1
|
Bothe U, Günther J, Nubbemeyer R, Siebeneicher H, Ring S, Bömer U, Peters M, Rausch A, Denner K, Himmel H, Sutter A, Terebesi I, Lange M, Wengner AM, Guimond N, Thaler T, Platzek J, Eberspächer U, Schäfer M, Steuber H, Zollner TM, Steinmeyer A, Schmidt N. Discovery of IRAK4 Inhibitors BAY1834845 (Zabedosertib) and BAY1830839. J Med Chem 2024; 67:1225-1242. [PMID: 38228402 PMCID: PMC10823478 DOI: 10.1021/acs.jmedchem.3c01714] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 01/18/2024]
Abstract
Interleukin-1 receptor-associated kinase 4 (IRAK4) plays a critical role in innate inflammatory processes. Here, we describe the discovery of two clinical candidate IRAK4 inhibitors, BAY1834845 (zabedosertib) and BAY1830839, starting from a high-throughput screening hit derived from Bayer's compound library. By exploiting binding site features distinct to IRAK4 using an in-house docking model, liabilities of the original hit could surprisingly be overcome to confer both candidates with a unique combination of good potency and selectivity. Favorable DMPK profiles and activity in animal inflammation models led to the selection of these two compounds for clinical development in patients.
Collapse
Affiliation(s)
- Ulrich Bothe
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Judith Günther
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| | | | | | - Sven Ring
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| | | | - Michaele Peters
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| | | | - Karsten Denner
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Herbert Himmel
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Andreas Sutter
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Ildiko Terebesi
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| | | | - Antje M. Wengner
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Nicolas Guimond
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Tobias Thaler
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Johannes Platzek
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Uwe Eberspächer
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| | | | | | - Thomas M. Zollner
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Andreas Steinmeyer
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Nicole Schmidt
- Bayer AG, Research &
Development, Pharmaceuticals, 13353 Berlin, Germany
| |
Collapse
|
2
|
Prantner D, Shirey KA, Lai W, Lu W, Cole AM, Vogel SN, Garzino-Demo A. The θ-defensin retrocyclin 101 inhibits TLR4- and TLR2-dependent signaling and protects mice against influenza infection. J Leukoc Biol 2017; 102:1103-1113. [PMID: 28729359 PMCID: PMC5597516 DOI: 10.1189/jlb.2a1215-567rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 06/20/2017] [Accepted: 07/03/2017] [Indexed: 01/08/2023] Open
Abstract
A member of the θ‐defensin family protects mice during infection with influenza, suggesting a new strategy for viral therapy in humans. Despite widespread use of annual influenza vaccines, seasonal influenza‐associated deaths number in the thousands each year, in part because of exacerbating bacterial superinfections. Therefore, discovering additional therapeutic options would be a valuable aid to public health. Recently, TLR4 inhibition has emerged as a possible mechanism for protection against influenza‐associated lethality and acute lung injury. Based on recent data showing that rhesus macaque θ‐defensins could inhibit TLR4‐dependent gene expression, we tested the hypothesis that a novel θ‐defensin, retrocyclin (RC)‐101, could disrupt TLR4‐dependent signaling and protect against viral infection. In this study, RC‐101, a variant of the humanized θ‐defensin RC‐1, blocked TLR4‐mediated gene expression in mouse and human macrophages in response to LPS, targeting both MyD88‐ and TRIF‐dependent pathways. In a cell‐free assay, RC‐101 neutralized the biologic activity of LPS at doses ranging from 0.5 to 50 EU/ml, consistent with data showing that RC‐101 binds biotinylated LPS. The action of RC‐101 was not limited to the TLR4 pathway because RC‐101 treatment of macrophages also inhibited gene expression in response to a TLR2 agonist, Pam3CSK4, but failed to bind that biotinylated agonist. Mouse macrophages infected in vitro with mouse‐adapted A/PR/8/34 influenza A virus (PR8) also produced lower levels of proinflammatory cytokine gene products in a TLR4‐independent fashion when treated with RC‐101. Finally, RC‐101 decreased both the lethality and clinical severity associated with PR8 infection in mice. Cumulatively, our data demonstrate that RC‐101 exhibits therapeutic potential for the mitigation of influenza‐related morbidity and mortality, potentially acting through TLR‐dependent and TLR‐independent mechanisms.
Collapse
Affiliation(s)
- Daniel Prantner
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Kari Ann Shirey
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Wendy Lai
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Wuyuan Lu
- Department of Biochemistry and Molecular Biology, University of Maryland, School of Medicine, Baltimore, Maryland, USA.,Institute for Human Virology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Alexander M Cole
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, Florida, USA; and
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA;
| | - Alfredo Garzino-Demo
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA.,Institute for Human Virology, University of Maryland, School of Medicine, Baltimore, Maryland, USA.,Department of Molecular Medicine, University of Padova, Padova, Italy
| |
Collapse
|
3
|
Biswas R, Kumar P, Pollard HB. Regulation of mRNA turnover in cystic fibrosis lung disease. WILEY INTERDISCIPLINARY REVIEWS-RNA 2016; 8. [PMID: 27863009 DOI: 10.1002/wrna.1408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/13/2016] [Accepted: 10/16/2016] [Indexed: 01/07/2023]
Abstract
Cystic fibrosis (CF) is an autosomal recessive disease due to mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, F508del-CFTR being the most frequent mutation. The CF lung is characterized by a hyperinflammatory phenotype and is regulated by multiple factors that coordinate its pathophysiology. In CF the expression of CFTR as well as proinflammatory genes are regulated at the level of messenger RNA (mRNA) stability, which subsequently affect translation. These mechanisms are mediated by inflammatory RNA-binding proteins as well as small endogenous noncoding microRNAs, in coordination with cellular signaling pathways. These regulatory factors exhibit altered expression and function in vivo in the CF lung, and play a key role in the pathophysiology of CF lung disease. In this review, we have described the role of mRNA stability and associated regulatory mechanisms in CF lung disease. WIREs RNA 2017, 8:e1408. doi: 10.1002/wrna.1408 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Roopa Biswas
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Parameet Kumar
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Harvey B Pollard
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
4
|
Gorbachev AV, Fairchild RL. Regulation of chemokine expression in the tumor microenvironment. Crit Rev Immunol 2015; 34:103-20. [PMID: 24940911 DOI: 10.1615/critrevimmunol.2014010062] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Chemokines are chemotactic cytokines critical for homeostatic and inflammation-induced trafficking of leukocytes during immune responses, hematopoesis, wound healing, and tumorigenesis. Despite three decades of intensive study of the chemokine network, the molecular mechanisms regulating chemokine expression during tumor growth are not well understood. In this review, we focus on the role of chemokines in both tumor growth and anti-tumor immune responses and on molecular mechanisms employed by tumor cells to regulate chemokine expression in the tumor microenvironment. Multiple mechanisms used by tumors to regulate chemokine production, including those revealed by very recent studies (such as DNA methylation or post-translational nitrosylation of chemokines) are discussed. Concluding the review, we discuss how understanding of these regulatory mechanisms can be used in cancer therapy to suppress tumor growth and/or to promote immune-mediated eradication of tumors.
Collapse
Affiliation(s)
| | - Robert L Fairchild
- Department of Immunology and Urological Institute, Cleveland Clinic Foundation, Cleveland, OH 44195 and Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| |
Collapse
|
5
|
Caldwell AB, Cheng Z, Vargas JD, Birnbaum HA, Hoffmann A. Network dynamics determine the autocrine and paracrine signaling functions of TNF. Genes Dev 2014; 28:2120-33. [PMID: 25274725 PMCID: PMC4180974 DOI: 10.1101/gad.244749.114] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A hallmark of the inflammatory response to pathogen exposure is the production of tumor necrosis factor (TNF) that coordinates innate and adaptive immune responses by functioning in an autocrine or paracrine manner. Numerous molecular mechanisms contributing to TNF production have been identified, but how they function together in macrophages remains unclear. Here, we pursued an iterative systems biology approach to develop a quantitative understanding of the regulatory modules that control TNF mRNA synthesis and processing, mRNA half-life and translation, and protein processing and secretion. By linking the resulting model of TNF production to models of the TLR-, the TNFR-, and the NFκB signaling modules, we were able to study TNF's functions during the inflammatory response to diverse TLR agonists. Contrary to expectation, we predicted and then experimentally confirmed that in response to lipopolysaccaride, TNF does not have an autocrine function in amplifying the NFκB response, although it plays a potent paracrine role in neighboring cells. However, in response to CpG DNA, autocrine TNF extends the duration of NFκB activity and shapes CpG-induced gene expression programs. Our systems biology approach revealed that network dynamics of MyD88 and TRIF signaling and of cytokine production and response govern the stimulus-specific autocrine and paracrine functions of TNF.
Collapse
Affiliation(s)
- Andrew B Caldwell
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, and San Diego Center for Systems Biology, University of California at San Diego, La Jolla, California 92093, USA
| | - Zhang Cheng
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, and San Diego Center for Systems Biology, University of California at San Diego, La Jolla, California 92093, USA; Institute for Quantitative and Computational Biosciences, Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California 90025, USA
| | - Jesse D Vargas
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, and San Diego Center for Systems Biology, University of California at San Diego, La Jolla, California 92093, USA; Institute for Quantitative and Computational Biosciences, Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California 90025, USA
| | - Harry A Birnbaum
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, and San Diego Center for Systems Biology, University of California at San Diego, La Jolla, California 92093, USA; Institute for Quantitative and Computational Biosciences, Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California 90025, USA
| | - Alexander Hoffmann
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, and San Diego Center for Systems Biology, University of California at San Diego, La Jolla, California 92093, USA; Institute for Quantitative and Computational Biosciences, Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California 90025, USA
| |
Collapse
|
6
|
Herjan T, Novotny M, Hamilton TA. Diversity in sequence-dependent control of GRO chemokine mRNA half-life. J Leukoc Biol 2013; 93:895-904. [PMID: 23519936 DOI: 10.1189/jlb.0812370] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neutrophil trafficking to sites of injury or infection is regulated, in part, by the closely related GRO family of chemokines (CXCL1, -2, and -3). Expression of the GRO chemokine genes is known to be determined by transcriptional bursts in response to proinflammatory stimulation, but post-transcriptional mechanisms that regulate mRNA half-life are now recognized as important determinants. mRNA half-life is regulated via distinct sequence motifs and sequence-specific, RNA-binding proteins, whose function is subject to regulation by extracellular proinflammatory stimuli. Moreover, such mechanisms exhibit cell-type and stimulus dependency. We now present evidence that in nonmyeloid cells, GRO2 and GRO3 isoforms exhibit at least two patterns of mRNA instability that are distinguished by differential sensitivity to specific mRNA-destabilizing proteins and stimulus-mediated prolongation of mRNA half-life, respectively. Although the 3' UTR regions of GRO2 and GRO3 mRNAs contain multiple AREs, GRO2 has eight AUUUA pentamers, whereas GRO3 has seven. These confer quantitative differences in half-life and show sensitivity for TTP and KSRP but not SF2/ASF. Moreover, these AUUUA determinants do not confer instability that can be modulated in response to IL-1α. In contrast, IL-1α-sensitive instability for GRO2 and GRO3 is conferred by sequences located proximal to the 3' end of the 3'UTR that are independent of the AUUUA sequence motif. These regions are insensitive to TTP and KSRP but show reduced half-life mediated by SF2/ASF. These sequence-linked, post-transcriptional activities provide substantial mechanistic diversity in the control of GRO family chemokine gene expression.
Collapse
Affiliation(s)
- Tomasz Herjan
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | |
Collapse
|
7
|
Li K, Li NL, Wei D, Pfeffer SR, Fan M, Pfeffer LM. Activation of chemokine and inflammatory cytokine response in hepatitis C virus-infected hepatocytes depends on Toll-like receptor 3 sensing of hepatitis C virus double-stranded RNA intermediates. Hepatology 2012; 55:666-75. [PMID: 22030901 PMCID: PMC3272326 DOI: 10.1002/hep.24763] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 10/04/2011] [Indexed: 12/14/2022]
Abstract
UNLABELLED Chemokines and inflammatory cytokines are key regulators of immunity and inflammation during viral infections. Hepatitis C virus (HCV) is a hepatotropic RNA virus frequently associated with chronic liver inflammation and hepatocellular carcinoma. Intrahepatic levels of chemokines and cytokines are elevated in chronic HCV infections, but the underlying mechanisms remain unclear. We found that Toll-like receptor-3 (TLR3) senses HCV infection in cultured hepatoma cells, leading to nuclear factor kappa B (NF-κB) activation and the production of numerous chemokines and inflammatory cytokines, such as regulated on activation normal T cell expressed and secreted (RANTES), macrophage inflammatory protein (MIP)-1α, MIP-1β, IP-10, and interleukin-6. The chemokine/cytokine induction occurred late in HCV infection and was abrogated when HCV was ultraviolet-inactivated before infection, indicating a dependence on the cellular recognition of HCV replication products. Gel-shift and chromatin immunoprecipitation assays revealed that NF-κB plays a pivotal role in HCV-induced chemokine/cytokine transcription. Mutations specifically disrupting the double-stranded RNA (dsRNA)-binding activity of TLR3 ablated the chemokine/cytokine response to HCV infection, indicating that HCV dsRNA was the pathogen-associated molecular pattern triggering TLR3 signaling. In vitro synthesized HCV dsRNAs, with a minimal length of ∼80-100 base pairs, activated TLR3-dependent chemokine expression, regardless of the genome position from which they derived. In contrast, HCV single-stranded RNAs, including those derived from the structured 3'nontranslated region highly potent for RIG-I activation, failed to do so. Moreover, robust production of chemokines and inflammatory cytokines was also observed in primary human hepatocytes after stimulation with extracellular poly-I:C, a TLR3 ligand. CONCLUSION Our data suggest that TLR3-mediated chemokine and inflammatory cytokine responses may play an important role in host immune responses to HCV and the pathogenesis of HCV-associated liver diseases.
Collapse
Affiliation(s)
- Kui Li
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Nan L. Li
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Dahai Wei
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Susan R. Pfeffer
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Meiyun Fan
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Lawrence M. Pfeffer
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
8
|
Hamilton T, Li X, Novotny M, Pavicic PG, Datta S, Zhao C, Hartupee J, Sun D. Cell type- and stimulus-specific mechanisms for post-transcriptional control of neutrophil chemokine gene expression. J Leukoc Biol 2011; 91:377-83. [PMID: 22167720 DOI: 10.1189/jlb.0811404] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
mRNAs encoding inflammatory chemokines that recruit neutrophils frequently exhibit short half-lives that serve to limit their expression under inappropriate conditions but are often prolonged to ensure adequate levels during inflammatory response. Extracellular stimuli that modulate the stability of such mRNAs may be the same as the transcriptional activator, as is the case with TLR ligands, or may cooperate with independent transcriptional stimuli, as with IL-17, which extends the half-life of TNF-induced transcripts. These different stimuli engage independent signaling pathways that target different instability mechanisms distinguished by dependence on different regulatory nucleotide sequence motifs within the 3'UTRs, which involve that action of different mRNA-binding proteins. The selective use of these pathways by different stimuli and in distinct cell populations provides the potential for tailoring of chemokine expression patterns to meet specific needs in different pathophysiologic circumstances.
Collapse
Affiliation(s)
- Thomas Hamilton
- Cleveland Clinic Foundation, 9500 Euclid Ave., Cleveland, OH 44195-0001, USA.
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Ryzhakov G, Lai CCK, Blazek K, To KW, Hussell T, Udalova I. IL-17 boosts proinflammatory outcome of antiviral response in human cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:5357-62. [PMID: 21964025 DOI: 10.4049/jimmunol.1100917] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Excessive inflammation during bacterial and viral infections is destructive to the host and involves elevated production of proinflammatory cytokines. It is especially deleterious in organs with space constraints such as lung and the CNS. Indeed, a number of viruses that infect lungs, such as avian influenza virus, SARS-associated coronavirus, and respiratory syncytial virus, elicit a very high level of proinflammatory cytokines; however, it is unclear what triggers their production. In this study, we show that IL-17 commonly produced during viral infection specifically augments a proinflammatory response by directly synergizing with antiviral signaling. Costimulation of primary human fibroblasts with IL-17 greatly enhanced respiratory syncytial virus-induced or synthetic dsRNA-based viral mimic polyinosinic:polycytidylic acid-induced expression of proinflammatory genes without affecting expression of IFN-β-stimulated or IFN-stimulated genes. Knockdown of expression of known mediators of the antiviral signaling pathway revealed that the IL-17-poly(I:C) synergy depends on the presence of the transcriptional factors RelA and IFN regulatory factor 3 and IκB kinases. Moreover, this synergy was blocked by an IκB kinase inhibitor, BAY 11-7082. These findings shed light on the molecular mechanisms behind IL-17-dependent immunopathology observed in viral infections.
Collapse
Affiliation(s)
- Grigory Ryzhakov
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, London W6 8LH, UK.
| | | | | | | | | | | |
Collapse
|
10
|
Flannery SM, Keating SE, Szymak J, Bowie AG. Human interleukin-1 receptor-associated kinase-2 is essential for Toll-like receptor-mediated transcriptional and post-transcriptional regulation of tumor necrosis factor alpha. J Biol Chem 2011; 286:23688-97. [PMID: 21606490 DOI: 10.1074/jbc.m111.248351] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Toll-like receptors (TLRs) are pattern-recognition receptors that recognize microbial ligands and subsequently trigger intracellular signaling pathways involving transcription factors such as NFκB and MAPKs such as p38. TLR signaling can regulate both transcriptional and post-transcriptional events leading to altered gene expression and thus appropriate immune responses. The interleukin-1 receptor-associated kinase (IRAK) family comprises four kinases that regulate TLR signaling. However, the role of IRAK-2 has remained unclear, especially in human cells. Recent studies using cells from in-bred Irak2(-/-) mice showed that murine IRAK-2 was not required for early TLR signaling events but had a role in delayed NFκB activation and in cytokine production. IRAK-2 in mice has four splice variants, two of which are inhibitory, whereas human IRAK-2 has no splice variants. Thus IRAK-2 in mice and humans may function differently, and therefore we analyzed the role of IRAK-2 in TLR responses in primary human cells. siRNA knockdown of IRAK-2 expression in human peripheral blood mononuclear cells showed a role for human IRAK-2 in both TLR4- and TLR8-mediated early NFκB and p38 MAPK activation and in induction of TNF mRNA. These data conflict with findings from the in-bred Irak2(-/-) mice but concur with what has been seen in wild-derived mice for TLR2. Moreover, human IRAK-2 was required for regulating MyD88-dependent TNFα mRNA stability via the TNF 3'UTR. Collectively, these data demonstrate for the first time an essential role for IRAK-2 in primary human cells for both transcriptional and post-transcriptional TLR responses.
Collapse
Affiliation(s)
- Sinead M Flannery
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland
| | | | | | | |
Collapse
|
11
|
Khabar KSA. Post-transcriptional control during chronic inflammation and cancer: a focus on AU-rich elements. Cell Mol Life Sci 2010; 67:2937-55. [PMID: 20495997 PMCID: PMC2921490 DOI: 10.1007/s00018-010-0383-x] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 04/01/2010] [Accepted: 04/21/2010] [Indexed: 12/21/2022]
Abstract
A considerable number of genes that code for AU-rich mRNAs including cytokines, growth factors, transcriptional factors, and certain receptors are involved in both chronic inflammation and cancer. Overexpression of these genes is affected by aberrations or by prolonged activation of several signaling pathways. AU-rich elements (ARE) are important cis-acting short sequences in the 3'UTR that mediate recognition of an array of RNA-binding proteins and affect mRNA stability and translation. This review addresses the cellular and molecular mechanisms that are common between inflammation and cancer and that also govern ARE-mediated post-transcriptional control. The first part examines the role of the ARE-genes in inflammation and cancer and sequence characteristics of AU-rich elements. The second part addresses the common signaling pathways in inflammation and cancer that regulate the ARE-mediated pathways and how their deregulations affect ARE-gene regulation and disease outcome.
Collapse
Affiliation(s)
- Khalid S A Khabar
- Program in BioMolecular Research, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia.
| |
Collapse
|
12
|
Hamilton T, Novotny M, Pavicic PJ, Herjan T, Hartupee J, Sun D, Zhao C, Datta S. Diversity in post-transcriptional control of neutrophil chemoattractant cytokine gene expression. Cytokine 2010; 52:116-22. [PMID: 20430641 DOI: 10.1016/j.cyto.2010.04.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 04/05/2010] [Indexed: 12/11/2022]
Abstract
Regulation of neutrophil chemokine gene expression represents an important feature in tissue inflammation. While chemokine gene transcription through the action of NFkappaB is recognized as an essential component of this process, it is now clear that post-transcriptional mechanisms, particularly the rates of decay of mature cytoplasmic mRNA, provides an essential component of this control. Chemokine and other cytokine mRNA half life is known to be controlled via adenine-uridine rich sequence motifs localized within 3' untranslated regions (UTRs), the most common of which contains one or more copies of the pentameric AUUUA sequence. In myeloid cells AUUUA sequences confer instability through the action of RNA binding proteins such as tristetraprolin (TTP). The resulting instability can be regulated in response to extra-cellular stimuli including Toll like receptor ligands that signal to control the function of TTP through pathways involving the activation of p38 MAP kinases. Recent findings indicate that substantial mechanistic diversity is operative in non-myeloid cells in response to alternate pro-inflammatory stimuli such as IL-17. These pathways target distinct instability sequences that do not contain the AUUUA pentamer motif, do not signal through p38 MAPK, and function independently of TTP.
Collapse
Affiliation(s)
- Thomas Hamilton
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Datta S, Novotny M, Pavicic PG, Zhao C, Herjan T, Hartupee J, Hamilton T. IL-17 regulates CXCL1 mRNA stability via an AUUUA/tristetraprolin-independent sequence. THE JOURNAL OF IMMUNOLOGY 2009; 184:1484-91. [PMID: 20042592 DOI: 10.4049/jimmunol.0902423] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
IL-17 contributes to inflammatory response in part by promoting enhanced expression of chemokines, such as CXCL1, by prolonging the t(1/2) of this constitutively unstable mRNA. Although IL-17 is a weak stimulus for transcription of the CXCL1 gene, it strongly potentiates message accumulation via stabilization when the mRNA is transcribed in cells stimulated with TNF. In myeloid cells, LPS-induced CXCL1 mRNA stabilization is dependent on AUUUA-containing sequence motifs that are recognized by the RNA binding protein tristetraprolin (TTP). Using deletion and site-specific mutagenesis, we report that IL-17-mediated stabilization of CXCL1 mRNA in nonmyeloid cells depends on a sequence that does not contain the AUUUA motif. Furthermore, a specific two-nucleotide mutation within this region markedly abrogates sensitivity for IL-17-mediated stabilization. Consistent with this finding, the IL-17-sensitive sequence does not exhibit increased instability in the presence of TTP, and CXCL1 mRNA remains unstable and can be stabilized in response to treatment with IL-17 in embryo fibroblasts from mice in which the TTP gene has been deleted. Whereas the RNA binding protein KSRP has been shown to participate in regulating the instability of human CXCL8 mRNA, inhibitory RNA-based reduction in KSRP does not effect the instability mediated by the IL-17-sensitive sequence motif. These findings suggest that IL-17-mediated chemokine mRNA stabilization in nonmyeloid cells uses a mechanism that is distinct from that operating to control AU-rich mRNA stability in myeloid cells.
Collapse
Affiliation(s)
- Shyamasree Datta
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
The effects of interleukin-1beta in tumor necrosis factor-alpha-induced acute pulmonary inflammation in mice. Mediators Inflamm 2009; 2009:958658. [PMID: 19901996 PMCID: PMC2773377 DOI: 10.1155/2009/958658] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Revised: 07/08/2009] [Accepted: 08/12/2009] [Indexed: 12/21/2022] Open
Abstract
We determined the role of interleukin-1β (IL-1β) signaling on tumor necrosis factor alpha-induced (TNF-α) lung neutrophil influx as well as neutrophil chemoattractant macrophage inflammatory protein (MIP-2) and KC and soluble TNF-α receptor (TNFR) levels utilizing wildtype (WT), TNF receptor double knockout (TNFR1/TNFR2 KO), and IL-1β KO mice after oropharyngeal instillation with TNF-α. A significant increase in neutrophil accumulation in bronchoalveolar lavage fluid (BALF) and lung interstitium was detected in the WT mice six hours after TNF-α exposure. This correlated with an increase in BALF MIP-2. In contrast, BALF neutrophil numbers were not increased by TNF-α treatment of IL-1β KOs, correlating with a failure to induce BALF MIP-2 and a trend toward increased BALF soluble TNFR1. TNF-α-instillation increased lavage and serum KC and soluble TNFR2 irrespective of IL-1β expression. These results suggest IL-1β contributes, in part, to TNF-α-mediated, chemokine release, and neutrophil recruitment to the lung, potentially associated with altered soluble TNFR1 release into the BALF.
Collapse
|
15
|
Balakathiresan NS, Bhattacharyya S, Gutti U, Long RP, Jozwik C, Huang W, Srivastava M, Pollard HB, Biswas R. Tristetraprolin regulates IL-8 mRNA stability in cystic fibrosis lung epithelial cells. Am J Physiol Lung Cell Mol Physiol 2009; 296:L1012-8. [PMID: 19363120 DOI: 10.1152/ajplung.90601.2008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cystic fibrosis (CF) is due to mutations in the CFTR gene and is characterized by hypersecretion of the proinflammatory chemokine IL-8 into the airway lumen. Consequently, this induces the highly inflammatory cellular phenotype typical of CF. Our initial studies revealed that IL-8 mRNA is relatively stable in CF cells compared with those that had been repaired with [WT]CFTR (wild-type CFTR). Relevantly, the 3'-UTR of IL-8 mRNA contains AU-rich sequences (AREs) that have been shown to mediate posttranscriptional regulation of proinflammatory genes upon binding to ARE-binding proteins including Tristetraprolin (TTP). We therefore hypothesized that very low endogenous levels of TTP in CF cells might be responsible for the relative stability of IL-8 mRNA. As predicted, increased expression of TTP in CF cells resulted in reduced stability of IL-8 mRNA. An in vitro analysis of IL-8 mRNA stability in CF cells also revealed a TTP-induced enhancement of deadenylation causing reduction of IL-8 mRNA stability. We conclude that enhanced stability of IL-8 mRNA in TTP-deficient CF lung epithelial cells serve to drive the proinflammatory cellular phenotype in the CF lung.
Collapse
Affiliation(s)
- Nagaraja Sethuraman Balakathiresan
- Departments of Health Systems, Risk, and Contingency Management, Graduate School of Nursing, Uniformed Services Univ. of the Health Sciences, Bethesda, MD 20814, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Hartupee J, Liu C, Novotny M, Sun D, Li X, Hamilton TA. IL-17 signaling for mRNA stabilization does not require TNF receptor-associated factor 6. THE JOURNAL OF IMMUNOLOGY 2009; 182:1660-6. [PMID: 19155515 DOI: 10.4049/jimmunol.182.3.1660] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
IL-17 alone is a relatively weak inducer of gene expression, but cooperates with other cytokines, including TNF-alpha, to generate a strong response in part via prolongation of mRNA t(1/2). Because TNFR-associated factor 6 (TRAF6) has been reported to be essential for signaling by IL-17, we examined its involvement in IL-17-mediated mRNA stabilization. Although overexpression of TRAF6 in HeLa cells activates NF-kappaB, it does not stabilize transfected KC mRNA. Furthermore, a dominant-negative TRAF6 abrogates NF-kappaB activation, but does not block IL-17-induced chemokine mRNA stabilization. IL-17 can stabilize KC and MIP-2 mRNAs comparably in TNF-alpha-treated mouse embryo fibroblasts from TRAF6(+/+) and TRAF6(-/-) mice. TRAF6 is known to couple upstream signals with activation of p38 MAPK and mitogen activated protein kinase activated protein kinase 2, both of which have been shown to be important for Toll/IL-1R-mediated mRNA stabilization in various cell types. Inhibition of p38 MAPK, however, does not block IL-17-induced KC mRNA stabilization, and IL-17 can stabilize KC mRNA equally in mouse embryo fibroblasts from both wild-type and mitogen activated protein kinase activated protein kinase 2/3 doubly-deficient mice. Finally, IL-17 can amplify the levels of multiple TNF-alpha-stimulated mRNAs in wild-type and TRAF6-deficient cells, but not in cells from Act1(-/-) mice. Collectively, these findings demonstrate the existence of a TRAF6/p38 MAPK-independent pathway that couples the IL-17R with enhanced mRNA stability. Because the most potent effects of IL-17 on gene expression are obtained in cooperation with other cytokines such as TNF-alpha, these findings suggest that this pathway is a major contributing mechanism for response to IL-17.
Collapse
Affiliation(s)
- Justin Hartupee
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | | | |
Collapse
|
17
|
Yamamoto M, Akira S. Lipid A receptor TLR4-mediated signaling pathways. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 667:59-68. [PMID: 20665200 DOI: 10.1007/978-1-4419-1603-7_6] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Lipid A is a strong activator of monocytes to release immune stimulators such as proinflammatory cytokines. Overproduction of inflammatory cytokines such as TNF and IL-6 is known to cause septic shock that frequently leads to multiple organ failure and finally to death. In recent years, Lipid A has also been recognized by a Toll-like receptor, TLR4. Activation of TLR4by LPS or Lipid A triggers signal transduction via the cytoplasmic domain called the Toll/IL-1 Receptor (TIR) domain. Intracellular TIR domain-containing adaptor molecules are involved in the TLR4-mediated signaling pathways. Moreover, a subset of LPS-inducible genes is regulated in two steps by the inducible nuclear protein. Additionally, the TLR4-mediated activation of signaling cascadesis elaborately down-regulated by a number of negative regulators. In this chapter, we discuss the mechanisms of the activation or de-activation program mediated by the Lipid A receptor TLR4.
Collapse
Affiliation(s)
- Masahiro Yamamoto
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | | |
Collapse
|
18
|
Datta S, Biswas R, Novotny M, Pavicic PG, Herjan T, Mandal P, Hamilton TA. Tristetraprolin regulates CXCL1 (KC) mRNA stability. THE JOURNAL OF IMMUNOLOGY 2008; 180:2545-52. [PMID: 18250465 DOI: 10.4049/jimmunol.180.4.2545] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
mRNAs encoding proinflammatory chemokines are regulated posttranscriptionally via adenine-uridine-rich sequences (AREs) located in the 3' untranslated region of the message, which are recognized by sequence-specific RNA-binding proteins. One ARE binding protein, tristetraprolin (TTP), has been implicated in regulating the stability of several ARE-containing mRNAs, including those encoding TNF-alpha and GM-CSF. In the present report we examined the role of TTP in regulating the decay of the mouse chemokine KC (CXCL1) mRNA. Using tetR-regulated control of transcription in TTP-deficient HEK293 cells, KC mRNA half-life was markedly decreased in the presence of TTP. Deletion and site-specific mutagenesis were used to identify multiple AUUUA sequence determinants responsible for TTP sensitivity. Although a number of studies suggest that the destabilizing activity of TTP is subject to modulation in response to ligands of Toll/IL-1 family receptors, decay mediated by TTP in 293 cells was not sensitive to stimulation with IL-1alpha. Using primary macrophages from wild-type and TTP-deficient mice, KC mRNA instability was found to be highly dependent on TTP. Furthermore, LPS-mediated stabilization of KC mRNA is blocked by inhibition of the p38 MAPK in macrophages from wild-type but not TTP-deficient mice. These findings demonstrate that TTP is the predominant regulator of KC mRNA decay in mononuclear phagocytes acting via multiple 3'-untranslated region-localized AREs. Nevertheless, KC mRNA remains highly unstable in cells that do not express TTP, suggesting that additional determinants of instability and stimulus sensitivity may operate in cell populations where TTP is not expressed.
Collapse
Affiliation(s)
- Shyamasree Datta
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Hartupee J, Li X, Hamilton T. Interleukin 1alpha-induced NFkappaB activation and chemokine mRNA stabilization diverge at IRAK1. J Biol Chem 2008; 283:15689-93. [PMID: 18411265 DOI: 10.1074/jbc.m801346200] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Interleukin 1alpha (IL-1alpha) is capable of driving pro-inflammatory gene expression through both the initiation of transcription and by prolonging the half-life of short-lived mRNAs. Although the signaling events linking the IL-1 receptor to the activation of NFkappaB and the initiation of transcription have been well characterized, less is known about the signaling events linking to mRNA stabilization. As a model to study the control of mRNA stability we have used the mouse chemokine KC, expression of which requires both NFkappaB-driven transcription and stabilization of the constitutively unstable mRNA. We have evaluated the role of signaling adaptors known to play a role in IL-1alpha-driven NFkappaB activation in the generation of mRNA stability. Surprisingly, although TRAF6 is essential for NFkappaB activation, it is not required for IL-1alpha-induced mRNA stabilization. IRAK1, which is recognized to function upstream of TRAF6, is required for both mRNA stabilization and activation of NFkappaB. Consistent with the previous findings, the TRAF6 interaction sites in IRAK1 are required for NFkappaB activation but do not play a role in mRNA stabilization. These findings indicate that signals from the IL-1 receptor segregate into at least two separate pathways at the level of IRAK1; one couples through TRAF6 to NFkappaB activation while a second utilizes a TRAF6-independent pathway that is responsible for mRNA stabilization.
Collapse
Affiliation(s)
- Justin Hartupee
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | |
Collapse
|
20
|
Hartupee J, Liu C, Novotny M, Li X, Hamilton T. IL-17 enhances chemokine gene expression through mRNA stabilization. THE JOURNAL OF IMMUNOLOGY 2007; 179:4135-41. [PMID: 17785852 DOI: 10.4049/jimmunol.179.6.4135] [Citation(s) in RCA: 225] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
IL-17 plays an important role in host defense and autoimmunity via the induction of proinflammatory gene expression, particularly in combination with TNF-alpha. The molecular mechanisms by which IL-17 regulates such expression are not well understood. Using the mouse chemokine CXCL1 (KC) gene as a model, we have examined the effects of IL-17 alone or in combination with TNF-alpha on transcriptional and posttranscriptional events. Although treatment of mouse embryonic fibroblasts with IL-17 alone only modestly increased KC expression, the combination of IL-17 with TNF-alpha induced a synergistic response. IL-17 treatment exerted a strong posttranscriptional effect by extending the t1/2 of the highly unstable, TNF-alpha-induced KC mRNA. Using a tetracycline-regulated transgene in HeLa cells, we determined that IL-17 treatment alone promoted stabilization of KC mRNA in the absence of TNF-alpha. IL-17 treatment exerted little effect on KC transcription or NF-kappaB activation, suggesting that it primarily acts posttranscriptionally. We identified a number of other mRNAs whose t1/2 are prolonged in response to IL-17, suggesting that this is a common mechanism by which IL-17 promotes enhanced gene expression. Finally, activator of NF-kappaB1 protein (Act1), an adaptor protein recently implicated in IL-17 signaling, was necessary for IL-17-induced stabilization, and overexpression of Act1 resulted in stabilization of KC mRNA, indicating that events downstream of Act1 are sufficient to initiate this process. Thus, the synergy between TNF-alpha and IL-17 reflects their independent actions on KC gene expression; TNF-alpha serves as a stimulus to initiate transcription through activation of NF-kappaB, whereas IL-17 drives mRNA stabilization through an Act1-dependent pathway.
Collapse
Affiliation(s)
- Justin Hartupee
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | |
Collapse
|
21
|
Abstract
The expression of neutrophil-specific chemokines is known to be regulated via adenine-uridine-rich sequence elements in the 3'-untranslated regions of their mRNAs that confer a high degree of mRNA instability. Although the presence of intron sequences in eukaryotic genes is known to enhance expression, the effect of intron content on the rate of mature, translatable mRNA degradation has not been demonstrated. In this study, we have determined the effects of intron content on the rate of decay of the chemokine CXCL1 (KC) mRNA. The half-life of KC mRNA was markedly prolonged when the primary transcript was obtained from a genomic clone containing three introns as compared with the half-life observed with sequence-identical KC mRNA derived from an intron-free cDNA construct. The effect of intron content was achieved with a single intron, and neither the intron sequences nor the intron positions were critical determinants of the outcome. The intron content produced the same effect when expressed in multiple cell types and when the sequences were stably integrated into the genome. The differential decay rates were not a consequence of differential nuclear to cytoplasmic transport. The intron content of the primary transcript did not influence the rate of KC mRNA translation and did not modulate the ability of interleukin-1 stimulation to stabilize the otherwise unstable mRNA. The intron effect on mRNA decay was seen with mRNAs containing two distinct instability determinants. These findings document that intron content marks the mRNA sequence leading to enhanced stability that is particularly evident in short lived ARE-containing mRNAs.
Collapse
Affiliation(s)
- Chenyang Zhao
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | | |
Collapse
|
22
|
Hamilton TA, Novotny M, Datta S, Mandal P, Hartupee J, Tebo J, Li X. Chemokine and chemoattractant receptor expression: post-transcriptional regulation. J Leukoc Biol 2007; 82:213-9. [PMID: 17409125 DOI: 10.1189/jlb.1206754] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The magnitude and character of the inflammatory process are determined in part via the trafficking of leukocytes into sites of injury and infection, and this process depends on proper control of the expression of genes encoding chemoattractant peptides and their receptors. Although these controls operate at multiple mechanistic levels, recent evidence indicates that post-transcriptional events governing the half-life of select mRNAs are important determinants. Adenine-uridine rich elements (AREs) located within 3' untranslated regions (UTRs) confer constitutive mRNA instability and in some cases, stabilization following stimulation by ligands of the Toll-IL-1 receptor (TIR) family. Although the importance of AREs in determining activity and mRNA half-life is well-recognized, the mechanistic scope and diversity remain poorly understood. Using the mouse KC or CXCL1 gene as a model, we have demonstrated that the abundance of mRNA and protein produced during an inflammatory response depends on multiple mechanistically distinct AREs present in the 3' UTR of the mRNA. The mRNA encoding the receptor for N-terminal formyl-methionine-containing peptides is also unstable and subject to stabilization in response to TIR ligands. These two models can, however, be readily distinguished from one another on the basis of specific stimulus sensitivity and the signaling pathways, through which such stimuli couple to the control of mRNA decay. These models demonstrate the substantial diversity operative in the post-transcriptional regulation of inflammatory gene expression.
Collapse
Affiliation(s)
- Thomas A Hamilton
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
There have been recent, significant advances about the role of mRNA turnover in controlling gene expression in immune cells. Post‐transcriptional regulation of gene expression contributes to the characteristics of many of the processes underlying the immune response by ensuring early, rapid, and transient action. The emphasis of this review is on current work that deals with the regulation of mRNA decay during innate immunity against microbes and T cell activation as a model of the adaptive response.
Collapse
Affiliation(s)
- Khalid S A Khabar
- Program in BioMolecular Research, King Faisal Specialist Hospital and Research Center, P3354, mBC-03, Riyadh 11211, Saudi Arabia.
| |
Collapse
|
24
|
Mandal P, Hamilton T. Signaling in Lipopolysaccharide-Induced Stabilization of Formyl Peptide Receptor 1 mRNA in Mouse Peritoneal Macrophages. THE JOURNAL OF IMMUNOLOGY 2007; 178:2542-8. [PMID: 17277163 DOI: 10.4049/jimmunol.178.4.2542] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To identify the TLR4-initiated signaling events that couple to formyl peptide receptor (FPR)1 mRNA stabilization, macrophages were treated with LPS along with a selection of compounds targeting several known signaling pathways. Although inhibitors of protein tyrosine kinases, MAPKs, and stress-activated kinases had little or no effect on the response to LPS, LY294002 (LY2) and parthenolide (an IkappaB kinase inhibitor) were both potent inhibitors. LY2 but not parthenolide blocked the LPS-induced stabilization of FPR1 mRNA. Although both LY2 and wortmannin effectively blocked PI3K activity, wortmannin had little effect on FPR1 expression and did not modulate the decay of FPR1 mRNA. Moreover, although LY2 was demonstrated to be a potent inhibitor of PI3K activity, a structural analog of LY2, LY303511 (LY3), which did not inhibit PI3K, was equally effective at preventing LPS-stimulated FPR1 expression. The mammalian target of rapamycin activity (measured as phospho-p70S6 kinase) was activated by LPS but not significantly blocked by LY2. In addition, although rapamycin blocked mTOR activity, it did not inhibit FPR1 mRNA expression. Finally, the mechanisms involved in stabilization of FPR1 by LPS could be distinguished from those involved in stabilization of AU-rich mRNAs because the prolonged half-life of FPR1 mRNA was insensitive to the inhibition of p38 MAPK. These findings demonstrate that LY2/LY3 targets a novel TLR4-linked signaling pathway that selectively couples to the stabilization of FPR1 mRNA.
Collapse
MESH Headings
- 3' Untranslated Regions/immunology
- 3' Untranslated Regions/metabolism
- Androstadienes/pharmacology
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Cell-Free System/immunology
- Cell-Free System/metabolism
- Chromones/pharmacology
- Female
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/immunology
- I-kappa B Kinase/antagonists & inhibitors
- I-kappa B Kinase/immunology
- I-kappa B Kinase/metabolism
- Lipopolysaccharides/pharmacology
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Mice
- Morpholines/pharmacology
- Phosphatidylinositol 3-Kinases/immunology
- Phosphoinositide-3 Kinase Inhibitors
- Piperazines/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Protein Kinases/immunology
- Protein Kinases/metabolism
- RNA Stability/drug effects
- RNA Stability/immunology
- RNA, Messenger/immunology
- RNA, Messenger/metabolism
- Receptors, Formyl Peptide/biosynthesis
- Receptors, Formyl Peptide/immunology
- Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors
- Ribosomal Protein S6 Kinases, 70-kDa/immunology
- Ribosomal Protein S6 Kinases, 70-kDa/metabolism
- Sesquiterpenes/pharmacology
- Signal Transduction/drug effects
- Signal Transduction/immunology
- TOR Serine-Threonine Kinases
- Toll-Like Receptor 4/immunology
- Toll-Like Receptor 4/metabolism
- Wortmannin
- p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
- p38 Mitogen-Activated Protein Kinases/immunology
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Palash Mandal
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | |
Collapse
|
25
|
Sun D, Ding A. MyD88-mediated stabilization of interferon-gamma-induced cytokine and chemokine mRNA. Nat Immunol 2006; 7:375-81. [PMID: 16491077 DOI: 10.1038/ni1308] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2005] [Accepted: 01/09/2006] [Indexed: 11/08/2022]
Abstract
The MyD88 adaptor protein is critical in Toll-like receptor and interleukin 1 receptor (IL-1R) signaling, but has not been linked to interferon-gamma receptor (IFN-gammaR) signaling. Here we demonstrate that MyD88 increased the half-life but not the synthesis of IFN-gamma-induced mRNA transcripts encoding tumor necrosis factor and IFN-gamma-inducible protein 10. IFN-gamma stimulation triggered a physical association between the IFN-gammaR1 and MyD88. Transcript stabilization required activation of mixed-lineage kinase 3 and p38 mitogen-activated protein kinase and the presence of an adenine-uridine-rich element in the transcript's 3' untranslated region. These results demonstrate a MyD88-dependent post-transcriptional mechanism through which IFN-gamma can enhance the expression of genes encoding proinflammatory molecules.
Collapse
MESH Headings
- 3' Untranslated Regions
- Adaptor Proteins, Signal Transducing/immunology
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Chemokine CXCL10
- Chemokines, CXC/biosynthesis
- Chemokines, CXC/genetics
- Chemokines, CXC/immunology
- Heterogeneous Nuclear Ribonucleoprotein D0
- Heterogeneous-Nuclear Ribonucleoprotein D/immunology
- Interferon-gamma/immunology
- Interferon-gamma/pharmacology
- MAP Kinase Kinase 3/immunology
- MAP Kinase Kinase Kinases
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Knockout
- Mitogen-Activated Protein Kinase Kinases/immunology
- Myeloid Differentiation Factor 88
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptors, Interferon/immunology
- Receptors, Interferon/metabolism
- Recombinant Proteins
- Signal Transduction
- Tumor Necrosis Factor-alpha/biosynthesis
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
- p38 Mitogen-Activated Protein Kinases/immunology
- p38 Mitogen-Activated Protein Kinases/metabolism
- Mitogen-Activated Protein Kinase Kinase Kinase 11
Collapse
Affiliation(s)
- Dongxu Sun
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York 10021, USA
| | | |
Collapse
|
26
|
Novotny M, Datta S, Biswas R, Hamilton T. Functionally independent AU-rich sequence motifs regulate KC (CXCL1) mRNA. J Biol Chem 2005; 280:30166-74. [PMID: 15994316 DOI: 10.1074/jbc.m502280200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Certain pro-inflammatory chemokine mRNAs containing adenine/uridine-rich sequence elements (AREs) in their 3' untranslated regions (3'-UTRs) are known to exhibit constitutive instability and sensitivity to proinflammatory stimuli resulting in the stabilization of the message. Using tetR-regulated transcription we now show that the 3'-UTR of the mouse CXCL1 (KC) mRNA contains at least two ARE motifs that are structurally and functionally distinct. A fragment of 77 nucleotides containing 4 clustered AUUUA pentamers located at the 5'-end of the KC 3'-UTR is only modestly unstable yet promotes markedly enhanced, post-transcriptional protein production in response to either interleukin-1alpha (IL-1alpha) or lipopolysaccharide (LPS), suggesting translational regulation. In contrast, a fragment containing 3 isolated AUUUA pentamers corresponding to the residual 3' 400 nucleotides of the KC 3'-UTR confers both instability and is stabilized in response to IL-1alpha. Although the clustered AUUUA pentamers in the upstream region are required for stimulus sensitivity, mutation of all three pentamers in the downstream region has little or no effect on either instability or stimulus sensitivity. The upstream region is comparably stabilized in response to either IL-1alpha or LPS, whereas the AUUUA-independent downstream determinant is differentially more sensitive to IL-1alpha. Finally, using UV-induced RNA cross-linking, these functionally independent sequences exhibit different patterns of interaction with RNA-binding proteins. Collectively, these findings document the presence of multiple independent determinants of KC mRNA function and demonstrate that these operate via distinct mechanisms.
Collapse
Affiliation(s)
- Michael Novotny
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | |
Collapse
|
27
|
Hirotani T, Yamamoto M, Kumagai Y, Uematsu S, Kawase I, Takeuchi O, Akira S. Regulation of lipopolysaccharide-inducible genes by MyD88 and Toll/IL-1 domain containing adaptor inducing IFN-β. Biochem Biophys Res Commun 2005; 328:383-92. [PMID: 15694359 DOI: 10.1016/j.bbrc.2004.12.184] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2004] [Indexed: 02/03/2023]
Abstract
Macrophages recognize lipopolysaccharide (LPS) by Toll-like receptor 4 and activate inflammatory responses by inducing expression of various genes. TLR4 activates intracellular signaling pathways via TIR domain containing adaptor molecules, MyD88, and Toll/IL-1 domain containing adaptor inducing IFN-beta (TRIF). Although macrophages lacking MyD88 or TRIF showed impaired cytokine production, activation of intracellular signaling molecules still occurred in response to LPS in these cells. In the present study, we implemented cDNA microarrays to investigate the contribution of MyD88 and TRIF in gene expression induced by LPS stimulation. Whereas wild-type macrophages induced 148 genes in response to LPS, macrophages lacking both MyD88 and TRIF did not upregulate any genes in response to LPS. Surprisingly, 80 LPS-inducible genes were redundantly regulated by either MyD88 or TRIF. In contrast, proinflammatory cytokines and chemokines were critically regulated by MyD88 or TRIF alone. Genes critically regulated by MyD88 alone tend to be induced quickly after LPS stimulation and regulated by mRNA stability as well as transcription. Genes known to be induced by type I interferons were simply dependent on TRIF for their expression. Taken together, MyD88 and TRIF play both redundant and distinct roles in LPS-induced gene expression.
Collapse
Affiliation(s)
- Tomonori Hirotani
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|