transcription eliminated the effect of SAA on sPLA(2) mRNA suggested that the increase was transcriptional. Transient transfections and electrophoretic mobility shift assays identified CAAT enhancer-binding protein (C/EBP) and nuclear factor kappaB (NFkappaB) as key regulatory sites mediating the induction of sPLA(2). Moreover, SAA activated the inhibitor of NF-kappaB kinase (IKK) in cultured smooth muscle cells. Previous reports showed that interleukin (IL)-1beta up-regulates Pla2g2a gene transcription via C/EBPbeta and NFkappaB. Interestingly, SAA activated smooth muscle cell IL-1beta mRNA expression, however, blocking IL-1 receptors had no effect on SAA-mediated activation of sPLA(2) expression. Thus, the observed changes in sPLA(2) expression were not secondary to SAA-induced IL-1 receptor activation. The association of SAA with high density lipoprotein abrogated the SAA-induced increase in sPLA(2) expression. These data suggest that during atherogenesis, SAA can amplify the involvement of smooth muscle cells in vascular inflammation and that this can lead to deposition of sPLA(2) and subsequent local changes in lipid homeostasis.
Collapse
Affiliation(s)
- Christopher P Sullivan
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | |
Collapse
|
16
|
Rebl A, Goldammer T, Fischer U, Köllner B, Seyfert HM. Characterization of two key molecules of teleost innate immunity from rainbow trout (Oncorhynchus mykiss): MyD88 and SAA. Vet Immunol Immunopathol 2009; 131:122-6. [DOI: 10.1016/j.vetimm.2009.03.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Revised: 02/24/2009] [Accepted: 03/06/2009] [Indexed: 01/12/2023]
|
17
|
Talbot AT, Smith TJ, Cairns MT. Characterisation of the differentially regulated trout protein 1 (DRTP1) gene in rainbow trout (Oncorhynchus mykiss). FISH & SHELLFISH IMMUNOLOGY 2009; 26:589-598. [PMID: 18992823 DOI: 10.1016/j.fsi.2008.09.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2007] [Revised: 09/10/2008] [Accepted: 09/18/2008] [Indexed: 05/27/2023]
Abstract
Increased levels of differentially regulated trout protein 1 (DRTP1) mRNA transcripts have been reported in fish after activation of the acute phase response. While the function of the DRTP1 protein still remains to be elucidated, this study focused on the genomic organisation of the gene, the quantification of the DRTP1 transcript in various tissues, and the isolation and analysis of the 5' regulatory region of the DRTP1 gene in rainbow trout (Oncorhynchus mykiss). Analysis of the DRTP1 genomic and cDNA sequences showed the gene to consist of four exons separated by three introns. Tissue localisation of the DRTP1 gene was performed by Northern analysis and validated by quantitative real-time PCR (qPCR). Six tissues (liver, intestine, spleen, brain, pituitary, and hypothalamus) were analysed. The tissues with the most abundant transcripts were the liver and the pituitary, with lesser amounts detected in the intestine, hypothalamus, brain and spleen. Genome walking allowed the isolation of a 934 bp sequence of the 5' regulatory region of the gene which was cloned, sequenced and in which potential transcription factor binding sites were identified. Promoter fragments of decreasing size were generated and transiently transfected into the human hepatoma cell line (HepG2). Inducibility of the promoter was determined by stimulation of the HepG2 cells containing the constructs with dexamethasone, polyinosinic:polycytidylic acid (poly I:C) and lipopolysaccharide (LPS) and tumor necrosis factor-alpha (TNFalpha). One construct, containing two potential C-EBP/beta sites and two NF-kappaB sites, exhibited the highest promoter induction (6.34 fold +/- SEM 0.5) when stimulated with human TNFalpha. A slightly shorter fragment containing one C-EBP/beta site and one NF-kappaB site did not show any significant inducibility when treated with TNFalpha. The loss of the C-EBP/beta and NF-kappaB in the shorter construct suggests that these sites, either individually or in combination, are critical for the induction of the DRTP1 promoter by TNFalpha.
Collapse
Affiliation(s)
- Anita T Talbot
- National Diagnostics Centre, National University of Ireland, Galway, Ireland.
| | | | | |
Collapse
|
18
|
Kumar D, Ray A, Ray BK. Transcriptional Synergy Mediated by SAF-1 and AP-1. J Biol Chem 2009; 284:1853-62. [DOI: 10.1074/jbc.m806289200] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
19
|
Qiu X, Aiken KJ, Chokas AL, Beachy DE, Nick HS. Distinct functions of CCAAT enhancer-binding protein isoforms in the regulation of manganese superoxide dismutase during interleukin-1beta stimulation. J Biol Chem 2008; 283:25774-85. [PMID: 18559338 PMCID: PMC2533776 DOI: 10.1074/jbc.m801178200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Revised: 06/03/2008] [Indexed: 01/29/2023] Open
Abstract
The mitochondrial antioxidant enzyme manganese superoxide dismutase (Mn-SOD) is crucial in maintaining cellular and organismal homeostasis. Mn-SOD expression is tightly regulated in a manner that synchronizes its cytoprotective functions during inflammatory challenges. Induction of Mn-SOD gene expression by the proinflammatory cytokine IL-1beta is mediated through a complex intronic enhancer element. To identify and characterize the transcription factors required for Mn-SOD enhancer function, a yeast one-hybrid assay was utilized, and two CCAAT enhancer-binding protein (C/EBP) members, C/EBP beta and C/EBP delta, were identified. These two transcription factors responded to IL-1beta treatment with distinct expression profiles, different temporal yet inducible interactions with the endogenous Mn-SOD enhancer, and also opposite effects on Mn-SOD transcription. C/EBP beta is expressed as three isoforms, LAP* (liver-activating protein), LAP, and LIP (liver-inhibitory protein). Our functional analysis demonstrated that only the full-length C/EBP beta/LAP* served as a true activator for Mn-SOD, whereas LAP, LIP, and C/EBP delta functioned as potential repressors. Finally, our systematic mutagenesis of the unique N-terminal 21 amino acids further solidified the importance of LAP* in the induction of Mn-SOD and emphasized the crucial role of this isoform. Our data demonstrating the physiological relevance of the N-terminal peptide also provide a rationale for revisiting the role of LAP* in the regulation of other genes and in pathways such as lipogenesis and development.
Collapse
Affiliation(s)
- Xiaolei Qiu
- Department of Neuroscience,
McKnight Brian Institute, and
Department of Biochemistry and Molecular
Biology, University of Florida, Gainesville, Florida 32610
| | - Kimberly J. Aiken
- Department of Neuroscience,
McKnight Brian Institute, and
Department of Biochemistry and Molecular
Biology, University of Florida, Gainesville, Florida 32610
| | - Ann L. Chokas
- Department of Neuroscience,
McKnight Brian Institute, and
Department of Biochemistry and Molecular
Biology, University of Florida, Gainesville, Florida 32610
| | - Dawn E. Beachy
- Department of Neuroscience,
McKnight Brian Institute, and
Department of Biochemistry and Molecular
Biology, University of Florida, Gainesville, Florida 32610
| | - Harry S. Nick
- Department of Neuroscience,
McKnight Brian Institute, and
Department of Biochemistry and Molecular
Biology, University of Florida, Gainesville, Florida 32610
| |
Collapse
|
20
|
Hattori H, Imai H, Kirai N, Furuhama K, Sato O, Konishi K, Nakagawa Y. Identification of a responsible promoter region and a key transcription factor, CCAAT/enhancer-binding protein epsilon, for up-regulation of PHGPx in HL60 cells stimulated with TNF alpha. Biochem J 2007; 408:277-86. [PMID: 17688422 PMCID: PMC2267347 DOI: 10.1042/bj20070245] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the present study we investigated promoter regions of the PHGPx [phospholipid hydroperoxide GPx (glutathione peroxidase)] gene and transcription factors involved in TNFalpha (tumour necrosis factor alpha)-induced up-regulation of PHGPx in non-differentiated HL60 cells. Non-differentiated HL60 cells displayed up-regulation of non-mitochondrial and mitochondrial PHGPx mRNA in response to TNFalpha stimulation. The promoter activity was up-regulated by TNFalpha stimulation in cells transfected with a luciferase reporter vector encoding the region from -282 to -123 of the human PHGPx gene compared with the non-stimulated control. The up-regulated promoter activity was effectively abrogated by a mutation in the C/EBP (CCAAT/enhancer-binding protein)-binding sequence in this region. ChIP (chromatin immunoprecipitation) assays demonstrated that C/EBPepsilon bound to the -247 to -34 region in HL60 cells, but C/EBPalpha, beta, gamma and delta did not. The binding of C/EBPepsilon to the promoter region was increased in HL60 cells stimulated with TNFalpha compared with that of the non-stimulated control. An increased binding of nuclear protein to the C/EBP-binding sequence was observed by EMSA (electrophoretic mobility-shift assay) in cells stimulated with TNFalpha, and it was inhibited by pre-treatment with an anti-C/EBPepsilon antibody, but not with other antibodies. The C/EBPepsilon mRNA was expressed in PMNs (polymorphonuclear cells), non-differentiated HL60 cells and neutrophil-like differentiated HL60 cells displaying TNFalpha-induced up-regulation of PHGPx mRNA, but not in macrophage-like differentiated HL60 cells, HEK-293 cells (human embryonic kidney-293 cells) and other cell lines exhibiting no up-regulation. The up-regulation of PHGPx mRNA, however, was detected in HEK-293 cells overexpressing C/EBPepsilon as a result of TNFalpha stimulation. These results indicate that C/EBPepsilon is a critical transcription factor in TNFalpha-induced up-regulation of PHGPx expression.
Collapse
Affiliation(s)
- Hiroyuki Hattori
- *School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
- †Medicinal Safety Research Laboratories, Daiichi Sankyo Corporation, 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Hirotaka Imai
- *School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
- ‡PRESTO (Precursory research for embryonic science and technology), Japan Science and Technology Agency, 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan
| | - Nozomu Kirai
- *School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Kazuhisa Furuhama
- §Veterinary Pharmacology, Iwate University, 3-18-8 Ueda, Morioka 020-8550, Japan
| | - Osamu Sato
- ∥Clinical Data and Biostatistics Department, Daiichi Sankyo Corporation, 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Kumiko Konishi
- *School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Yasuhito Nakagawa
- *School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
- To whom correspondence should be addressed (email )
| |
Collapse
|
21
|
Venteclef N, Smith JC, Goodwin B, Delerive P. Liver receptor homolog 1 is a negative regulator of the hepatic acute-phase response. Mol Cell Biol 2006; 26:6799-807. [PMID: 16943422 PMCID: PMC1592867 DOI: 10.1128/mcb.00579-06] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The orphan nuclear receptor liver receptor homolog 1 (LRH-1) has been reported to play an important role in bile acid biosynthesis and reverse cholesterol transport. Here, we show that LRH-1 is a key player in the control of the hepatic acute-phase response. Ectopic expression of LRH-1 with adenovirus resulted in strong inhibition of both interleukin-6 (IL-6)- and IL-1beta-stimulated haptoglobin, serum amyloid A, and fibrinogen beta gene expression in hepatocytes. Furthermore, induction of the hepatic inflammatory response was significantly exacerbated in HepG2 cells expressing short hairpin RNA targeting LRH-1 expression. Moreover, transient-transfection experiments and electrophoretic mobility shift and chromatin immunoprecipitation assays revealed that LRH-1 regulates this cytokine-elicited inflammatory response by, at least in part, antagonizing the CCAAT/enhancer binding protein beta signaling pathway. Finally, we show, by using LRH-1 heterozygous mice, that LRH-1 is involved in the control of the inflammatory response at the hepatic level in vivo. Taken together, our results outline an unexpected role for LRH-1 in the modulation of the hepatic acute-phase response.
Collapse
Affiliation(s)
- Nicolas Venteclef
- GlaxoSmithKline R&D, CVU CEDD, 25 Avenue du Quebec, 91951 Les Ulis, France
| | | | | | | |
Collapse
|
22
|
Ray A, Shakya A, Kumar D, Benson MD, Ray BK. Inflammation-responsive transcription factor SAF-1 activity is linked to the development of amyloid A amyloidosis. THE JOURNAL OF IMMUNOLOGY 2006; 177:2601-9. [PMID: 16888022 DOI: 10.4049/jimmunol.177.4.2601] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Abundantly expressed serum amyloid A (SAA) protein under chronic inflammatory conditions gives rise to insoluble aggregates of SAA derivatives in multiple organs resulting in reactive amyloid A (AA) amyloidosis, a consequence of rheumatoid arthritis, Crohn's disease, ankylosing spondylitis, familial Mediterranean fever, and Castleman's disease. An inflammation-responsive transcription factor, SAF (for SAA activating factor), has been implicated in the sustained expression of amyloidogenic SAA under chronic inflammatory conditions. However, its role in the pathogenesis of AA amyloidosis has thus far remained obscure. In this paper we have shown that SAF-1, a major member of the SAF family, is abundantly present in human AA amyloidosis patients. To assess whether SAF-1 is directly linked to the pathogenesis of AA amyloidosis, we have developed a SAF-1 transgenic mouse model. SAF-1-overexpressing mice spontaneously developed AA amyloidosis at the age of 14 mo or older. Immunohistochemical analysis confirmed the nature of the amyloid deposits as an AA type derived from amyloidogenic SAA1. Furthermore, SAF-1 transgenic mice rapidly developed severe AA amyloidosis in response to azocasein injection, indicating increased susceptibility to inflammation. Also, during inflammation SAF-1 transgenic mice exhibited a prolonged acute phase response, leading to an extended period of SAA synthesis. Together, these results provide direct evidence that SAF-1 plays a key role in the development of AA amyloidosis, a consequence of chronic inflammation.
Collapse
Affiliation(s)
- Alpana Ray
- Department of Veterinary Pathobiology, University of Missouri, 126A Connaway Hall, Columbia, MO 65211, USA.
| | | | | | | | | |
Collapse
|
23
|
Sun Y, Boyd K, Xu W, Ma J, Jackson CW, Fu A, Shillingford JM, Robinson GW, Hennighausen L, Hitzler JK, Ma Z, Morris SW. Acute myeloid leukemia-associated Mkl1 (Mrtf-a) is a key regulator of mammary gland function. Mol Cell Biol 2006; 26:5809-26. [PMID: 16847333 PMCID: PMC1592762 DOI: 10.1128/mcb.00024-06] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Transcription of immediate-early genes--as well as multiple genes affecting muscle function, cytoskeletal integrity, apoptosis control, and wound healing/angiogenesis--is regulated by serum response factor (Srf). Extracellular signals regulate Srf in part via a pathway involving megakaryoblastic leukemia 1 (Mkl1, also known as myocardin-related transcription factor A [Mrtf-a]), which coactivates Srf-responsive genes downstream of Rho GTPases. Here we investigate Mkl1 function using gene targeting and show the protein to be essential for the physiologic preparation of the mammary gland during pregnancy and the maintenance of lactation. Lack of Mkl1 causes premature involution and impairs expression of Srf-dependent genes in the mammary myoepithelial cells, which control milk ejection following oxytocin-induced contraction. Despite the importance of Srf in multiple transcriptional pathways and widespread Mkl1 expression, the spectrum of abnormalities associated with Mkl1 absence appears surprisingly restricted.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Apoptosis
- Child
- Failure to Thrive
- Female
- Gene Expression Profiling
- Gene Expression Regulation
- Gene Targeting
- Heart/anatomy & histology
- Heart/embryology
- Humans
- Infant
- Lactation/physiology
- Leukemia, Megakaryoblastic, Acute
- Male
- Mammary Glands, Animal/abnormalities
- Mammary Glands, Animal/anatomy & histology
- Mammary Glands, Animal/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Milk
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/ultrastructure
- Oligonucleotide Array Sequence Analysis
- Oxytocin/metabolism
- Pregnancy
- Prolactin/metabolism
- STAT3 Transcription Factor
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Trans-Activators/genetics
- Trans-Activators/metabolism
Collapse
Affiliation(s)
- Yi Sun
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Shen F, Hu Z, Goswami J, Gaffen SL. Identification of common transcriptional regulatory elements in interleukin-17 target genes. J Biol Chem 2006; 281:24138-48. [PMID: 16798734 DOI: 10.1074/jbc.m604597200] [Citation(s) in RCA: 236] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interleukin (IL)-17 is the founding member of a novel family of inflammatory cytokines. Although produced by T cells, IL-17 activates genes and signals typical of innate immune mediators such as tumor necrosis factor (TNF)-alpha and IL-1beta. Most IL-17 target genes characterized to date are cytokines or neutrophil-attractive chemokines. Our recent microarray studies identified an acute phase response gene, 24p3/lipocalin 2, as a novel IL-17-induced gene. Here we describe a detailed analysis of the 24p3 promoter. We find that, unlike cytokine or chemokine gene target genes, 24p3 is regulated primarily at the level of transcription rather than mRNA stability and that synergy between IL-17 and TNFalpha occurs at the level of the 24p3 promoter. Two key transcription factor binding sites (TFBS) were identified, corresponding to NF-kappaB and CCAAT/enhancer-binding protein (C/EBP). Deletion of either site eliminated 24p3 promoter activity in response to IL-17. These findings were strikingly similar to the IL-6 promoter, where IL-17-mediated regulation of both NF-kappaB and C/EBP is essential. To determine whether joint use of NF-kappaB and C/EBP is common to all IL-17 target genes, we performed a computational analysis on 18 well documented IL-17 target promoters to assess statistical enrichment of specific TFBSs. Indeed, NF-kappaB and C/EBP sites were over-represented in these genes, as were AP1 and OCT1 sites. Moreover, these promoters fell into three definable subcategories based on TFBS location and usage. Analysis of IL-17 target gene regulation is key for understanding this important host-defense molecule and also contributes to an understanding of upstream signaling mechanisms used by IL-17, either alone or in concert with TNFalpha.
Collapse
Affiliation(s)
- Fang Shen
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, State University of New York, Buffalo, New York 14214, USA
| | | | | | | |
Collapse
|
25
|
Kalvakolanu DV, Roy SK. CCAAT/enhancer binding proteins and interferon signaling pathways. J Interferon Cytokine Res 2006; 25:757-69. [PMID: 16375604 DOI: 10.1089/jir.2005.25.757] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Interferons (IFNs) regulate a number of host responses, including innate and adaptive immunity against viruses, microbes, and neoplastic cells. These responses are dependent on the expression of IFN-stimulated genes (ISGs). Given the diversities in these responses and their kinetics, it is conceivable that a number of different factors are required for controlling them. Here, we describe one such pathway wherein transcription factor CAAAT/enhancer binding protein-beta (C/EBP-beta) is controlled via IFN-gamma-induced MAPK signaling pathways. At least two IFN-gamma-induced MAPK signals converge on to C/EBP-beta for inducing transcription. One of these, driven by extracellular signal-regulated kinases (ERKs), phosphorylates the C/EBP-beta protein in its regulatory domain. The second, driven by the mixed-lineage kinases (MLKs), induces a dephosphorylation leading to the recruitment of transcriptional coactivators.
Collapse
Affiliation(s)
- Dhananjaya V Kalvakolanu
- Greenebaum Cancer Center, Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | |
Collapse
|
26
|
Paz-Priel I, Cai DH, Wang D, Kowalski J, Blackford A, Liu H, Heckman CA, Gombart AF, Koeffler HP, Boxer LM, Friedman AD. CCAAT/enhancer binding protein alpha (C/EBPalpha) and C/EBPalpha myeloid oncoproteins induce bcl-2 via interaction of their basic regions with nuclear factor-kappaB p50. Mol Cancer Res 2006; 3:585-96. [PMID: 16254192 DOI: 10.1158/1541-7786.mcr-05-0111] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The CEBPA gene is mutated in 10% of acute myeloid leukemia (AML) cases. We find that CEBPA and Bcl-2 RNA levels correlate highly in low-risk human AMLs, suggesting that inhibition of apoptosis via induction of bcl-2 by CCAAT/enhancer binding protein alpha (C/EBPalpha) or its mutant variants contributes to transformation. C/EBPalphap30, lacking a NH2-terminal transactivation domain, or C/EBPalphaLZ, carrying in-frame mutations in the leucine zipper that prevent DNA binding, induced bcl-2 in hematopoietic cell lines, and C/EBPalpha induced bcl-2 in normal murine myeloid progenitors and in the splenocytes of H2K-C/EBPalpha-Emu transgenic mice. C/EBPalpha protected Ba/F3 cells from apoptosis on interleukin-3 withdrawal but not if bcl-2 was knocked down. Remarkably, C/EBPalphaLZ oncoproteins activated the bcl-2 P2 promoter despite lack of DNA binding, and C/EBPalphap30 also activated the promoter. C/EBPalpha and the C/EBPalpha oncoproteins cooperated with nuclear factor-kappaB (NF-kappaB) p50, but not p65, to induce bcl-2 transcription. Endogenous C/EBPalpha preferentially coimmunoprecipitated with p50 versus p65 in myeloid cell extracts. Mutation of residues 297 to 302 in the C/EBPalpha basic region prevented induction of endogenous bcl-2 or the bcl-2 promoter and interaction with p50 but not p65. These findings suggest that C/EBPalpha or its mutant variants tether to a subset of NF-kappaB target genes, including Bcl-2, via p50 to facilitate gene activation and offer an explanation for preferential in-frame rather than out-of-frame mutation of the leucine zipper with sparing of the basic region in C/EBPalphaLZ oncoproteins. Targeting interaction between C/EBPalpha basic region and NF-kappaB p50 may contribute to the therapy of AML and other malignancies expressing C/EBPs.
Collapse
MESH Headings
- Adult
- Animals
- Apoptosis/physiology
- CCAAT-Enhancer-Binding Protein-alpha/genetics
- CCAAT-Enhancer-Binding Protein-alpha/metabolism
- Cell Line
- HL-60 Cells
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/metabolism
- Humans
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- NF-kappa B p50 Subunit/metabolism
- Oncogene Proteins/genetics
- Oncogene Proteins/metabolism
- Promoter Regions, Genetic
- Proto-Oncogene Proteins c-bcl-2/biosynthesis
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Rats
- Transcriptional Activation
Collapse
Affiliation(s)
- Ido Paz-Priel
- Division of Pediatric Oncology, Johns Hopkins University, CRB 253, 1650 Orleans Street, Baltimore, Maryland 21231, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Hagihara K, Nishikawa T, Sugamata Y, Song J, Isobe T, Taga T, Yoshizaki K. Essential role of STAT3 in cytokine-driven NF-kappaB-mediated serum amyloid A gene expression. Genes Cells 2005; 10:1051-63. [PMID: 16236134 DOI: 10.1111/j.1365-2443.2005.00900.x] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Serum amyloid A (SAA) is a sensitive marker of acute-phase responses and known as a precursor protein of amyloid fibril in amyloid A (AA) (secondary) amyloidosis. Since the serum SAA level is also closely related to activity of chronic inflammatory disease and coronary artery disease, it is important to clarify the exact induction mechanism of SAA from the clinical point of view. Here we provide evidence that STAT3 plays an essential role in cytokine-driven SAA expression, although the human SAA gene shows no typical STAT3 response element (RE) in its promoters. STAT3 and nuclear factor kappaB (NF-kappaB) p65 first form a complex following interleukin (IL)-1 and IL-6 (IL-1+6) stimulation, after which STAT3 interacts with nonconsensus sequences at a 3' site of the SAA gene promoter's NF-kappaB RE. Moreover, co-expression of p300 with STAT3 dramatically enhances the transcriptional activity of SAA. The formation of a complex with STAT3, NF-kappaB p65, and p300 is thus essential for the synergistic induction of the SAA gene by IL-1+6 stimulation. Our findings are expected to aid the understanding of the inflammatory status of AA amyloidosis to aid development of a therapeutic strategy for this disease by means of normalization of serum SAA levels.
Collapse
Affiliation(s)
- Keisuke Hagihara
- Molecular Medicine, Osaka University Graduate School of Medicine, Osaka University, 2-1 Yamada-Oka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|
28
|
López-Bojórquez LN, Dehesa AZ, Reyes-Terán G. Molecular mechanisms involved in the pathogenesis of septic shock. Arch Med Res 2005; 35:465-79. [PMID: 15631870 DOI: 10.1016/j.arcmed.2004.07.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pathogenesis of the development of sepsis is highly complex and has been the object of study for many years. The inflammatory phenomena underlying septic shock are described in this review, as well as the enzymes and genes involved in the cellular activation that precedes this condition. The most important molecular aspects are discussed, ranging from the cytokines involved and their respective transduction pathways to the cellular mechanisms related to accelerated catabolism and multi-organic failure.
Collapse
Affiliation(s)
- Lucia Nikolaia López-Bojórquez
- Departamento de Biología Celular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, D.F., Mexico.
| | | | | |
Collapse
|
29
|
Gorrill TS, Khalili K. Cooperative interaction of p65 and C/EBPbeta modulates transcription of BKV early promoter. Virology 2005; 335:1-9. [PMID: 15823601 DOI: 10.1016/j.virol.2005.02.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2004] [Revised: 12/30/2004] [Accepted: 02/07/2005] [Indexed: 10/25/2022]
Abstract
Reactivation of the human polyomavirus BK (BKV) has emerged as an important cause of allograft rejection in renal transplant recipients. Expression of the viral early promoter that leads to production of T-antigen is the first event in viral lytic infection. In an effort to understand the mechanism involved in the activation of BKV early gene (BKV(E)) expression, we analyzed the promoter/enhancer region of the virus and identified binding motifs for the inducible transcription factors NF-kappaB and C/EBPbeta, which are in juxtaposition to each other downstream from the early gene transcription initiation site. Results from transfection studies demonstrate that overexpression of the p65 subunit of NF-kappaB, but not C/EBPbeta stimulates transcription of the BKV(E) promoter in CV-1 cells. Interestingly, low level expression of C/EBPbeta showed a synergistic effect on p65 activation of the BKV(E) promoter, suggesting a functional cooperativity between these two regulators upon viral gene transcription. Results from DNA-binding studies showed the ability of p65 and C/EBPbeta to bind independently with BKV DNA as removal of the binding site for p65 or C/EBPbeta had no significant effect on the interaction of p65 and C/EBPbeta with their motifs, respectively. Functional evaluation of the mutant promoter with no binding sites for either NF-kappaB or C/EBPbeta showed that the observed synergism requires the p65 but not the C/EBPbeta binding site, suggesting cross-talk between C/EBPbeta and p65 in this event. Results from the co-expression of p65 and C/EBPbeta showed no evidence for the formation of a DNA-protein complex containing both p65 and C/EBPbeta, although results from protein-protein interaction studies verified the ability of C/EBPbeta to interact with p65. A dominant-negative isoform of C/EBPbeta which contains the DNA binding but not activation domain of full-length C/EBPbeta cooperated with p65 in activating the BKV(E) promoter, suggesting a functional interaction between the b-ZIP domain of C/EBPbeta and NF-kappaB.
Collapse
Affiliation(s)
- Timothy S Gorrill
- Center for Neurovirology and Cancer Biology, College of Science and Technology, Temple University, 1900 North 12th Street, 015-96, Room 203, Philadelphia, PA 19122, USA
| | | |
Collapse
|
30
|
Ghadially H, Ross XL, Kerst C, Dong J, Reske-Kunz AB, Ross R. Differential Regulation of CCL22 Gene Expression in Murine Dendritic Cells and B Cells. THE JOURNAL OF IMMUNOLOGY 2005; 174:5620-9. [PMID: 15843561 DOI: 10.4049/jimmunol.174.9.5620] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The activated T cell-attracting CC chemokine CCL22 is expressed by stimulated B cells and mature dendritic cells (DC). We have cloned and sequenced the complete mouse gene, including 4 kb of the 5'-flanking promoter region, and detected two distinct sites for initiation of transcription by 5'-RACE. Reporter gene assays indicate that the promoter reflects the specificity of the endogenous gene. Within the proximal promoter region, we identified potential binding sites for NF-kappaB, Ikaros, and a putative GC box. All three regions bind proteins. The NF-kappaB site was shown to specifically bind NF-kappaB subunits p50 and p65 from nuclear extracts of LPS-stimulated B cells, B cell line A20/2J, TNF-alpha-stimulated bone marrow-derived DC, and DC line XS106. Furthermore, promoter activity was affected by targeted mutagenesis of the NF-kappaB site and transactivation with p50 and p65. The region harboring the putative Ikaros site contributes to promoter activity, but the binding protein does not belong to the Ikaros family. The GC box was shown to specifically bind Sp1 using extracts from LPS-stimulated B cells and A20/2J but not from DC and DC line XS106. Additionally, Sp1 transactivated the promoter in A20/2J but not in XS106 cells, and mutation of the Sp1 site diminished transactivation. Furthermore, binding of the protein complex at the GC box is required for NF-kappaB activity, and the spatial alignment of the binding sites is of critical importance for promoter activity. Thus, identical and distinct proteins contribute to expression of CCL22 in DC and B cells.
Collapse
Affiliation(s)
- Hormas Ghadially
- Clinical Research Unit Allergology, Department of Dermatology, Johannes Gutenberg-University, Mainz, Germany
| | | | | | | | | | | |
Collapse
|
31
|
Chen J, Zhao M, Rao R, Inoue H, Hao CM. C/EBPβ and Its Binding Element Are Required for NFκB-induced COX2 Expression Following Hypertonic Stress. J Biol Chem 2005; 280:16354-9. [PMID: 15713664 DOI: 10.1074/jbc.m411134200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
NFkappaB plays a critical role mediating COX2 expression in renal medullary interstitial cells (RMICs). The trans-activating ability of NFkappaB can be modified by another nuclear factor C/EBPbeta that can physically bind to NFkappaB and regulate its activity. Because the COX2 promoter also contains a C/EBPbeta site adjacent to the NFkappaB site, the present study examined whether these two transcription factors cooperate to induce COX2 expression following hypertonic stress. Hypertonicity markedly induced COX2 expression in cultured medullary interstitial cells by immunoblot analysis. The tonicity-induced COX2 expression was suppressed by mutant IkappaB (IkappaBm) that blocks NFkappaB activation, demonstrating that tonicity-induced COX2 expression depends on NFkappaB activation. However, mutation of the NFkappaB site in the COX2 promoter failed to abolish tonicity-induced COX2 reporter activity. IkappaB kinase-1 (IKK1) significantly induced COX2-luciferase activity by 2.3-fold (n = 10, p < 0.01); mutation of the NFkappaB site also failed to abolish IKK1-stimulated COX2 reporter activity (86 +/- 3.1% of wild type, p > 0.05, n = 4). Interestingly, mutation of the C/EBPbeta site of the COX2 gene significantly reduced both IKK1 and hypertonicity-induced COX2 reporter activity (p < 0.01). To further examine the potential role of C/EBPbeta in tonicity-induced COX2 expression, a dominant negative C/EBPbeta-p20 was transduced into RMICs. C/EBPbeta-p20 markedly suppressed hypertonic (550 mOsm) induction of COX2 (immunoblot) to a similar extent as IkappaBm. No additional suppression was observed when both NFkappaB and C/EBPbeta were simultaneously blocked by IkappaBm and C/EBPbeta-p20. Interestingly, IKK-induced COX2 expression was not only blocked by IkappaBm, but also completely abolished by C/EBPbeta-p20. Further studies demonstrated physical association of C/EBPbeta to NFkappaB p65 by coimmunoprecipitation. Importantly, this interaction between C/EBPbeta and NFkappaB was greatly enhanced following hypertonic stress. These studies indicate C/EBPbeta is required for the transcriptional activation of COX2 by NFkappaB, suggesting a dominant role for the C/EBPbeta pathway in regulating induction of RMIC COX2 by hypertonicity.
Collapse
Affiliation(s)
- Jing Chen
- Division of Nephrology, Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | | | | | | | |
Collapse
|
32
|
Migita K, Abiru S, Nakamura M, Komori A, Yoshida Y, Yokoyama T, Daikoku M, Ueki T, Takii Y, Yano K, Yastuhashi H, Eguchi K, Ishibashi H. Lipopolysaccharide signaling induces serum amyloid A (SAA) synthesis in human hepatocytes in vitro. FEBS Lett 2004; 569:235-9. [PMID: 15225640 DOI: 10.1016/j.febslet.2004.05.072] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2004] [Revised: 05/19/2004] [Accepted: 05/27/2004] [Indexed: 11/24/2022]
Abstract
To investigate the role of lipopolysaccharide (LPS) in hepatocyte activation, we examined the expression of Toll-like receptor 4 (TLR4), the putative receptor for LPS in human hepatocytes. TLR4 mRNA and protein expression was confirmed in human hepatocytes. Stimulation of human hepatocytes with LPS results in rapid degradation of IkappaB-alpha and mitogen activated protein kinase activation. Human hepatocytes stimulated by LPS produced serum amyloid A protein. Our data suggest that human hepatocytes utilize components of TLR4 signal transduction pathways in response to LPS and these direct LPS-mediated effects on hepatocytes may contribute to liver inflammation and injury.
Collapse
Affiliation(s)
- Kiyoshi Migita
- Clinical Research Center, National Nagasaki Medical Center, Kubara 2-1001-1, Omura 856-8562, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Friedman JR, Larris B, Le PP, Peiris TH, Arsenlis A, Schug J, Tobias JW, Kaestner KH, Greenbaum LE. Orthogonal analysis of C/EBPbeta targets in vivo during liver proliferation. Proc Natl Acad Sci U S A 2004; 101:12986-91. [PMID: 15317935 PMCID: PMC516505 DOI: 10.1073/pnas.0402875101] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
CCAAT enhancer-binding protein beta (C/EBPbeta), a basic-leucine zipper transcription factor, is an important effector of signals in physiologic growth and cancer. The identification of direct C/EBPbeta targets in vivo has been limited by functional compensation by other C/EBP family proteins and the low stringency of the consensus sequence. Here we use the combined power of expression profiling and high-throughput chromatin immunoprecipitation to identify direct and biologically relevant targets of C/EBPbeta. We identified 25 potential C/EBPbeta targets, of which 88% of those tested were confirmed as in vivo C/EBPbeta-binding sites. Six of these genes also displayed differential expression in C/EBPbeta-/- livers. Computational analysis revealed that bona fide C/EBPbeta target genes can be distinguished by the presence of binding motifs for specific additional transcription factors in the vicinity of the C/EBPbeta site. This approach is generally applicable to the discovery of direct, biologically relevant targets of mammalian transcription factors.
Collapse
Affiliation(s)
- Joshua R Friedman
- Department of Genetics, Bioinformatics Core, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Chapman LM, Roling JA, Bingham LK, Herald MR, Baldwin WS. Construction of a subtractive library from hexavalent chromium treated winter flounder (Pseudopleuronectes americanus) reveals alterations in non-selenium glutathione peroxidases. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2004; 67:181-94. [PMID: 15003702 DOI: 10.1016/j.aquatox.2003.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2003] [Revised: 12/05/2003] [Accepted: 12/12/2003] [Indexed: 05/24/2023]
Abstract
Chromium is released during several industrial processes and has accumulated in some estuarine areas. Its effects on mammals have been widely studied, but relatively little information is available on its effects on fish. Gene expression changes are useful biomarkers that can provide information about toxicant exposure and effects, as well as the health of an organism and its ability to adapt to its surroundings. Therefore, we investigated the effects of Cr(VI) on gene expression in the sediment dwelling fish, winter flounder (Pseudopleuronectes americanus). Winter flounder ranging from 300 to 360 g were injected i.p. with Cr(VI) as chromium oxide at 25 microg/kg chromium in 0.15N KCl. Twenty-four hours following injections, winter flounder were euthanized with MS-222 and the livers were excised. Half of the livers were used to make cytosol and the other half were used to isolate mRNA for subtractive hybridization. Subtractive clones obtained were spotted onto nylon filters, which revealed several genes with potentially altered expression due to Cr(VI), including an alpha class GST, 1-Cys peroxiredoxin (a non-selenium glutathione peroxidase), a P-450 2X subfamily member, two elongation factors (EF-1 gamma and EF-2), and complement component C3. Semi-quantitative RT-PCR was performed and confirmed that Cr(VI) down-regulated complement component C3, an EST, and two potential glutathione peroxidases, GSTA3 and 1-Cys peroxiredoxin. In addition, cytosolic GSH peroxidase activity was reduced, and silver stained SDS-PAGE gels from glutathione-affinity purified cytosol demonstrated that a 27.1 kDa GSH-binding protein was down-regulated greater than 50%. Taken together, Cr(VI) significantly altered the expression of several genes including two potential glutathione peroxidases in winter flounder.
Collapse
Affiliation(s)
- Laura M Chapman
- Biological Sciences, University of Texas at El Paso, 500 W. University Avenue, El Paso, TX 79968, USA
| | | | | | | | | |
Collapse
|
35
|
Abstract
More than a half a century ago, interferons (IFN) were identified as antiviral cytokines. Since that discovery, IFN have been in the forefront of basic and clinical cytokine research. The pleiotropic nature of these cytokines continues to engage a large number of investigators to define their actions further. IFN paved the way for discovery of Janus tyrosine kinase (JAK)-signal transducing activators of transcription (STAT) pathways. A number of important tumor suppressive pathways are controlled by IFN. Several infectious pathogens counteract IFN-induced signaling pathways. Recent studies indicate that IFN activate several new protein kinases, including the MAP kinase family, and downstream transcription factors. This review not only details the established IFN signaling paradigms but also provides insights into emerging alternate signaling pathways and mechanisms of pathogen-induced signaling interference.
Collapse
Affiliation(s)
- Dhananjaya V Kalvakolanu
- Molecular and Cellular Biology Graduate Program, Greenebaum Cancer Center, Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
36
|
Cho K, Pham TN, Crivello SD, Jeong J, Green TL, Greenhalgh DG. Involvement of CD14 and Toll-Like Receptor 4 in the Acute Phase Response of Serum Amyloid A Proteins and Serum Amyloid P Component in the Liver After Burn Injury. Shock 2004; 21:144-50. [PMID: 14752288 DOI: 10.1097/01.shk.0000108398.56565.ae] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Acute phase proteins such as serum amyloid A proteins (SAAs) and serum amyloid P component (SAP) are induced in the liver after various insults (e.g., infection, injury). The cellular and molecular mechanisms controlling the expression of these acute phase proteins may be specifically designed for different insults. The roles of two central molecules of the lipopolysaccharide (LPS)-mediated inflammation pathway (CD14 and toll-like receptor 4 [Tlr4]) were investigated for the regulation of SAAs and SAP in the liver of mice after an 18% total body surface area burn injury. RT-PCR analysis revealed a subtype- and time-dependent induction of SAA mRNAs between 3 h and 3 days, while there was a peak induction of SAP mRNA at day 1. Marked elevations of SAA and SAP protein levels at day 1 supported the mRNA data. Furthermore, a differential regulation of SAAs and SAP mRNAs was noted between CD14 knockout (KO) and their control mice after injury. SAA protein was induced to a lesser degree after injury in C3H/HeJ (Tlr4-defective) mice than in their control mice. In addition, in both CD14 KO and C3H/HeJ mice, the induction of SAP protein was significantly reduced compared with respective controls. These data provide evidence that CD14 and Tlr4 participate, at least in part, in a cascade of signaling events that control the immediate-early and differential induction of SAAs and SAP in the liver after injury. They also suggest that LPS may be one of the initial inducing agents associated with these acute phase responses in the liver after injury.
Collapse
Affiliation(s)
- Kiho Cho
- Burn Research, Shriners Hospitals for Children Northern California, Department of Surgery, University of California at Davis, Sacramento 95817, USA
| | | | | | | | | | | |
Collapse
|
37
|
Lindstrom T, Bennett P. Transcriptional regulation of genes for enzymes of the prostaglandin biosynthetic pathway. Prostaglandins Leukot Essent Fatty Acids 2004; 70:115-35. [PMID: 14683688 DOI: 10.1016/j.plefa.2003.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Numerous studies over the years have demonstrated changes in prostaglandin (PG) levels in intrauterine tissues in association with labour, and PG administration has long been used to induce delivery. While it is now widely accepted that PGs play a major role in human parturition, the complex regulation of their levels is still being elucidated, with the focus on the transcriptional control of the enzymes responsible for the various steps in PG biosynthesis and catabolism.
Collapse
Affiliation(s)
- Tamsin Lindstrom
- Faculty of Medicine, Institute of Reproductive and Developmental Biology, Parturition Research Group, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK.
| | | |
Collapse
|
38
|
Papin S, Cazeneuve C, Duquesnoy P, Jeru I, Sahali D, Amselem S. The tumor necrosis factor alpha-dependent activation of the human mediterranean fever (MEFV) promoter is mediated by a synergistic interaction between C/EBP beta and NF kappaB p65. J Biol Chem 2003; 278:48839-47. [PMID: 14514692 DOI: 10.1074/jbc.m305166200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
MEFV is a gene expressed specifically in myeloid cells and whose mutations underlie an autosomal recessive auto-inflammatory disease, called familial Mediterranean fever (FMF), characterized by recurrent episodes of serosal inflammation. This gene, which encodes a protein with unclear physiological functions, has been shown to be up-regulated by the pro-inflammatory cytokine tumor necrosis factor alpha (TNFalpha). However, the mechanism of this regulation is unknown, and the MEFV promoter is still to be characterized. Here, we show that 243 bp of the 5'-flanking region of the human MEFV gene are sufficient to direct high level expression of MEFV in TNFalpha-treated cells. The TNFalpha-induced expression of MEFV is dependent on both NFkappaB p65 and C/EBPbeta that bind to evolutionarily conserved sites located, in the human promoter, at positions -163 and -55, respectively. As shown by a series of transcription and gel shift assays performed with wild-type and mutated promoter sequences, these two transcription factors act differently on the TNFalpha-dependent transcription of MEFV: C/EBPbeta is the key regulatory factor required to confer cell responsiveness to TNFalpha, whereas NFkappaB p65 increases this response by means of a synergistic interaction with C/EBPbeta that is dependent on the integrity of the identified -55 C/EBP binding site. Given the phenotype of patients with FMF, this C/EBP-NFkappaB interaction may represent a key step in the control of an inflammatory response that is abnormally high in this disease. These data, which shed novel light on the pathophysiology of FMF, represent an unusual example of cross-talk between C/EBP and NFkappaB pathways in TNFalpha signaling.
Collapse
Affiliation(s)
- Stéphanie Papin
- INSERM, Unit 468 and Unit 581, Hôpital Henri-Mondor, 51, Avenue du Maréchal de-Lattre-de-Tassigny, 94010 Créteil, France
| | | | | | | | | | | |
Collapse
|
39
|
Vallée S, Fouchier F, Brémond P, Briand C, Marvaldi J, Champion S. Insulin-like growth factor-1 downregulates nuclear factor kappa B activation and upregulates interleukin-8 gene expression induced by tumor necrosis factor alpha. Biochem Biophys Res Commun 2003; 305:831-9. [PMID: 12767906 DOI: 10.1016/s0006-291x(03)00866-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Pretreatment of HT29-D4 epithelial adenocarcinoma colic cells with des-IGF-1 upregulated TNF alpha-mediated activation of IL-8 expression at different levels (protein, mRNA, and hnRNA). RNA transcription but not RNA stabilization was found to be involved. In this cell line, cooperation of NF-kappa B with other factors appeared essential for IL-8 expression. Indeed, TNF alpha-induced NF-kappa B translocation was not sufficient to support enhancement of the transcription and des-IGF-1 did not promote but partly inhibited both the TNF alpha-induced NF-kappa B activation and I kappa B alpha degradation through a PI-3K-dependent pathway. A CCAAT/enhancer binding protein (C/EBP) site located on the IL-8 gene enhancer cooperated with a NF-kappa B binding site and led to the upregulation of IL-8 expression. Binding of C/EBP alpha to this sequence disappeared in IGF-1 treated cells. This event may be important for the cross-talk between IGF-1- and TNF alpha-mediated pathways leading to the control of inflammatory processes and the decision concerning apoptosis or cell survival.
Collapse
Affiliation(s)
- Sébastien Vallée
- Laboratoire de Biochimie Cellulaire, UMR 6032 CNRS, Faculté de Pharmacie, 27 Bd Jean Moulin, F-13385 Marseille, France
| | | | | | | | | | | |
Collapse
|
40
|
Borger P, Black JL, Roth M. Asthma and the CCAAT-enhancer binding proteins: a holistic view on airway inflammation and remodeling. J Allergy Clin Immunol 2002; 110:841-6. [PMID: 12464948 DOI: 10.1067/mai.2002.130047] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Asthma is an airway disease with increasing prevalence characterized by intermittent reversible airway obstruction, airway inflammation, and airway wall remodeling. The disease is generally triggered by inhalation of allergens, but nonallergic asthma triggers are quite common. The pathogenesis of asthma is well documented, and a great deal of research has been carried out to elucidate the underlying mechanisms. A multitude of articles have focused on cells alleged to be involved in the pathogenesis, including circulating cells from the immunologic compartment (ie, eosinophils and T lymphocytes) and resident cells, such as fibroblasts, airway smooth muscle cells, and, more recently, the airway epithelium. Despite the enormous amount of research, it is still unclear what exactly causes asthma. A general feature of most studies is an enhanced activation status of asthmatic cells, suggesting a general defect with respect to regulation of cellular responses. Here we discuss the ubiquitous transcription factor family of CCAAT-enhancer binding proteins (C/EBPs) and its involvement in inflammation and proliferation. We propose that an imbalance of C/EBP isoform expression might lead to an enhanced activity of asthmatic cells and provide an overall hypothesis that both airway inflammation and remodeling can be conceived as the result of an imbalance of C/EBP isoform expression.
Collapse
Affiliation(s)
- Peter Borger
- Department of Pharmacology and the Woolcock Institute of Medical Research, Royal Prince Alfred Hospital, University of Sydney, Australia
| | | | | |
Collapse
|
41
|
Yu Z, Cheng G, Wen X, Wu GD, Lee WT, Pleasure D. Tumor necrosis factor alpha increases neuronal vulnerability to excitotoxic necrosis by inducing expression of the AMPA-glutamate receptor subunit GluR1 via an acid sphingomyelinase- and NF-kappaB-dependent mechanism. Neurobiol Dis 2002; 11:199-213. [PMID: 12460558 DOI: 10.1006/nbdi.2002.0530] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Acid sphingomyelinase (ASMase) and NF-kappaB participate in tumor necrosis factor alpha (TNFalpha) signal transduction. Mice in which the genes encoding ASMase or the p50 subunit of NF-kappaB are disrupted have been reported to be less vulnerable than wild-type mice to focal brain ischemia. We now demonstrate selective diminution in expression of GluR1, an alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate-type glutamate receptor (AMPA-GluR) protein subunit, in these two groups of knockout mice. To confirm that neuronal GluR1 expression is regulated by ASMase and NF-kappaB, and to learn whether this regulation has pathophysiological significance, we treated cultured human NT2-N neurons with TNFalpha. This induced GluR1 expression and increased susceptibility of the neurons to kainate necrosis. Both induction of GluR1 and heightened vulnerability to kainate were blocked by inhibiting ASMase or by antisense knockdown of NF-kappaB p50. We conclude that TNFalpha can sensitize neurons to excitotoxic necrosis by inducing expression of GluR1 via an ASMase- and NF-kappaB-dependent mechanism. TNFalpha levels are frequently elevated during ischemia and other CNS diseases in which excitotoxicity contributes to neuronal loss. Our results suggest that inhibiting TNFalpha signal transduction will diminish neuronal necrosis in these diseases.
Collapse
Affiliation(s)
- ZaiFang Yu
- Department of Neurology Research, University of Pennsylvania School of Medicine, Philadelphia 19104, USA
| | | | | | | | | | | |
Collapse
|
42
|
Jin R, De Smaele E, Zazzeroni F, Nguyen DU, Papa S, Jones J, Cox C, Gelinas C, Franzoso G. Regulation of the gadd45beta promoter by NF-kappaB. DNA Cell Biol 2002; 21:491-503. [PMID: 12162804 DOI: 10.1089/104454902320219059] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In addition to coordinating immune and inflammatory responses, NF-kappaB/Rel transcription factors control cell survival. The NF-kappaB antiapoptotic function is crucial to oncogenesis, cancer chemoresistance, and to antagonize tumor necrosis factor (TNF) receptor-induced killing. Recently, we have shown that the suppression of the c-Jun-N-terminal kinase (JNK) cascade is a pivotal protective mechanism by NF-kappaB, and that this suppression involves the upregulation of gadd45beta/myd118. Induction of gadd45beta by stress and cytokines requires NF-kappaB; however, the regulatory mechanisms underlying this induction are not known. Here, we report that, in HeLa cells, the NF-kappaB subunit RelA is sufficient to activate gadd45beta expression, whereas Rel and p50 are not. Activation of gadd45beta by RelA depends on three kappaB elements at positions -447/-438 (kappaB-1), -426/-417 (kappaB-2), and -377/-368 (kappaB-3) of the gadd45beta promoter. Each of these sites binds to NF-kappaB complexes in vitro, and is required for optimal promoter transactivation. The data establish the direct participation of NF-kappaB in the regulation of Gadd45beta, thereby providing important mechanistic insights into the control of apoptosis by the transcription factor.
Collapse
Affiliation(s)
- Rongguan Jin
- The Gwen Knapp Center for Lupus and Immunology Research, and The Ben May Institute for Cancer Research, Committees on Immunology and Cancer Biology, The University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Frazier-Jessen MR, Thompson CD, Brown R, Rawat R, Nordan RP, Feldman GM. NF-kappaB elements contribute to junB inducibility by lipopolysaccharide in the murine macrophage cell line RAW264.7. FEBS Lett 2002; 513:203-7. [PMID: 11904151 DOI: 10.1016/s0014-5793(02)02295-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Macrophages respond to bacterial lipopolysaccharide (LPS) by activating latent cis-acting factors that initiate transcription of immediate early genes. One such immediate early gene, junB, is induced by LPS in macrophages within 30 min. To identify elements that mediate the induction of junB by LPS, upstream and downstream sequences flanking the junB gene were examined by transient expression in the RAW264.7 murine macrophage cell line using a luciferase reporter gene vector containing the junB minimal promoter. A >10-fold enhancement was associated with a 222 bp region downstream of the junB promoter in response to LPS. Transient reporter assays demonstrated that multiple nuclear factor (NF) kappaB sites are required for inducibility of junB by LPS in RAW264.7 cells. Electrophoretic mobility shift assays confirmed binding of LPS-induced nuclear proteins included p50/p65 heterodimers at these NF-kappaB sites.
Collapse
Affiliation(s)
- Michelle R Frazier-Jessen
- Laboratory of Immunobiology, Division of Monoclonal Antibodies, Office of Therapeutics Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20892, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Ray A, Yu GY, Ray BK. Cytokine-responsive induction of SAF-1 activity is mediated by a mitogen-activated protein kinase signaling pathway. Mol Cell Biol 2002; 22:1027-35. [PMID: 11809795 PMCID: PMC134650 DOI: 10.1128/mcb.22.4.1027-1035.2002] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
SAF-1, a zinc finger transcription factor, is activated by a number of inflammatory agents, including interleukin-1 (IL-1) and IL-6. It is involved in the cytokine-mediated transcriptional induction of serum amyloid A, an acute-phase plasma protein that is associated with the pathogenesis of reactive amyloidosis, rheumatoid arthritis, and atherosclerosis. Here, we show that the mitogen-activated protein (MAP) kinase signaling pathway regulates cytokine-mediated induction of the DNA-binding activity and transactivation potential of SAF-1. Phosphorylation of endogenous SAF-1 in response to IL-1 and IL-6 was markedly inhibited by the addition of MAP kinase inhibitors. Consistent with this finding, we show that a consensus MAP kinase phosphorylation site, PPTP, within SAF-1 could be phosphorylated by MAP kinase in vitro. To analyze the contribution of MAP kinase in the activation of SAF-1, we prepared two independent mutant proteins in which the threonine residue of the PPTP motif was altered to either valine or alanine. These mutant proteins lost the ability to be phosphorylated by MAP kinase both in vivo and in vitro and exhibited a significantly reduced ability to promote expression of the SAF-1-regulated promoter. While the DNA-binding activity of wild-type SAF-1 protein was markedly increased upon phosphorylation with MAP kinase, no such increase could be detected with the mutant SAF-1 proteins. Further analysis with the GAL-4 reporter system showed that mutation of the MAP kinase phosphorylation site considerably lowers the transactivation potential of SAF-1. Together, these results show that activation of SAF-1 in response to IL-1 and -6 is mediated via MAP kinase-regulated phosphorylation.
Collapse
Affiliation(s)
- Alpana Ray
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri 65211, USA.
| | | | | |
Collapse
|
45
|
Ray A, Kumar D, Ray BK. Promoter-binding activity of inflammation-responsive transcription factor SAF is regulated by cyclic AMP signaling pathway. DNA Cell Biol 2002; 21:31-40. [PMID: 11879578 DOI: 10.1089/10445490252810294] [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: 11/12/2022] Open
Abstract
The serum amyloid A activating factor (SAF) was identified as a family of inducible transcription factors that is activated by many mediators of inflammation. Its activation involves a phosphorylation event, whose mechanism is not fully understood. Here, we show that cAMP treatment of several cell types, including mouse liver-derived BNL CL.2, human monocyte-derived THP-1, and a primary culture of vascular smooth muscle cells from porcine aorta, activated cellular SAF's ability to bind DNA. The protein kinase A (PKA) activity in cytoplasmic extracts of cAMP-treated cells was responsible for the potentiation of the DNA-binding activity of the cellular SAF proteins. Furthermore, treatment of nuclear extracts of untreated cells with purified PKA increased the DNA-binding activity of cellular SAF proteins, and specific inhibitors of PKA abrogated the enhanced DNA-binding ability of SAF in the cAMP-treated cells. Consistent with these findings, overexpression of the catalytic subunit of PKA markedly increased expression of the SAF-regulated promoter. These results imply a functional role for the previously detected protein-protein interaction between SAF-1 transcription factor and the catalytic subunit of PKA and further demonstrate the consequences of cAMP-mediated signaling for the expression of SAF-regulated genes.
Collapse
Affiliation(s)
- Alpana Ray
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri 65211, USA
| | | | | |
Collapse
|
46
|
Kubota T, Hirama T, Verbeek W, Kawano S, Chih DY, Chumakov AM, Taguchi H, Koeffler HP. DNase I hypersensitivity analysis of the human CCAAT enhancer binding protein epsilon (C/EBPepsilon) gene. Leuk Res 2001; 25:981-95. [PMID: 11597733 DOI: 10.1016/s0145-2126(01)00065-0] [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: 12/14/2022]
Abstract
Human C/EBPepsilon is a recently cloned member of the C/EBP family of transcriptional factors. Previous studies demonstrated that the expression of this gene is tightly regulated in a tissue-specific manner; it is expressed almost exclusively in myeloid cells. To understand the mechanism by which the expression of C/EBPepsilon gene is controlled, we cloned a large genomic region surrounding the C/EBPepsilon gene and performed a DNase I hypersensitivity analysis of this locus. These sites probably represent areas of binding of proteins modulating gene transcription. Hypersensitive (HS) regions in 30 kb of DNA surrounding the C/EBPepsilon gene were examined in C/EBPepsilon high-expressing (NB4, HL-60), low-expressing (Jurkat), very-low-expressing (KG-1), and non-expressing (K562) hematopoietic cells as well as in non-hematopoietic-non-expressing cells (MCF-7, DU 145, PC-3). Three HS sites were detected near the first exon of C/EBPepsilon gene. They were found only in hematopoietic cells and were especially prominent in C/EBPepsilon expressing cells, suggesting that these sites play an important role in transcribing the gene. These hypersensitive bands did not change when the cells were cultured with retinoids. Gel-shift assays using 200 bp of nucleotide sequences that encompassed the hypersensitive sites and nuclear extracts from NB4 and Jurkat cells (C/EBPepsilon expressing) as well as K562 and MCF-7 cells (non-expressing) showed different retarded bands on gel electrophoresis. A fourth HS site, located about 11 kb upstream of exon 1, was found only in cells highly expressing C/EBPepsilon. Two sites, one about 4.5 kb upstream of exon 1 and another about 8.5 kb downstream of exon 2, were positive only in non-expressing cell lines, suggesting that repressors may bind in these areas. Taken together, we have found six specific DNase I hypersensitive sites in the region of C/EBPepsilon that may be involved in regulating transcription of this gene.
Collapse
Affiliation(s)
- T Kubota
- Division of Hematology/Oncology, Cedars-Sinai Research Institute, UCLA School of Medicine, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Vaidyanathan H, Gentry JD, Weatherman A, Schwartzbach SD, Petro TM. Differential response of the murine IL-12 p35 gene to lipopolysaccharide compared with interferon-gamma and CD40 ligation. Cytokine 2001; 16:1-9. [PMID: 11669581 DOI: 10.1006/cyto.2001.0938] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Expression of the heterodimeric cytokine interleukin-(IL-)12 is induced by pattern recognition receptors responding to microbial stimuli such as lipopolysaccharide (LPS) and products of the immune system such as interferon-gamma (IFN-gamma) and CD40L. The formation of bioactive IL-12 requires equimolar synthesis of p35 and p40 subunits. However, p35 expression limits the amount of IL-12 formed. Transcription of the gene for the p35 subunit of IL-12 initiates within the first exon, an alternate first exon (exon 1a), or second exon. Here we show that LPS and IFN-gamma/CD40 ligation increase the amount of total p35 mRNA in splenic adherent cells (SAC) to a similar extent. However, the exon 1 transcript was a smaller fraction of total p35 mRNA in IFN-gamma/CD40-stimulated cells than in unstimulated or LPS-stimulated cells. Despite comparable levels of total p35 mRNA, LPS-induced p35 exon 1 transcripts led to significantly more bioactive IL-12 from SAC than IFN-gamma/CD40-induced exon 1a/exon 2 transcripts as measured by ELISA. The data suggest that LPS-inducible p35 synthesis from exon 1 p35 transcripts leads to greater amount of bioactive IL-12 than IFN-gamma/CD40-induced p35 expression from alternate p35 exon 1a/exon 2 transcripts.
Collapse
Affiliation(s)
- H Vaidyanathan
- Center for Biological Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68583-0740, USA
| | | | | | | | | |
Collapse
|
48
|
Tsuzuki H, Tani T, Ueyama H, Kodama M. Lipopolysaccharide: neutralization by polymyxin B shuts down the signaling pathway of nuclear factor kappaB in peripheral blood mononuclear cells, even during activation. J Surg Res 2001; 100:127-34. [PMID: 11516215 DOI: 10.1006/jsre.2001.6227] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND There have been many studies on anti-lipopolysaccharide (LPS) agents and LPS-neutralizing agents; however, there have been no reports on the changes in clinical status and mediators that occur when these agents are used. Polymyxin (PMX) (treatment using a column containing polymyxin B-immobilized fiber) removed circulating endotoxin, and reduced various cytokines within 120 min, even in patients with high levels of plasma cytokines. Our purpose was examine the mechanisms of PMX treatment by which plasma cytokines are reduced by endotoxin neutralization with polymyxin B, even during therapy for sepsis and/or endotoxin shock. METHODS We studied the interaction between nuclear factor kappaB (NF-kappaB) binding activity and tumor necrosis factor alpha (TNF-alpha) secretion in an experimental system using LPS-stimulated human peripheral blood mononuclear cells (PBMCs), after neutralization of LPS with polymyxin B. PBMCs were incubated with LPS in vitro, and TNF-alpha secretion and NF-kappaB activation were assessed. We then studied the changes in NF-kappaB activation and TNF-alpha secretion when both polymyxin B and LPS were added simultaneously and when polymyxin B was added after 30 or 120 min of incubation with LPS. RESULTS Immediate inhibition of NF-kappaB binding activity and suppression of TNF-alpha secretion were observed after LPS neutralization with polymyxin B regardless of whether PBMCs were already producing TNF-alpha. CONCLUSIONS These findings may indicate one of the mechanisms operating in the clinical changes that occur after circulating endotoxin removal, and are likely to have therapeutic value, even for patients with high proinflammatory cytokine levels.
Collapse
Affiliation(s)
- H Tsuzuki
- First Department of Surgery, Shiga University of Medical Science, Seta-tsukinowa-cho, Otsu-city, Shiga, 520-2192, Japan.
| | | | | | | |
Collapse
|
49
|
Ely S, Bonatesta R, Ancsin JB, Kindy M, Kisilevsky R. The in-vitro influence of serum amyloid A isoforms on enzymes that regulate the balance between esterified and un-esterified cholesterol. Amyloid 2001; 8:169-81. [PMID: 11676294 DOI: 10.3109/13506120109007360] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The intracellular balance between un-esterified and esterified cholesterol is regulated by two enzyme activities, cholesterol ester hydrolases, which drive the balance in favor of un-esterified cholesterol, and acyl-CoA:cholesterol acyl transferase (ACAT) which acts in the opposite direction. During acute inflammation apo-serum amyloid A (apoSAA) isoforms 1.1 and 2.1 become major constituents of high density lipoprotein and this complex is internalized by macrophages. Mixtures of the two isoforms have been shown to enhance cholesterol esterase activity. Using a purified form of the pancreatic enzyme we have explored the mechanism by which apoSAA may accomplish this stimulation. The pancreatic esterase cleaves cholesteryl-oleate with a Km of 0.255 mM, releasing both cholesterol and oleate. Cholesterol exhibits a product inhibition which is relieved by isoform 2.1 but not 1.1 nor apolipoprotein A-I. The NH2-terminal 16 residues of isoform 2.1 had no effect on the esterase, but the 80 residue peptide constituting its COOH-terminus possessed the stimulatory property. Purified isoforms 1.1, 2.1, 2.2, apolipoprotein A-I, the NH2-terminal 16 residues and COOH-terminal 80 residues of isoform 2.1 were also examined for their effects on macrophage ACAT activity. Isoforms 2.1 and 2.2 produced dose dependent inhibitions of up to 50%, (p<0.001). Isoform 1.1, and apoA-I had no effect on ACAT activity. The NH2-terminal 16 residue peptide of isoform 2.1 reduced the ACAT activity in a dose dependent manner by 74% (p<0.001), whereas the COOH-terminal 80 residues, in contrast to its enhancing effect on the esterase, had no inhibitory effect on ACAT. Such complementary but opposite effects of isoform 2.1 on ACAT and the esterase are consistent with a role for this protein in shifting the balance between unesterified (transportable) and esterified (storage) forms of cholesterol in favor of the latter. They suggest that apoSAA2.1 may mediate cholesterol mobilization at sites of tissue injury.
Collapse
Affiliation(s)
- S Ely
- Department of Pathology, Queen's University, Kingston General Hospital, Ontario, Canada
| | | | | | | | | |
Collapse
|
50
|
Ray BK, Chen J, Ray A. Catalytic subunit of protein kinase A is an interacting partner of the inflammation-responsive transcription factor serum amyloid A-activating factor-1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:2343-8. [PMID: 11490023 DOI: 10.4049/jimmunol.167.4.2343] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Serum amyloid A-activating factor-1 (SAF-1) is a zinc finger transcription factor that is activated by many mediators of inflammation including IL-1, IL-6, and bacterial LPS. However, the mechanism of activation is not fully understood. To identify possible activation partners for SAF-1, we used a yeast two-hybrid system that detected interaction between the catalytic subunit of cyclic AMP-dependent protein kinase (PKA-Calpha) and SAF-1. Immunofluorescence and combined immunoprecipitation-Western blot analyses revealed colocalization and interaction between SAF-1 and PKA-Calpha. In vivo evidence of SAF-1 and PKA-Calpha interaction was further revealed by coimmunoprecipitation of these two proteins in cAMP-activated liver cells. We further show that SAF-1 is phosphorylated in vitro by PKA-Calpha and that addition of cAMP markedly induces in vivo phosphorylation of SAF-1 and transcription of SAF-regulated reporter genes. These results showed that SAF1-PKA-Calpha interaction is involved in functional activation of SAF-1.
Collapse
Affiliation(s)
- B K Ray
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | | | | |
Collapse
|