1
|
Huang S, Yang J, Xie T, Jiang Y, Hong Y, Liu X, He X, Buratto D, Zhang D, Zhou R, Liang T, Bai X. Inhibition of DEF-p65 Interactions as a Potential Avenue to Suppress Tumor Growth in Pancreatic Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401845. [PMID: 38757623 PMCID: PMC11267266 DOI: 10.1002/advs.202401845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/28/2024] [Indexed: 05/18/2024]
Abstract
The limited success of current targeted therapies for pancreatic cancer underscores an urgent demand for novel treatment modalities. The challenge in mitigating this malignancy can be attributed to the digestive organ expansion factor (DEF), a pivotal yet underexplored factor in pancreatic tumorigenesis. The study uses a blend of in vitro and in vivo approaches, complemented by the theoretical analyses, to propose DEF as a promising anti-tumor target. Analysis of clinical samples reveals that high expression of DEF is correlated with diminished survival in pancreatic cancer patients. Crucially, the depletion of DEF significantly impedes tumor growth. The study further discovers that DEF binds to p65, shielding it from degradation mediated by the ubiquitin-proteasome pathway in cancer cells. Based on these findings and computational approaches, the study formulates a DEF-mimicking peptide, peptide-031, designed to disrupt the DEF-p65 interaction. The effectiveness of peptide-031 in inhibiting tumor proliferation has been demonstrated both in vitro and in vivo. This study unveils the oncogenic role of DEF while highlighting its prognostic value and therapeutic potential in pancreatic cancer. In addition, peptide-031 is a promising therapeutic agent with potent anti-tumor effects.
Collapse
Affiliation(s)
- Sicong Huang
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Key Laboratory of Pancreatic Disease of Zhejiang ProvinceHangzhou310000China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang ProvinceHangzhou310000China
| | - Jiaqi Yang
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Key Laboratory of Pancreatic Disease of Zhejiang ProvinceHangzhou310000China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang ProvinceHangzhou310000China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic DiseasesHangzhou310000China
| | - Teng Xie
- Institute of Quantitative Biology, College of Life SciencesZhejiang UniversityHangzhou310000China
- Shanghai Institute for Advanced StudyZhejiang UniversityShanghai200000China
| | - Yangwei Jiang
- Institute of Quantitative Biology, College of Life SciencesZhejiang UniversityHangzhou310000China
| | - Yifan Hong
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Key Laboratory of Pancreatic Disease of Zhejiang ProvinceHangzhou310000China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang ProvinceHangzhou310000China
| | - Xinyuan Liu
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Key Laboratory of Pancreatic Disease of Zhejiang ProvinceHangzhou310000China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang ProvinceHangzhou310000China
| | - Xuyan He
- Life Sciences InstituteZhejiang UniversityHangzhou310000China
| | - Damiano Buratto
- Institute of Quantitative Biology, College of Life SciencesZhejiang UniversityHangzhou310000China
- Shanghai Institute for Advanced StudyZhejiang UniversityShanghai200000China
| | - Dong Zhang
- Institute of Quantitative Biology, College of Life SciencesZhejiang UniversityHangzhou310000China
- Shanghai Institute for Advanced StudyZhejiang UniversityShanghai200000China
| | - Ruhong Zhou
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Institute of Quantitative Biology, College of Life SciencesZhejiang UniversityHangzhou310000China
- Shanghai Institute for Advanced StudyZhejiang UniversityShanghai200000China
- Department of ChemistryColumbia UniversityNew York10027USA
- Cancer CenterZhejiang UniversityHangzhou310000China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Key Laboratory of Pancreatic Disease of Zhejiang ProvinceHangzhou310000China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang ProvinceHangzhou310000China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic DiseasesHangzhou310000China
- Cancer CenterZhejiang UniversityHangzhou310000China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Key Laboratory of Pancreatic Disease of Zhejiang ProvinceHangzhou310000China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang ProvinceHangzhou310000China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic DiseasesHangzhou310000China
- Cancer CenterZhejiang UniversityHangzhou310000China
| |
Collapse
|
2
|
Feoktistov AV, Georgieva SG, Soshnikova NV. Chromatin Remodeling Complex PBAF Activates and Represses Inflammatory Genes. DOKL BIOCHEM BIOPHYS 2023; 513:332-336. [PMID: 38066320 DOI: 10.1134/s1607672923700539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 01/26/2024]
Abstract
The PBAF chromatin remodeling complex regulates chromatin state and gene transcription in higher eukaryotes. In this work, we studied the role of PBAF in the regulation of NF-κB-and JAK/STAT-dependent activation of inflammatory genes. We performed knockdown of specific module subunit BAF200, which resulted in destruction of the entire PBAF specific module and changed the level of the genes transcription of both pathways. PBAF can be both an activator and a repressor of inflammatory genes. Thus, PBAF is an important regulator of inflammatory gene expression.
Collapse
Affiliation(s)
- A V Feoktistov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | - S G Georgieva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - N V Soshnikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
3
|
Tan SYX, Zhang J, Tee WW. Epigenetic Regulation of Inflammatory Signaling and Inflammation-Induced Cancer. Front Cell Dev Biol 2022; 10:931493. [PMID: 35757000 PMCID: PMC9213816 DOI: 10.3389/fcell.2022.931493] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/23/2022] [Indexed: 01/10/2023] Open
Abstract
Epigenetics comprise a diverse array of reversible and dynamic modifications to the cell’s genome without implicating any DNA sequence alterations. Both the external environment surrounding the organism, as well as the internal microenvironment of cells and tissues, contribute to these epigenetic processes that play critical roles in cell fate specification and organismal development. On the other hand, dysregulation of epigenetic activities can initiate and sustain carcinogenesis, which is often augmented by inflammation. Chronic inflammation, one of the major hallmarks of cancer, stems from proinflammatory cytokines that are secreted by tumor and tumor-associated cells in the tumor microenvironment. At the same time, inflammatory signaling can establish positive and negative feedback circuits with chromatin to modulate changes in the global epigenetic landscape. In this review, we provide an in-depth discussion of the interconnected crosstalk between epigenetics and inflammation, specifically how epigenetic mechanisms at different hierarchical levels of the genome control inflammatory gene transcription, which in turn enact changes within the cell’s epigenomic profile, especially in the context of inflammation-induced cancer.
Collapse
Affiliation(s)
- Shawn Ying Xuan Tan
- Chromatin Dynamics and Disease Epigenetics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Jieqiong Zhang
- Chromatin Dynamics and Disease Epigenetics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wee-Wei Tee
- Chromatin Dynamics and Disease Epigenetics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
4
|
Alizada A, Khyzha N, Wang L, Antounians L, Chen X, Khor M, Liang M, Rathnakumar K, Weirauch MT, Medina-Rivera A, Fish JE, Wilson MD. Conserved regulatory logic at accessible and inaccessible chromatin during the acute inflammatory response in mammals. Nat Commun 2021; 12:567. [PMID: 33495464 PMCID: PMC7835376 DOI: 10.1038/s41467-020-20765-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 12/18/2020] [Indexed: 12/18/2022] Open
Abstract
The regulatory elements controlling gene expression during acute inflammation are not fully elucidated. Here we report the identification of a set of NF-κB-bound elements and common chromatin landscapes underlying the acute inflammatory response across cell-types and mammalian species. Using primary vascular endothelial cells (human/mouse/bovine) treated with the pro-inflammatory cytokine, Tumor Necrosis Factor-α, we identify extensive (~30%) conserved orthologous binding of NF-κB to accessible, as well as nucleosome-occluded chromatin. Regions with the highest NF-κB occupancy pre-stimulation show dramatic increases in NF-κB binding and chromatin accessibility post-stimulation. These 'pre-bound' regions are typically conserved (~56%), contain multiple NF-κB motifs, are utilized by diverse cell types, and overlap rare non-coding mutations and common genetic variation associated with both inflammatory and cardiovascular phenotypes. Genetic ablation of conserved, 'pre-bound' NF-κB regions within the super-enhancer associated with the chemokine-encoding CCL2 gene and elsewhere supports the functional relevance of these elements.
Collapse
Affiliation(s)
- Azad Alizada
- Hospital for Sick Children, Genetics and Genome Biology, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Nadiya Khyzha
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, Canada
| | - Liangxi Wang
- Hospital for Sick Children, Genetics and Genome Biology, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Lina Antounians
- Hospital for Sick Children, Genetics and Genome Biology, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Xiaoting Chen
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Melvin Khor
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, Canada
| | - Minggao Liang
- Hospital for Sick Children, Genetics and Genome Biology, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Kumaragurubaran Rathnakumar
- Hospital for Sick Children, Genetics and Genome Biology, Toronto, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, Canada
| | - Matthew T Weirauch
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Alejandra Medina-Rivera
- Hospital for Sick Children, Genetics and Genome Biology, Toronto, Canada
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| | - Jason E Fish
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
- University Health Network, Toronto General Hospital Research Institute, Toronto, Canada.
- University Health Network, Peter Munk Cardiac Centre, Toronto, Canada.
| | - Michael D Wilson
- Hospital for Sick Children, Genetics and Genome Biology, Toronto, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, Canada.
| |
Collapse
|
5
|
Lobb IT, Morin P, Martin K, Thoms HC, Wills JC, Lleshi X, Olsen KCF, Duncan RR, Stark LA. A Role for the Autophagic Receptor, SQSTM1/p62, in Trafficking NF-κB/RelA to Nucleolar Aggresomes. Mol Cancer Res 2020; 19:274-287. [PMID: 33097627 DOI: 10.1158/1541-7786.mcr-20-0336] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 09/18/2020] [Accepted: 10/16/2020] [Indexed: 11/16/2022]
Abstract
Elevated NF-κB activity is a contributory factor in many hematologic and solid malignancies. Nucleolar sequestration of NF-κB/RelA represses this elevated activity and mediates apoptosis of cancer cells. Here, we set out to understand the mechanisms that control the nuclear/nucleolar distribution of RelA and other regulatory proteins, so that agents can be developed that specifically target these proteins to the organelle. We demonstrate that RelA accumulates in intranucleolar aggresomes in response to specific stresses. We also demonstrate that the autophagy receptor, SQSTM1/p62, accumulates alongside RelA in these nucleolar aggresomes. This accumulation is not a consequence of inhibited autophagy. Indeed, our data suggest nucleolar and autophagosomal accumulation of p62 are in active competition. We identify a conserved motif at the N-terminus of p62 that is essential for nucleoplasmic-to-nucleolar transport of the protein. Furthermore, using a dominant-negative mutant deleted for this nucleolar localization signal (NoLS), we demonstrate a role for p62 in trafficking RelA and other aggresome-related proteins to nucleoli, to induce apoptosis. Together, these data identify a novel role for p62 in trafficking nuclear proteins to nucleolar aggresomes under conditions of cell stress, thus maintaining cellular homeostasis. They also provide invaluable information on the mechanisms that regulate the nuclear/nucleolar distribution of RelA that could be exploited for therapeutic purpose. IMPLICATIONS: The data open up avenues for the development of a unique class of therapeutic agents that act by targeting RelA and other aberrantly active proteins to nucleoli, thus killing cancer cells.
Collapse
Affiliation(s)
- Ian T Lobb
- Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Pierre Morin
- Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Kirsty Martin
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot Watt University, Edinburgh, Scotland
| | - Hazel C Thoms
- Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | | | - Xhordi Lleshi
- Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Karl C F Olsen
- Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Rory R Duncan
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot Watt University, Edinburgh, Scotland
| | - Lesley A Stark
- Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland.
| |
Collapse
|
6
|
Zhao Z, Sun W, Guo Z, Zhang J, Yu H, Liu B. Mechanisms of lncRNA/microRNA interactions in angiogenesis. Life Sci 2020; 254:116900. [DOI: 10.1016/j.lfs.2019.116900] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 09/09/2019] [Accepted: 09/20/2019] [Indexed: 12/12/2022]
|
7
|
Ariel O, Gendron D, Dudemaine PL, Gévry N, Ibeagha-Awemu EM, Bissonnette N. Transcriptome Profiling of Bovine Macrophages Infected by Mycobacterium avium spp. paratuberculosis Depicts Foam Cell and Innate Immune Tolerance Phenotypes. Front Immunol 2020; 10:2874. [PMID: 31969876 PMCID: PMC6960179 DOI: 10.3389/fimmu.2019.02874] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/22/2019] [Indexed: 12/18/2022] Open
Abstract
Mycobacterium avium spp. paratuberculosis (MAP) is the causative agent of Johne's disease (JD), also known as paratuberculosis, in ruminants. The mechanisms of JD pathogenesis are not fully understood, but it is known that MAP subverts the host immune system by using macrophages as its primary reservoir. MAP infection in macrophages is often studied in healthy cows or experimentally infected calves, but reports on macrophages from naturally infected cows are lacking. In our study, primary monocyte-derived macrophages (MDMs) from cows diagnosed as positive (+) or negative (–) for JD were challenged in vitro with live MAP. Analysis using next-generation RNA sequencing revealed that macrophages from JD(+) cows did not present a definite pattern of response to MAP infection. Interestingly, a considerable number of genes, up to 1436, were differentially expressed in JD(–) macrophages. The signatures of the infection time course of 1, 4, 8, and 24 h revealed differential expression of ARG2, COL1A1, CCL2, CSF3, IL1A, IL6, IL10, PTGS2, PTX3, SOCS3, TNF, and TNFAIP6 among other genes, with major effects on host signaling pathways. While several immune pathways were affected by MAP, other pathways related to hepatic fibrosis/hepatic stellate cell activation, lipid homeostasis, such as LXR/RXR (liver X receptor/retinoid X receptor) activation pathways, and autoimmune diseases (rheumatoid arthritis or atherosclerosis) also responded to the presence of live MAP. Comparison of the profiles of the unchallenged MDMs from JD(+) vs. JD(–) cows showed that 868 genes were differentially expressed, suggesting that these genes were already affected before monocytes differentiated into macrophages. The downregulated genes predominantly modified the general cell metabolism by downregulating amino acid synthesis and affecting cholesterol biosynthesis and other energy production pathways while introducing a pro-fibrotic pattern associated with foam cells. The upregulated genes indicated that lipid homeostasis was already supporting fat storage in uninfected JD(+) MDMs. For JD(+) MDMs, differential gene expression expounds long-term mechanisms established during disease progression of paratuberculosis. Therefore, MAP could further promote disease persistence by influencing long-term macrophage behavior by using both tolerance and fat-storage states. This report contributes to a better understanding of MAP's controls over the immune cell response and mechanisms of MAP survival.
Collapse
Affiliation(s)
- Olivier Ariel
- Sherbrooke Research and Development Center, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada.,Department of Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Daniel Gendron
- Sherbrooke Research and Development Center, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada
| | - Pier-Luc Dudemaine
- Sherbrooke Research and Development Center, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada.,Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Nicolas Gévry
- Department of Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Eveline M Ibeagha-Awemu
- Sherbrooke Research and Development Center, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada
| | - Nathalie Bissonnette
- Sherbrooke Research and Development Center, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada.,Department of Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
8
|
Gougelet A. Epigenetic modulation of immunity: towards new therapeutic avenues in hepatocellular carcinoma? Gut 2019; 68:1727-1728. [PMID: 31243054 DOI: 10.1136/gutjnl-2019-319084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/03/2019] [Accepted: 06/06/2019] [Indexed: 01/10/2023]
|
9
|
Ren J, Li L, Wang Y, Zhai J, Chen G, Hu K. Gambogic acid induces heme oxygenase-1 through Nrf2 signaling pathway and inhibits NF-κB and MAPK activation to reduce inflammation in LPS-activated RAW264.7 cells. Biomed Pharmacother 2019; 109:555-562. [DOI: 10.1016/j.biopha.2018.10.112] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 12/20/2022] Open
|
10
|
Pulugulla SH, Packard TA, Galloway NLK, Grimmett ZW, Doitsh G, Adamik J, Galson DL, Greene WC, Auron PE. Distinct mechanisms regulate IL1B gene transcription in lymphoid CD4 T cells and monocytes. Cytokine 2018; 111:373-381. [PMID: 30300855 DOI: 10.1016/j.cyto.2018.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/26/2018] [Accepted: 10/01/2018] [Indexed: 02/07/2023]
Abstract
Interleukin 1β is a pro-inflammatory cytokine important for both normal immune responses and chronic inflammatory diseases. The regulation of the 31 kDa proIL-1β precursor coded by the IL1B gene has been extensively studied in myeloid cells, but not in lymphoid-derived CD4 T cells. Surprisingly, we found that some CD4 T cell subsets express higher levels of proIL-1β than unstimulated monocytes, despite relatively low IL1B mRNA levels. We observed a significant increase in IL1B transcription and translation in CD4 T cells upon ex vivo CD3/CD28 activation, and a similar elevation in the CCR5+ effector memory population compared to CCR5- T cells in vivo. The rapid and vigorous increase in IL1B gene transcription for stimulated monocytes has previously been associated with the presence of Spi-1/PU.1 (Spi1), a myeloid-lineage transcription factor, pre-bound to the promoter. In the case of CD4 T cells, this increase occurred despite the lack of detectable Spi1 at the IL1B promoter. Additionally, we found altered epigenetic regulation of the IL1B locus in CD3/CD28-activated CD4 T cells. Unlike monocytes, activated CD4 T cells possess bivalent H3K4me3+/H3K27me3+ nucleosome marks at the IL1B promoter, reflecting low transcriptional activity. These results support a model in which the IL1B gene in CD4 T cells is transcribed from a low-activity bivalent promoter independent of Spi1. Accumulated cytoplasmic proIL-1β may ultimately be cleaved to mature 17 kDa bioactive IL-1β, regulating T cell polarization and pathogenic chronic inflammation.
Collapse
Affiliation(s)
- Sree H Pulugulla
- Duquesne University, Department of Biological Sciences, Pittsburgh, PA 15282, United States
| | - Thomas A Packard
- Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, United States
| | - Nicole L K Galloway
- Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, United States
| | - Zachary W Grimmett
- Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, United States
| | - Gilad Doitsh
- Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, United States; Department of Medicine, University of California, San Francisco, CA 94143, United States
| | - Juraj Adamik
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, United States
| | - Deborah L Galson
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, United States; University of Pittsburgh Hillman Cancer Center & McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15213, United States
| | - Warner C Greene
- Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, United States; Department of Medicine, University of California, San Francisco, CA 94143, United States; Department of Microbiology and Immunology, University of California, San Francisco, CA 94143, United States
| | - Philip E Auron
- Duquesne University, Department of Biological Sciences, Pittsburgh, PA 15282, United States; Department of Microbiology & Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, United States.
| |
Collapse
|
11
|
Proteasome inhibition induces IKK-dependent interleukin-8 expression in triple negative breast cancer cells: Opportunity for combination therapy. PLoS One 2018; 13:e0201858. [PMID: 30089134 PMCID: PMC6082561 DOI: 10.1371/journal.pone.0201858] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/23/2018] [Indexed: 12/14/2022] Open
Abstract
Triple negative breast cancer (TNBC) cells express increased levels of the pro-inflammatory and pro-angiogenic chemokine interleukin-8 (IL-8, CXCL8), which promotes their proliferation and migration. Because TNBC patients are unresponsive to current targeted therapies, new therapeutic strategies are urgently needed. While proteasome inhibition by bortezomib (BZ) or carfilzomib (CZ) has been effective in treating hematological malignancies, it has been less effective in solid tumors, including TNBC, but the mechanisms are incompletely understood. Here we report that proteasome inhibition significantly increases expression of IL-8, and its receptors CXCR1 and CXCR2, in TNBC cells. Suppression or neutralization of the BZ-induced IL-8 potentiates the BZ cytotoxic and anti-proliferative effect in TNBC cells. The IL-8 expression induced by proteasome inhibition in TNBC cells is mediated by IκB kinase (IKK), increased nuclear accumulation of p65 NFκB, and by IKK-dependent p65 recruitment to IL-8 promoter. Importantly, inhibition of IKK activity significantly decreases proliferation, migration, and invasion of BZ-treated TNBC cells. These data provide the first evidence demonstrating that proteasome inhibition increases the IL-8 signaling in TNBC cells, and suggesting that IKK inhibitors may increase effectiveness of proteasome inhibitors in treating TNBC.
Collapse
|
12
|
Zhang H, Jia R, Wang F, Qiu G, Qiao P, Xu X, Wu D. Catalpol protects mice against Lipopolysaccharide/D-galactosamine-induced acute liver injury through inhibiting inflammatory and oxidative response. Oncotarget 2017; 9:3887-3894. [PMID: 29423091 PMCID: PMC5790508 DOI: 10.18632/oncotarget.23242] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/01/2017] [Indexed: 01/10/2023] Open
Abstract
The purpose of this study was to investigate the protective effect of catalpol on Lipopolysaccharide (LPS)/D-galactosamine (D-gal)-induced acute liver injury in mice. The mouse model was established by injection of LPS and D-gal. Catalpol (2.5, 5, 10 mg/kg) were pretreated intraperitoneally 1 h before LPS and D-gal. The survival rate, AST, ALT, MDA, MPO activity, hepatic tissue histology, TNF-α level, and NF-κB activation were assayed. The results revealed that catalpol dose-dependently elevated the survival rate. Furthermore, catalpol reduced the activities of AST, ALT, MDA, and MPO. The production of TNF-α was also inhibited by treatment of catalpol. In addition, catalpol inhibited LPS/D-gal-induced NF-κB activation. The expression of Nrf2 and HO-1 were up-regulated by treatment of catalpol. These results indicated that pretreatment with catalpol could attenuate LPS/D-gal-induced acute liver injury in mice and the underlying mechanism may due to the inhibition of NF-κB signaling pathway and the activation of Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Haogang Zhang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Ruichun Jia
- Department of Blood Transfusion, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Fujing Wang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Gongcai Qiu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Pengfei Qiao
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Xunzheng Xu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Dequan Wu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| |
Collapse
|
13
|
Vancurova I, Uddin MM, Zou Y, Vancura A. Combination Therapies Targeting HDAC and IKK in Solid Tumors. Trends Pharmacol Sci 2017; 39:295-306. [PMID: 29233541 DOI: 10.1016/j.tips.2017.11.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/21/2017] [Accepted: 11/27/2017] [Indexed: 12/18/2022]
Abstract
The rationale for developing histone deacetylase (HDAC) inhibitors (HDACi) as anticancer agents was based on their ability to induce apoptosis and cell cycle arrest in cancer cells. However, while HDACi have been remarkably effective in the treatment of hematological malignancies, clinical studies with HDACi as single agents in solid cancers have been disappointing. Recent studies have shown that, in addition to inducing apoptosis in cancer cells, class I HDACi induce IκB kinase (IKK)-dependent expression of proinflammatory chemokines, such as interleukin-8 (IL8; CXCL8), resulting in the increased proliferation of tumor cells, and limiting the effectiveness of HDACi in solid tumors. Here, we discuss the mechanisms responsible for HDACi-induced CXCL8 expression, and opportunities for combination therapies targeting HDACs and IKK in solid tumors.
Collapse
Affiliation(s)
- Ivana Vancurova
- Department of Biological Sciences, St John's University, New York, NY 11439, USA.
| | - Mohammad M Uddin
- Department of Biological Sciences, St John's University, New York, NY 11439, USA
| | - Yue Zou
- Department of Biological Sciences, St John's University, New York, NY 11439, USA
| | - Ales Vancura
- Department of Biological Sciences, St John's University, New York, NY 11439, USA
| |
Collapse
|
14
|
Andy SN, Chan CK, Kadir HA. Deoxyelephantopin from Elephantopus scaber modulates neuroinflammatory response through MAPKs and PI3K/Akt-dependent NF-κB signaling pathways in LPS-stimulated BV-2 microglial cells. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
15
|
Rahnamoun H, Lu H, Duttke SH, Benner C, Glass CK, Lauberth SM. Mutant p53 shapes the enhancer landscape of cancer cells in response to chronic immune signaling. Nat Commun 2017; 8:754. [PMID: 28963538 PMCID: PMC5622043 DOI: 10.1038/s41467-017-01117-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 08/18/2017] [Indexed: 01/15/2023] Open
Abstract
Inflammation influences cancer development, progression, and the efficacy of cancer treatments, yet the mechanisms by which immune signaling drives alterations in the cancer cell transcriptome remain unclear. Using ChIP-seq, RNA-seq, and GRO-seq, here we demonstrate a global overlap in the binding of tumor-promoting p53 mutants and the master proinflammatory regulator NFκB that drives alterations in enhancer and gene activation in response to chronic TNF-α signaling. We show that p53 mutants interact directly with NFκB and that both factors impact the other's binding at diverse sets of active enhancers. In turn, the simultaneous and cooperative binding of these factors is required to regulate RNAPII recruitment, the synthesis of enhancer RNAs, and the activation of tumor-promoting genes. Collectively, these findings establish a mechanism by which chronic TNF-α signaling orchestrates a functional interplay between mutant p53 and NFκB that underlies altered patterns of cancer-promoting gene expression.Inflammation is known to affect cancer development, yet the mechanisms by which immune signaling drives transformation remain unclear. Here, the authors provide evidence that chronic TNF-α signaling promotes the enhancer binding and transcriptional interplay between mutant p53 and NFκB.
Collapse
Affiliation(s)
- Homa Rahnamoun
- Section of Molecular Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Hanbin Lu
- Section of Molecular Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Sascha H Duttke
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0651, USA
| | - Christopher Benner
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0651, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0651, USA
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0651, USA
| | - Shannon M Lauberth
- Section of Molecular Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
16
|
Asadipour M, Hassan-Zadeh V, Aryaeian N, Shahram F, Mahmoudi M. Histone variants expression in peripheral blood mononuclear cells of patients with rheumatoid arthritis. Int J Rheum Dis 2017; 21:1831-1837. [DOI: 10.1111/1756-185x.13126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Maryam Asadipour
- Department of Cell and Molecular Biology; Kish International Campus; University of Tehran; Kish Iran
| | - Vahideh Hassan-Zadeh
- Department of Cell and Molecular Biology; Faculty of Biology; College of Science; University of Tehran; Tehran Iran
| | - Naheed Aryaeian
- Department of Nutrition; School of Public Health; Iran University of Medical Sciences; Tehran Iran
| | - Farhad Shahram
- Rheumatology Research Center (RRC); Tehran University of Medical Sciences; Tehran Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center (RRC); Tehran University of Medical Sciences; Tehran Iran
| |
Collapse
|
17
|
Song XM, Yu Q, Dong X, Yang HO, Zeng KW, Li J, Tu PF. Aldose reductase inhibitors attenuate β-amyloid-induced TNF-α production in microlgia via ROS-PKC-mediated NF-κB and MAPK pathways. Int Immunopharmacol 2017. [PMID: 28623716 DOI: 10.1016/j.intimp.2017.06.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Microglia-mediated neuroinflammation is a key risk factor to the development of Alzheimer' disease (AD). Aldose reductase (AR) has been found to be widely involved in inflammation-related diseases; however, whether aldose reductase inhibitors (ARIs) could be used to treat neuroinflammation is rarely reported. This study aims to evaluate the anti-neuroinflammatory effects of two major ARIs of Sorbinil (Sor) and Zopolrestat (Zol) in β-amyloid protein (Aβ)-induced microglia (BV-2). We find that Sor and Zol significantly inhibit TNF-α, IL-1β, IL-6 production from microglia in response to Aβ stimulation. Mechanism study showed that Sor and Zol decreased the production of intracellular ROS which resulted in an effective inhibition on the phosphorylation of several protein kinase C (PKC) isoforms including PKCα/β, δ, ζ/λ and mu. Moreover, Sor and Zol inactivated PCK-associated IKKβ-IκB-NF-κB and mitogen-activated protein kinase (JNK, p38, ERK) inflammation pathways. In summary, our findings suggest that Sor and Zol could inhibit Aβ-induced neuroinflammation by regulating ROS/PKC-dependent NF-κB and MAPK signaling pathways, indicating that ARIs could be promising agents for treating inflammation-related neurodegenerative diseases such as AD.
Collapse
Affiliation(s)
- Xiao-Min Song
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qian Yu
- Research Studio of Integration of Traditional and Western Medicine, First Hospital, Peking University, Beijing 100034, China
| | - Xin Dong
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hyun Ok Yang
- Natural Products Research Center, Korea Institute of Science and Technology (KIST) Gangneung Institute, Kangneung 210-340, Republic of Korea
| | - Ke-Wu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Jun Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Peng-Fei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
18
|
Worzfeld T, Pogge von Strandmann E, Huber M, Adhikary T, Wagner U, Reinartz S, Müller R. The Unique Molecular and Cellular Microenvironment of Ovarian Cancer. Front Oncol 2017; 7:24. [PMID: 28275576 PMCID: PMC5319992 DOI: 10.3389/fonc.2017.00024] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/07/2017] [Indexed: 12/13/2022] Open
Abstract
The reciprocal interplay of cancer cells and host cells is an indispensable prerequisite for tumor growth and progression. Cells of both the innate and adaptive immune system, in particular tumor-associated macrophages (TAMs) and T cells, as well as cancer-associated fibroblasts enter into a malicious liaison with tumor cells to create a tumor-promoting and immunosuppressive tumor microenvironment (TME). Ovarian cancer, the most lethal of all gynecological malignancies, is characterized by a unique TME that enables specific and efficient metastatic routes, impairs immune surveillance, and mediates therapy resistance. A characteristic feature of the ovarian cancer TME is the role of resident host cells, in particular activated mesothelial cells, which line the peritoneal cavity in huge numbers, as well as adipocytes of the omentum, the preferred site of metastatic lesions. Another crucial factor is the peritoneal fluid, which enables the transcoelomic spread of tumor cells to other pelvic and peritoneal organs, and occurs at more advanced stages as a malignancy-associated effusion. This ascites is rich in tumor-promoting soluble factors, extracellular vesicles and detached cancer cells as well as large numbers of T cells, TAMs, and other host cells, which cooperate with resident host cells to support tumor progression and immune evasion. In this review, we summarize and discuss our current knowledge of the cellular and molecular interactions that govern this interplay with a focus on signaling networks formed by cytokines, lipids, and extracellular vesicles; the pathophysiologial roles of TAMs and T cells; the mechanism of transcoelomic metastasis; and the cell type selective processing of signals from the TME.
Collapse
Affiliation(s)
- Thomas Worzfeld
- Institute of Pharmacology, Biochemical-Pharmacological Center (BPC), Philipps University, Marburg, Germany; Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Elke Pogge von Strandmann
- Experimental Tumor Research, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology, Philipps University , Marburg , Germany
| | - Magdalena Huber
- Institute of Medical Microbiology and Hygiene, Biomedical Research Center, Philipps University , Marburg , Germany
| | - Till Adhikary
- Institute of Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University , Marburg , Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital of Giessen and Marburg (UKGM) , Marburg , Germany
| | - Silke Reinartz
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Philipps University , Marburg , Germany
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University , Marburg , Germany
| |
Collapse
|
19
|
Sette M, D'Addabbo P, Kelly G, Cicconi A, Micheli E, Cacchione S, Poma A, Gargioli C, Giambra V, Frezza D. Evidence for a quadruplex structure in the polymorphic hs1.2 enhancer of the immunoglobulin heavy chain 3' regulatory regions and its conservation in mammals. Biopolymers 2017; 105:768-78. [PMID: 27287611 PMCID: PMC5516150 DOI: 10.1002/bip.22891] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 06/01/2016] [Indexed: 11/09/2022]
Abstract
Regulatory regions in the genome can act through a variety of mechanisms that range from the occurrence of histone modifications to the presence of protein-binding loci for self-annealing sequences. The final result is often the induction of a conformational change of the DNA double helix, which alters the accessibility of a region to transcription factors and consequently gene expression. A ∼300 kb regulatory region on chromosome 14 at the 3' end (3'RR) of immunoglobulin (Ig) heavy-chain genes shows very peculiar features, conserved in mammals, including enhancers and transcription factor binding sites. In primates, the 3'RR is present in two copies, both having a central enhancer named hs1.2. We previously demonstrated the association between different hs1.2 alleles and Ig plasma levels in immunopathology. Here, we present the analysis of a putative G-quadruplex structure (tetraplex) consensus site embedded in a variable number tandem repeat (one to four copies) of hs1.2 that is a distinctive element among the enhancer alleles, and an investigation of its three-dimensional structure using bioinformatics and spectroscopic approaches. We suggest that both the role of the enhancer and the alternative effect of the hs1.2 alleles may be achieved through their peculiar three-dimensional-conformational rearrangement. © 2016 Wiley Periodicals, Inc. Biopolymers 105: 768-778, 2016.
Collapse
Affiliation(s)
- Marco Sette
- Department of Chemical Sciences and Technology, University of Roma "Tor Vergata,", Roma, Italy
| | - Pietro D'Addabbo
- Department of Biology, University of Bari "Aldo Moro", Bari, Italy
| | - Geoffrey Kelly
- MRC Biomedical NMR Centre, The Francis Crick Institute, Mill Hill Laboratory, London, UK
| | - Alessandro Cicconi
- Department of Biology and Biotechnology, Sapienza University, Roma, Italy.,Institute Pasteur-Fondazione Cenci-Bolognetti, Roma, Italy
| | - Emanuela Micheli
- Department of Biology and Biotechnology, Sapienza University, Roma, Italy.,Institute Pasteur-Fondazione Cenci-Bolognetti, Roma, Italy
| | - Stefano Cacchione
- Department of Biology and Biotechnology, Sapienza University, Roma, Italy.,Institute Pasteur-Fondazione Cenci-Bolognetti, Roma, Italy
| | - Anna Poma
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Cesare Gargioli
- Department of Biology, University of Roma "Tor Vergata,", Roma, Italy
| | | | - Domenico Frezza
- Department of Biology, University of Roma "Tor Vergata,", Roma, Italy
| |
Collapse
|
20
|
Gatla HR, Zou Y, Uddin MM, Singha B, Bu P, Vancura A, Vancurova I. Histone Deacetylase (HDAC) Inhibition Induces IκB Kinase (IKK)-dependent Interleukin-8/CXCL8 Expression in Ovarian Cancer Cells. J Biol Chem 2017; 292:5043-5054. [PMID: 28167529 DOI: 10.1074/jbc.m116.771014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 01/20/2017] [Indexed: 12/18/2022] Open
Abstract
Overexpression of the pro-angiogenic chemokine IL-8 (CXCL8) is associated with a poor prognosis in several solid tumors, including epithelial ovarian cancer (EOC). Even though histone deacetylase (HDAC) inhibition has shown remarkable antitumor activity in hematological malignancies, it has been less effective in solid tumors, including EOC. Here we report results that may explain the decreased efficiency of HDAC inhibition in EOC, based on our data demonstrating that HDAC inhibition specifically induces expression of IL-8/CXCL8 in SKOV3, CAOV3, and OVCAR3 cells. Suppression or neutralization of vorinostat-induced IL-8/CXCL8 potentiates the vorinostat inhibitory effect on cell viability and proliferation. The IL-8/CXCL8 expression induced by vorinostat in EOC cells is dependent on IκB kinase (IKK) activity and associated with a gene-specific recruitment of IKKβ and IKK-dependent recruitment of p65 NFκB to the IL-8/CXCL8 promoter. In addition, HDAC inhibition induces acetylation of p65 and histone H3 and their IL-8/CXCL8 promoter occupancy. In vivo results demonstrate that combining vorinostat and the IKK inhibitor Bay 117085 significantly reduces tumor growth in nude mice compared with control untreated mice or either drug alone. Mice in the combination group had the lowest IL-8/CXCL8 tumor levels and the lowest tumor expression of the murine neutrophil [7/4] antigen, indicating reduced neutrophil infiltration. Together, our results demonstrate that HDAC inhibition specifically induces IL-8/CXCL8 expression in EOC cells and that the mechanism involves IKK, suggesting that using IKK inhibitors may increase the effectiveness of HDAC inhibitors when treating ovarian cancer and other solid tumors characterized by increased IL-8/CXCL8 expression.
Collapse
Affiliation(s)
- Himavanth R Gatla
- From the Department of Biological Sciences, St. John's University, New York, New York 11439 and
| | - Yue Zou
- From the Department of Biological Sciences, St. John's University, New York, New York 11439 and
| | - Mohammad M Uddin
- From the Department of Biological Sciences, St. John's University, New York, New York 11439 and
| | | | - Pengli Bu
- From the Department of Biological Sciences, St. John's University, New York, New York 11439 and
| | - Ales Vancura
- From the Department of Biological Sciences, St. John's University, New York, New York 11439 and
| | - Ivana Vancurova
- From the Department of Biological Sciences, St. John's University, New York, New York 11439 and
| |
Collapse
|
21
|
Günther J, Petzl W, Zerbe H, Schuberth HJ, Seyfert HM. TLR ligands, but not modulators of histone modifiers, can induce the complex immune response pattern of endotoxin tolerance in mammary epithelial cells. Innate Immun 2016; 23:155-164. [PMID: 27913794 PMCID: PMC5410871 DOI: 10.1177/1753425916681076] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Excessive stimulation of the TLR4 axis through LPS reduces the expression of some
cytokine genes in immune cells, while stimulating the expression of immune
defense genes during a subsequent bacterial infection. This endotoxin tolerance
(ET) is mediated via epigenetic mechanisms. Priming the udder of cows with LPS
was shown to induce ET in mammary epithelial cells (MEC), thereby protecting the
udder against reinfection for some time. Seeking alternatives to LPS priming we
tried to elicit ET by priming MEC with either lipopeptide (Pam2CSK4) via the
TLR2/6 axis or inhibitors of histone-modifying enzymes. Pre-incubation of MEC
with Pam2CSK4 enhanced baseline and induced expression of bactericidal
(β-defensin; SLPI) and membrane protecting factors
(SAA3, TGM3), while reducing the
expression of cytokine- and chemokine-encoding genes (TNF,
IL1β) after a subsequent pathogen challenge, the latter,
however, not as efficiently as after LPS priming. Pre-treating MEC with various
inhibitors of histone H3 modifiers (for demethylation, acetylation or
deacetylation) all failed to induce any of the protective factors and only
resulted in some dampening of cytokine gene expression after the re-challenge.
Hence, triggering immune functions via the TLR axis, but not through those
histone modifiers, induced the beneficial phenomenon of ET in MEC.
Collapse
Affiliation(s)
- Juliane Günther
- 1 Leibniz Institute for Farm Animal Biology, Institute for Genome Biology, Dummerstorf, Germany
| | - Wolfram Petzl
- 2 Clinic for Ruminants with Ambulance and Herd Health Services, Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Holm Zerbe
- 2 Clinic for Ruminants with Ambulance and Herd Health Services, Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | | | - Hans-Martin Seyfert
- 1 Leibniz Institute for Farm Animal Biology, Institute for Genome Biology, Dummerstorf, Germany
| |
Collapse
|
22
|
Sasaki-Hamada S, Hoshi M, Niwa Y, Ueda Y, Kokaji A, Kamisuki S, Kuramochi K, Sugawara F, Oka JI. Neoechinulin A induced memory improvements and antidepressant-like effects in mice. Prog Neuropsychopharmacol Biol Psychiatry 2016; 71:155-61. [PMID: 27495355 DOI: 10.1016/j.pnpbp.2016.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 07/20/2016] [Accepted: 08/01/2016] [Indexed: 12/13/2022]
Abstract
Neoechinulin A is an isoprenyl indole alkaloid that exhibits scavenging, neurotrophic factor-like, and anti-apoptotic activities. However, the effectiveness of neoechinulin A in animal models of disease has not yet been explored. In the present study, we investigated the effects of neoechinulin A on memory impairment in lipopolysaccharide (LPS)-treated mice and its antidepressant-like effects in mice. In the Y-maze test, the intracerebroventicular (i.c.v.) administration of LPS (10μg/mouse) significantly decreased spontaneous alternation behavior, which was prevented by the prior administration of neoechinulin A (300ng/mouse, i.c.v.). None of the treatments altered the locomotor activity of mice. Moreover, the administration of neoechinulin A decreased the immobility time in the forced-swim test or tail suspension test, which was prevented by the prior administration of WAY100635 (an antagonist of 5-HT1A receptors) and parachlorophenylalanine (an inhibitor of tryptophan hydroxylase). These results suggest that neoechinulin A improves memory functions in LPS-treated mice, and also exerts antidepressant-like effects through changes in the 5-HT system.
Collapse
Affiliation(s)
- Sachie Sasaki-Hamada
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Maho Hoshi
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Yuki Niwa
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Yudai Ueda
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Aya Kokaji
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Shinji Kamisuki
- Department of Applied Biological Science, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Kouji Kuramochi
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Sakyo-ku, Kyoto 606-8522, Japan
| | - Fumio Sugawara
- Department of Applied Biological Science, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Jun-Ichiro Oka
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan.
| |
Collapse
|
23
|
Down-regulation of IKKβ expression in glioma-infiltrating microglia/macrophages is associated with defective inflammatory/immune gene responses in glioblastoma. Oncotarget 2016; 6:33077-90. [PMID: 26427514 PMCID: PMC4741750 DOI: 10.18632/oncotarget.5310] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 09/15/2015] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma (GBM) is an aggressive malignancy associated with profound host immunosuppression. Microglia and macrophages infiltrating GBM acquire the pro-tumorigenic, M2 phenotype and support tumor invasion, proliferation, survival, angiogenesis and block immune responses both locally and systematically. Mechanisms responsible for immunological deficits in GBM patients are poorly understood. We analyzed immune/inflammatory gene expression in five datasets of low and high grade gliomas, and performed Gene Ontology and signaling pathway analyses to identify defective transcriptional responses. The expression of many immune/inflammatory response and TLR signaling pathway genes was reduced in high grade gliomas compared to low grade gliomas. In particular, we found the reduced expression of the IKBKB, a gene coding for IKKβ, which phosphorylates IκB proteins and represents a convergence point for most signal transduction pathways leading to NFκB activation. The reduced IKBKB expression and IKKβ levels in GBM tissues were demonstrated by qPCR, Western blotting and immunohistochemistry. The IKKβ expression was down-regulated in microglia/macrophages infiltrating glioblastoma. NFκB activation, prominent in microglia/macrophages infiltrating low grade gliomas, was reduced in microglia/macrophages in glioblastoma tissues. Down-regulation of IKBKB expression and NFκB signaling in microglia/macrophages infiltrating glioblastoma correlates with defective expression of immune/inflammatory genes and M2 polarization that may result in the global impairment of anti-tumor immune responses in glioblastoma.
Collapse
|
24
|
Morooka N, Ueguri K, Yee KKL, Yanase T, Sato T. Androgen-androgen receptor system improves chronic inflammatory conditions by suppressing monocyte chemoattractant protein-1 gene expression in adipocytes via transcriptional regulation. Biochem Biophys Res Commun 2016; 477:895-901. [PMID: 27392713 DOI: 10.1016/j.bbrc.2016.06.155] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 06/29/2016] [Indexed: 12/14/2022]
Abstract
Age-related decreases in sex hormones are closely related to chronic inflammation in obesity and metabolic diseases. Particularly, the molecular basis of androgen activity in regulating inflammation and controlling metabolism remains largely unknown. Obese adipocytes secrete monocyte chemoattractant protein-1 (MCP-1), a key chemokine that promotes the infiltration of monocytes/macrophages into adipose tissue, thereby leading to metabolic disorders. Here, we studied the role of androgen-androgen receptor (AR) action in regulating MCP-1 expression in adipose tissue. We observed the induction of Mcp-1 expression in 3T3-L1 adipocytes co-cultured with RAW264.7 macrophages. Additionally, Mcp-1 expression was upregulated by culturing in conditioned medium derived from inflammatory macrophages (M1-Mφ) containing tumor necrosis factor-alpha (TNF-α). We found that sex hormones downregulated TNF-α-induced Mcp-1 and interleukin (Il)-6 expression in 3T3-L1 adipocytes. Furthermore, luciferase-reporter analysis indicated that MCP-1 promoter activity was predominantly suppressed by dihydrotestosterone (DHT)-AR interactions through functional canonical nuclear factor-kappa B (NF-κB) sites, whereas non-canonical NF-κB site containing important flanking sequences exhibited minor contributions to DHT-AR transcriptional repression. These findings suggested that androgen-AR suppressed obesity-induced chronic inflammation in adipose tissue.
Collapse
Affiliation(s)
- Nobukatsu Morooka
- Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8512, Japan.
| | - Kei Ueguri
- Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8512, Japan
| | - Karen Kar Lye Yee
- Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8512, Japan; Human Resources Cultivation Center, Gunma University, 1-5-1 Tenjin-cho, Kiryushi, Gunma, 376-8515, Japan
| | - Toshihiko Yanase
- Department of Endocrinology and Diabetes Mellitus, School of Medicine, Fukuoka University, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Takashi Sato
- Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8512, Japan
| |
Collapse
|
25
|
Thangjam GS, Birmpas C, Barabutis N, Gregory BW, Clemens MA, Newton JR, Fulton D, Catravas JD. Hsp90 inhibition suppresses NF-κB transcriptional activation via Sirt-2 in human lung microvascular endothelial cells. Am J Physiol Lung Cell Mol Physiol 2016; 310:L964-74. [PMID: 27036868 DOI: 10.1152/ajplung.00054.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/13/2016] [Indexed: 11/22/2022] Open
Abstract
The ability of anti-heat shock protein 90 (Hsp90) drugs to attenuate NF-κB-mediated transcription is the major basis for their anti-inflammatory properties. While the molecular mechanisms underlying this effect are not clear, they appear to be distinct in human endothelial cells. We now show for the first time that type 2 sirtuin (Sirt-2) histone deacetylase binds human NF-κB target gene promoter and prevents the recruitment of NF-κB proteins and subsequent assembly of RNA polymerase II complex in human lung microvascular endothelial cells. Hsp90 inhibitors stabilize the Sirt-2/promoter interaction and impose a "transcriptional block," which is reversed by either inhibition or downregulation of Sirt-2 protein expression. Furthermore, this process is independent of NF-κB (p65) Lysine 310 deacetylation, suggesting that it is distinct from known Sirt-2-dependent mechanisms. We demonstrate that Sirt-2 is recruited to NF-κB target gene promoter via interaction with core histones. Upon inflammatory challenge, chromatin remodeling and core histone H3 displacement from the promoter region removes Sirt-2 and allows NF-κB/coactivator recruitment essential for RNA Pol II-dependent mRNA induction. This novel mechanism may have important implications in pulmonary inflammation.
Collapse
Affiliation(s)
- Gagan S Thangjam
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia
| | - Charalampos Birmpas
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia
| | - Nektarios Barabutis
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia
| | - Betsy W Gregory
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia
| | - Mary Ann Clemens
- Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, Virginia
| | - Joseph R Newton
- Department of Surgery, Eastern Virginia Medical School, Norfolk, Virginia
| | - David Fulton
- Vascular Biology Center, Augusta University, Augusta, Georgia; and
| | - John D Catravas
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia; School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, Virginia
| |
Collapse
|
26
|
Markó L, Vigolo E, Hinze C, Park JK, Roël G, Balogh A, Choi M, Wübken A, Cording J, Blasig IE, Luft FC, Scheidereit C, Schmidt-Ott KM, Schmidt-Ullrich R, Müller DN. Tubular Epithelial NF-κB Activity Regulates Ischemic AKI. J Am Soc Nephrol 2016; 27:2658-69. [PMID: 26823548 DOI: 10.1681/asn.2015070748] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 11/26/2015] [Indexed: 12/20/2022] Open
Abstract
NF-κB is a key regulator of innate and adaptive immunity and is implicated in the pathogenesis of AKI. The cell type-specific functions of NF-κB in the kidney are unknown; however, the pathway serves distinct functions in immune and tissue parenchymal cells. We analyzed tubular epithelial-specific NF-κB signaling in a mouse model of ischemia-reperfusion injury (IRI)-induced AKI. NF-κB reporter activity and nuclear localization of phosphorylated NF-κB subunit p65 analyses in mice revealed that IRI induced widespread NF-κB activation in renal tubular epithelia and in interstitial cells that peaked 2-3 days after injury. To genetically antagonize tubular epithelial NF-κB activity, we generated mice expressing the human NF-κB super-repressor IκBαΔN in renal proximal, distal, and collecting duct epithelial cells. Compared with control mice, these mice exhibited improved renal function, reduced tubular apoptosis, and attenuated neutrophil and macrophage infiltration after IRI-induced AKI. Furthermore, tubular NF-κB-dependent gene expression profiles revealed temporally distinct functional gene clusters for apoptosis, chemotaxis, and morphogenesis. Primary proximal tubular cells isolated from IκBαΔN-expressing mice and exposed to hypoxia-mimetic agent cobalt chloride exhibited less apoptosis and expressed lower levels of chemokines than cells from control mice did. Our results indicate that postischemic NF-κB activation in renal tubular epithelia aggravates tubular injury and exacerbates a maladaptive inflammatory response.
Collapse
Affiliation(s)
- Lajos Markó
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany; Max Delbrück Center for Molecular Medicine, Berlin, Germany;
| | - Emilia Vigolo
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | | | - Giulietta Roël
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - András Balogh
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany; Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Mira Choi
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Anne Wübken
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Jimmi Cording
- Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany; and
| | - Ingolf E Blasig
- Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany; and
| | - Friedrich C Luft
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany; Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | - Kai M Schmidt-Ott
- Max Delbrück Center for Molecular Medicine, Berlin, Germany; Department of Nephrology, Charité Medical Faculty, Berlin, Germany
| | | | - Dominik N Müller
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany; Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
27
|
Cieślik M, Bekiranov S. Genome-wide predictors of NF-κB recruitment and transcriptional activity. BioData Min 2015; 8:37. [PMID: 26617673 PMCID: PMC4661973 DOI: 10.1186/s13040-015-0071-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 11/18/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Inducible transcription factors (TFs) mediate transcriptional responses to environmental cues. In response to multiple inflammatory signals active NF-κB dimers enter the nucleus and trigger cell-type-, and stimulus-specific transcriptional programs. Although much is known about NF-κB inducing pathways and about locus-specific mechanisms of transcriptional control, it is poorly understood how the pre-existing chromatin landscape determines NF-κB target selection and activation. Specifically, it is not known which epigenetic marks and pre-bound TFs serve genome-wide as positive (negative) cues for active NF-κB. RESULTS We applied multivariate and combinatorial data mining techniques on a comprehensive dataset of DNA methylation, DNase I hypersensitivity, eight epigenetic marks, and 34 TFs to arrive at genome-wide patterns that predict NF-κB binding. Strikingly, we observed NF-κB recruitment to accessible and nucleosome-bound sites. Within nucleosomal DNA NF-κB binding was primed by H3K4me1 and H2A.Z, but also hyper-methylated DNA outside of promoters and CpG-islands. Many of these predictors showed combinatorial cooperativity and statistically significant interactions. Recruitment to pre-accessible sites was more frequent and influenced by chromatin-associated TFs. We observed that specific TF-combinations are greatly enriched for (or depleted of) NF-κB binding events. CONCLUSIONS We provide evidence of NF-κB binding within genomic regions that lack classical marks of activity. These pioneer binding events are relatively often associated with transcriptional regulation. Further, our predictive models indicate that specific combinations of epigenetic marks and transcription factors predetermine the NF-κB cistrome, supporting the feasibility of using statistical approaches to identify "histone codes".
Collapse
Affiliation(s)
- Marcin Cieślik
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109 USA
| | - Stefan Bekiranov
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia USA
| |
Collapse
|
28
|
Vallabhapurapu SD, Noothi SK, Pullum DA, Lawrie CH, Pallapati R, Potluri V, Kuntzen C, Khan S, Plas DR, Orlowski RZ, Chesi M, Kuehl WM, Bergsagel PL, Karin M, Vallabhapurapu S. Transcriptional repression by the HDAC4-RelB-p52 complex regulates multiple myeloma survival and growth. Nat Commun 2015; 6:8428. [PMID: 26455434 DOI: 10.1038/ncomms9428] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 08/21/2015] [Indexed: 12/12/2022] Open
Abstract
Although transcriptional activation by NF-κB is well appreciated, physiological importance of transcriptional repression by NF-κB in cancer has remained elusive. Here we show that an HDAC4-RelB-p52 complex maintains repressive chromatin around proapoptotic genes Bim and BMF and regulates multiple myeloma (MM) survival and growth. Disruption of RelB-HDAC4 complex by a HDAC4-mimetic polypeptide blocks MM growth. RelB-p52 also represses BMF translation by regulating miR-221 expression. While the NIK-dependent activation of RelB-p52 in MM has been reported, we show that regardless of the activation status of NIK and the oncogenic events that cause plasma cell malignancy, several genetically diverse MM cells including Bortezomib-resistant MM cells are addicted to RelB-p52 for survival. Importantly, RelB is constitutively phosphorylated in MM and ERK1 is a RelB kinase. Phospho-RelB remains largely nuclear and is essential for Bim repression. Thus, ERK1-dependent regulation of nuclear RelB is critical for MM survival and explains the NIK-independent role of RelB in MM.
Collapse
Affiliation(s)
- Subrahmanya D Vallabhapurapu
- The Vontz Center for Molecular Studies, Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA
| | - Sunil K Noothi
- The Vontz Center for Molecular Studies, Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA
| | - Derek A Pullum
- The Vontz Center for Molecular Studies, Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA
| | - Charles H Lawrie
- Department of Oncology, Biodonostia Research Institute, San Sebastián 20014, Spain.,Nuffield Department of Clinical Laboratory Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Rachel Pallapati
- The Vontz Center for Molecular Studies, Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA
| | - Veena Potluri
- The Vontz Center for Molecular Studies, Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA
| | - Christian Kuntzen
- Department of Medicine, Bridgeport Hospital, 267 Grant Street, Bridgeport, Connecticut 06610, USA
| | - Sohaib Khan
- The Vontz Center for Molecular Studies, Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA
| | - David R Plas
- The Vontz Center for Molecular Studies, Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA
| | - Robert Z Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Marta Chesi
- Department of Hematology/Oncology , Mayo Clinic, 13400 E. Shea Boulevard, Scottsdale, Arizona 85259, USA
| | - W Michael Kuehl
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Building 37, Room 6002C, Bethesda, Maryland 20892, USA
| | - P Leif Bergsagel
- Department of Hematology/Oncology , Mayo Clinic, 13400 E. Shea Boulevard, Scottsdale, Arizona 85259, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California, San Diego, California 92093, USA
| | - Sivakumar Vallabhapurapu
- The Vontz Center for Molecular Studies, Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA
| |
Collapse
|
29
|
Supriady H, Kamarudin MNA, Chan CK, Goh BH, Kadir HA. SMEAF attenuates the production of pro-inflammatory mediators through the inactivation of Akt-dependent NF-κB, p38 and ERK1/2 pathways in LPS-stimulated BV-2 microglial cells. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.05.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
30
|
Vlahopoulos SA, Cen O, Hengen N, Agan J, Moschovi M, Critselis E, Adamaki M, Bacopoulou F, Copland JA, Boldogh I, Karin M, Chrousos GP. Dynamic aberrant NF-κB spurs tumorigenesis: a new model encompassing the microenvironment. Cytokine Growth Factor Rev 2015; 26:389-403. [PMID: 26119834 PMCID: PMC4526340 DOI: 10.1016/j.cytogfr.2015.06.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 06/15/2015] [Indexed: 12/15/2022]
Abstract
Recently it was discovered that a transient activation of transcription factor NF-κB can give cells properties essential for invasiveness and cancer initiating potential. In contrast, most oncogenes to date were characterized on the basis of mutations or by their constitutive overexpression. Study of NF-κB actually leads to a far more dynamic perspective on cancer: tumors caused by diverse oncogenes apparently evolve into cancer after loss of feedback regulation for NF-κB. This event alters the cellular phenotype and the expression of hormonal mediators, modifying signals between diverse cell types in a tissue. The result is a disruption of stem cell hierarchy in the tissue, and pervasive changes in the microenvironment and immune response to the malignant cells.
Collapse
Affiliation(s)
- Spiros A Vlahopoulos
- First Department of Pediatrics, University of Athens, Horemeio Research Laboratory, Athens, Greece.
| | - Osman Cen
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, United States
| | - Nina Hengen
- Bernard J. Dunn School of Pharmacy, Shenandoah University, United States
| | - James Agan
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, United States
| | - Maria Moschovi
- First Department of Pediatrics, University of Athens, Horemeio Research Laboratory, Athens, Greece
| | - Elena Critselis
- First Department of Pediatrics, University of Athens, Horemeio Research Laboratory, Athens, Greece
| | - Maria Adamaki
- First Department of Pediatrics, University of Athens, Horemeio Research Laboratory, Athens, Greece
| | - Flora Bacopoulou
- First Department of Pediatrics, University of Athens, Horemeio Research Laboratory, Athens, Greece
| | - John A Copland
- Mayo Clinic Comprehensive Cancer Center, Department of Cancer Biology, United States
| | - Istvan Boldogh
- Department of Microbiology and Immunology, School of Medicine, University of Texas Medical Branch at Galveston, United States
| | - Michael Karin
- Department of Pharmacology, University of California, San Diego, United States
| | - George P Chrousos
- First Department of Pediatrics, University of Athens, Horemeio Research Laboratory, Athens, Greece
| |
Collapse
|
31
|
Schmitz ML, de la Vega L. New Insights into the Role of Histone Deacetylases as Coactivators of Inflammatory Gene Expression. Antioxid Redox Signal 2015; 23:85-98. [PMID: 24359078 DOI: 10.1089/ars.2013.5750] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
SIGNIFICANCE The expression and/or activity of histone deacetylases (HDACs) can be regulated by a variety of environmental conditions, including inflammation and oxidative stress. These events result in diminished or exaggerated protein acetylation, both of which can be causative for many ailments. While the anti-inflammatory activity of HDAC inhibitors (HDACis) is well known, recent studies started unraveling details of the molecular mechanisms underlying the pro-inflammatory function of HDACs. RECENT ADVANCES Recent evidence shows that HDACs are found in association with transcribed regions and ensure proper transcription by maintaining acetylation homeostasis. We also discuss current insights in the molecular mechanisms mediating acetylation-dependent inhibition of pro-inflammatory transcription factors of the NF-κB, HIF-1, IRF, and STAT families. CRITICAL ISSUES The high number of acetylations and the complexity of the regulatory consequences make it difficult to assign biological effects directly to a single acetylation event. The vast majority of acetylated proteins are nonhistone proteins, and it remains to be shown whether the therapeutic effects of HDACis are attributable to altered histone acetylation. FUTURE DIRECTIONS In the traditional view, only exaggerated acetylation is harmful and causative for diseases. Recent data show the relevance of acetylation homeostasis and suggest that both diminished and inflated acetylation can enable the development of ailments. Since acetylation of nonhistone proteins is essential for the induction of a substantial part of the inflammatory gene expression program, HDACis are more than "epigenetic drugs." The identification of substrates for individual HDACs will be the prerequisite for the adequate use of highly specific HDACis.
Collapse
Affiliation(s)
- Michael Lienhard Schmitz
- 1 Medical Faculty, Institute of Biochemistry, Justus-Liebig-University , Giessen, Germany .,2 The German Center for Lung Research, Giessen, Germany
| | - Laureano de la Vega
- 3 Division of Cancer Research, Medical Research Institute, Jacqui Wood Cancer Centre, University of Dundee , Ninewells Hospital and Medical School, Dundee, United Kingdom
| |
Collapse
|
32
|
Mizutani T, Ishizaka A, Furuichi Y. The Werner Protein Acts as a Coactivator of Nuclear Factor κB (NF-κB) on HIV-1 and Interleukin-8 (IL-8) Promoters. J Biol Chem 2015; 290:18391-9. [PMID: 26037922 DOI: 10.1074/jbc.m115.657155] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Indexed: 12/22/2022] Open
Abstract
The Werner syndrome helicase (WRN) plays a role in maintaining genomic stability. The lack of WRN results in Werner syndrome, a rare autosomal recessive genetic disorder, which causes premature aging accompanied by many complications such as rare forms of cancer and type 2 diabetes. However, the underlying mechanisms of these complications, arising due to the loss of WRN, are poorly understood. In this study, we demonstrated the function of WRN in transcriptional regulation of NF-κB targets. WRN physically interacts via its RecQ C-terminal (RQC) domain with the Rel homology domain of both the RelA (p65) and the p50 subunits of NF-κB. In the steady state, WRN is recruited to HIV-1 long terminal repeat (LTR), a typical NF-κB-responsive promoter, as well as the p50/p50 homodimer, in an NF-κB site-dependent manner. The amount of WRN on LTR increased along with the transactivating RelA/p50 heterodimer in response to TNF-α stimulation. Further, a knockdown of WRN reduced the transactivation of LTR in exogenous RelA/p50-introduced or TNF-α-stimulated cells. Additionally, knockdown of WRN reduced TNF-α stimulation-induced activation of the endogenous promoter of IL-8, an NF-κB-responsive gene, and WRN increased its association with the IL-8 promoter region together with RelA/p50 after TNF-α stimulation. In conjunction with studies that have shown NF-κB to be a key regulator of aging and inflammation, our results indicate a novel role of WRN in transcriptional regulation. Along with NF-κB, the loss of WRN is expected to result in incorrect regulation of downstream targets and leads to immune abnormalities and homeostatic disruption.
Collapse
Affiliation(s)
- Taketoshi Mizutani
- From the Laboratory of Virology, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (BIKAKEN), Tokyo, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021 and
| | - Aya Ishizaka
- From the Laboratory of Virology, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (BIKAKEN), Tokyo, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021 and
| | - Yasuhiro Furuichi
- GeneCare Research Institute Co., Ltd., 19-2 Kajiwara, Kamakura, Kanagawa 247-0063, Japan
| |
Collapse
|
33
|
Sohn C, Lee A, Qiao Y, Loupasakis K, Ivashkiv LB, Kalliolias GD. Prolonged tumor necrosis factor α primes fibroblast-like synoviocytes in a gene-specific manner by altering chromatin. Arthritis Rheumatol 2015; 67:86-95. [PMID: 25199798 DOI: 10.1002/art.38871] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 09/02/2014] [Indexed: 12/27/2022]
Abstract
OBJECTIVE During the course of rheumatoid arthritis (RA), fibroblast-like synoviocytes (FLS) are chronically exposed to an inflammatory milieu. The purpose of this study was to test the hypothesis that prolonged exposure of FLS to tumor necrosis factor α (TNFα) augments inflammatory responses to secondary stimuli (priming effect). METHODS FLS obtained from RA patients were exposed to TNFα for 3 days and were then stimulated with interferons (IFNs). Expression of IFN target genes was measured by real-time quantitative reverse transcription-polymerase chain reaction analysis and enzyme-linked immunosorbent assay. Total STAT-1 protein and IFN-mediated STAT-1 activation were evaluated by Western blotting. Total histone levels, histone acetylation, and NF-κB p65 and RNA polymerase II (Pol II) recruitment were measured at the CXCL10 promoter (encodes IFNγ-inducible 10-kd protein [IP-10]) by chromatin immunoprecipitation assays. RESULTS Prolonged pre-exposure of FLS to TNFα enhanced the magnitude and extended the kinetics of CXCL10/IP-10, CXCL9, and CXCL11 production upon subsequent IFN stimulation. This phenotype was retained over a period of days, even after the removal of TNFα. Prolonged TNFα exposure decreased histone levels, increased acetylation of the remaining histones, and heightened recruitment of NF-κB p65 and Pol II to the CXCL10 promoter. In parallel, an increase in intracellular STAT-1 led to amplification of IFN-induced STAT-1 activation. CONCLUSION Our study reveals a novel pathogenic function of TNFα, namely, prolonged and gene-specific priming of FLS for enhanced transcription of inflammatory chemokine genes due to the priming of chromatin, the sustained activation of NF-κB, and the amplification of STAT-1 activation downstream of IFNs. These data also suggest that FLS gain an "inflammatory memory" upon prolonged exposure to TNFα.
Collapse
|
34
|
Zhou J, Ching YQ, Chng WJ. Aberrant nuclear factor-kappa B activity in acute myeloid leukemia: from molecular pathogenesis to therapeutic target. Oncotarget 2015; 6:5490-5500. [PMID: 25823927 PMCID: PMC4467382 DOI: 10.18632/oncotarget.3545] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 02/15/2015] [Indexed: 02/07/2023] Open
Abstract
The overall survival of patients with acute myeloid leukemia (AML) has not been improved significantly over the last decade. Molecularly targeted agents hold promise to change the therapeutic landscape in AML. The nuclear factor kappa B (NF-κB) controls a plethora of biological process through switching on and off its long list of target genes. In AML, constitutive NF-κB has been detected in 40% of cases and its aberrant activity enable leukemia cells to evade apoptosis and stimulate proliferation. These facts suggest that NF-κB signaling pathway plays a fundamental role in the development of AML and it represents an attractive target for the intervention of AML. This review summarizes our current knowledge of NF-κB signaling transduction including canonical and non-canonical NF-κB pathways. Then we specifically highlight what factors contribute to the aberrant activation of NF-κB activity in AML, followed by an overview of 8 important clinical trials of the first FDA approved proteasome inhibitor, Bortezomib (Velcade), which is a NF-κB inhibitor too, in combination with other therapeutic agents in patients with AML. Finally, this review discusses the future directions of NF-κB inhibitor in treatment of AML, especially in targeting leukemia stem cells (LSCs).
Collapse
Affiliation(s)
- Jianbiao Zhou
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, Republic of Singapore
| | - Ying Qing Ching
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, Republic of Singapore
| | - Wee-Joo Chng
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, Republic of Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Department of Hematology-Oncology, National University Cancer Institute of Singapore (NCIS), The National University Health System (NUHS), Singapore, Republic of Singapore
| |
Collapse
|
35
|
Transcriptional regulation of chemokine expression in ovarian cancer. Biomolecules 2015; 5:223-43. [PMID: 25790431 PMCID: PMC4384120 DOI: 10.3390/biom5010223] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/04/2015] [Accepted: 03/09/2015] [Indexed: 12/14/2022] Open
Abstract
The increased expression of pro-inflammatory and pro-angiogenic chemokines contributes to ovarian cancer progression through the induction of tumor cell proliferation, survival, angiogenesis, and metastasis. The substantial potential of these chemokines to facilitate the progression and metastasis of ovarian cancer underscores the need for their stringent transcriptional regulation. In this Review, we highlight the key mechanisms that regulate the transcription of pro-inflammatory chemokines in ovarian cancer cells, and that have important roles in controlling ovarian cancer progression. We further discuss the potential mechanisms underlying the increased chemokine expression in drug resistance, along with our perspective for future studies.
Collapse
|
36
|
Franco HL, Nagari A, Kraus WL. TNFα signaling exposes latent estrogen receptor binding sites to alter the breast cancer cell transcriptome. Mol Cell 2015; 58:21-34. [PMID: 25752574 DOI: 10.1016/j.molcel.2015.02.001] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 11/18/2014] [Accepted: 01/27/2015] [Indexed: 01/08/2023]
Abstract
The interplay between mitogenic and proinflammatory signaling pathways plays key roles in determining the phenotypes and clinical outcomes of breast cancers. Using GRO-seq in MCF-7 cells, we defined the immediate transcriptional effects of crosstalk between estradiol (E2) and TNFα, identifying a large set of target genes whose expression is rapidly altered with combined E2 + TNFα treatment, but not with either agent alone. The pleiotropic effects on gene transcription in response to E2 + TNFα are orchestrated by extensive remodeling of the ERα enhancer landscape in an NF-κB- and FoxA1-dependent manner. In addition, expression of the de novo and synergistically regulated genes is strongly associated with clinical outcomes in breast cancers. Together, our genomic and molecular analyses indicate that TNFα signaling, acting in pathways culminating in the redistribution of NF-κB and FoxA1 binding sites across the genome, creates latent ERα binding sites that underlie altered patterns of gene expression and clinically relevant cellular responses.
Collapse
Affiliation(s)
- Hector L Franco
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences and Division of Basic Reproductive Biology Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anusha Nagari
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences and Division of Basic Reproductive Biology Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - W Lee Kraus
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences and Division of Basic Reproductive Biology Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
37
|
Deoxysappanone B, a homoisoflavone from the Chinese medicinal plant Caesalpinia sappan L., protects neurons from microglia-mediated inflammatory injuries via inhibition of IκB kinase (IKK)-NF-κB and p38/ERK MAPK pathways. Eur J Pharmacol 2015; 748:18-29. [DOI: 10.1016/j.ejphar.2014.12.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/09/2014] [Accepted: 12/11/2014] [Indexed: 01/27/2023]
|
38
|
Kunes P, Mandak J, Holubcova Z, Kolackova M, Krejsek J. Actual position of interleukin(IL)-33 in atherosclerosis and heart failure: Great Expectations or En attendant Godot? Perfusion 2014; 30:356-74. [DOI: 10.1177/0267659114562269] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Atherosclerosis has been recognized as an inflammatory/autoimmune disease. The long-standing low-grade inflammation which fuels its development is primarily focused on the components of the vessel wall. Originally, inflammation in atherogenesis was supposed to be driven by the pro-inflammatory Th1 cellular and cytokine immune response. On the basis of accumulating evidence, this view has been re-evaluated to include the Th17/Th1 axis which is shared by most diseases of sterile inflammation. The anti-inflammatory Th2 cellular and cytokine immune response is initiated concomitantly with the former two, the latter dampening their harmful reactions which culminate in full-blown atherosclerosis. Interleukin-33, a novel member of the IL-1 cytokine superfamily, was suggested to take part in the anti-atherogenic response by mediating the Th1-to-Th2 switch of the immune reactions. However, IL-33 is a multifaceted mediator with both pro- and anti-inflammatory activities, also called a “dual factor” or a “Janus face” interleukin. IL-33 occurs both in an extracellular (cytokine-like) and in a nuclear-bound (transcription factor-like) form, each of them performing distinct activities of their own. This review article presents the latest data relevant to IL-33’s role in atherosclerosis and cardiac diseases as perceived by a cardiologist and a cardiac surgeon.
Collapse
Affiliation(s)
- P Kunes
- Department of Cardiac Surgery, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| | - J Mandak
- Department of Cardiac Surgery, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| | - Z Holubcova
- Department of Cardiac Surgery, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| | - M Kolackova
- Department of Clinical Immunology and Allergology, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| | - J Krejsek
- Department of Clinical Immunology and Allergology, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Czech Republic
| |
Collapse
|
39
|
Deficiency in p53 is required for doxorubicin induced transcriptional activation of NF-кB target genes in human breast cancer. Oncotarget 2014; 5:196-210. [PMID: 24344116 PMCID: PMC3960201 DOI: 10.18632/oncotarget.1556] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
NF-κB has been linked to doxorubicin resistance in breast cancer patients. NF-κB nuclear translocation and DNA binding in doxorubicin treated-breast cancer cells have been extensively examined; however its functional relevance at transcriptional level on NF-κB -dependent genes and the biological consequences are unclear. We studied NF-κB -dependent gene expression induced by doxorubicin in breast cancer cells and fresh human cancer specimens with different genetic backgrounds focusing on their p53 status. NF-κB -dependent signature of doxorubicin was identified by gene expression microarrays in breast cancer cells treated with doxorubicin and the IKKβ-inhibitor MLN120B, and confirmed ex vivo in human cancer samples. The association with p53 was functionally validated. Finally, NF-κB activation and p53 status was determined in a cohort of breast cancer patients treated with adjuvant doxorubicin-based chemotherapy. Doxorubicin treatment in the p53-mutated MDA-MB-231 cells resulted in NF NF-κB driven-gene transcription signature. Modulation of genes related with invasion, metastasis and chemoresistance (ICAM-1, CXCL1, TNFAIP3, IL8) were confirmed in additional doxorubicin-treated cell lines and fresh primary human breast tumors. In both systems, p53-defcient background correlated with the activation of the NF-κB -dependent signature. Furthermore, restoration of p53WT in the mutant p53 MDA-MB-231 cells impaired NF-κB driven transcription induced by doxorubicin. Moreover, a p53 deficient background and nuclear NF-κB /p65 in breast cancer patients correlated with reduced disease free-survival. This study supports that p53 deficiency is necessary for a doxorubicin driven NF-κB -response that limits doxorubicin cytotoxicity in breast cancer and is linked to an aggressive clinical behavior.
Collapse
|
40
|
Diermeier S, Kolovos P, Heizinger L, Schwartz U, Georgomanolis T, Zirkel A, Wedemann G, Grosveld F, Knoch TA, Merkl R, Cook PR, Längst G, Papantonis A. TNFα signalling primes chromatin for NF-κB binding and induces rapid and widespread nucleosome repositioning. Genome Biol 2014; 15:536. [PMID: 25608606 PMCID: PMC4268828 DOI: 10.1186/s13059-014-0536-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 11/07/2014] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The rearrangement of nucleosomes along the DNA fiber profoundly affects gene expression, but little is known about how signalling reshapes the chromatin landscape, in three-dimensional space and over time, to allow establishment of new transcriptional programs. RESULTS Using micrococcal nuclease treatment and high-throughput sequencing, we map genome-wide changes in nucleosome positioning in primary human endothelial cells stimulated with tumour necrosis factor alpha (TNFα) - a proinflammatory cytokine that signals through nuclear factor kappa-B (NF-κB). Within 10 min, nucleosomes reposition at regions both proximal and distal to NF-κB binding sites, before the transcription factor quantitatively binds thereon. Similarly, in long TNFα-responsive genes, repositioning precedes transcription by pioneering elongating polymerases and appears to nucleate from intragenic enhancer clusters resembling super-enhancers. By 30 min, widespread repositioning throughout megabase pair-long chromosomal segments, with consequential effects on three-dimensional structure (detected using chromosome conformation capture), is seen. CONCLUSIONS Whilst nucleosome repositioning is viewed as a local phenomenon, our results point to effects occurring over multiple scales. Here, we present data in support of a TNFα-induced priming mechanism, mostly independent of NF-κB binding and/or elongating RNA polymerases, leading to a plastic network of interactions that affects DNA accessibility over large domains.
Collapse
Affiliation(s)
- Sarah Diermeier
- />Department of Biochemistry III, University of Regensburg, Universität Strasse 31, 93053 Regensburg, Germany
- />Present address: Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, 11724 NY USA
| | - Petros Kolovos
- />Cell Biology and Genetics, Erasmus Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
- />Biophysical Genomics, Erasmus Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Leonhard Heizinger
- />Institute of Biophysics and Physical Biochemistry, University of Regensburg, 93040 Regensburg, Germany
| | - Uwe Schwartz
- />Department of Biochemistry III, University of Regensburg, Universität Strasse 31, 93053 Regensburg, Germany
| | - Theodore Georgomanolis
- />Centre for Molecular Medicine, University of Cologne, Robert-Koch-Strasse 21, 50931 Cologne, Germany
| | - Anne Zirkel
- />Centre for Molecular Medicine, University of Cologne, Robert-Koch-Strasse 21, 50931 Cologne, Germany
| | - Gero Wedemann
- />Institute for Applied Computer Science, University of Applied Sciences Stralsund, Zur Schwedenschanze 15, 18435 Stralsund, Germany
| | - Frank Grosveld
- />Cell Biology and Genetics, Erasmus Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Tobias A Knoch
- />Biophysical Genomics, Erasmus Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
- />BioQuant & German Cancer Research Center, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Rainer Merkl
- />Institute of Biophysics and Physical Biochemistry, University of Regensburg, 93040 Regensburg, Germany
| | - Peter R Cook
- />Sir William Dunn School of Pathology, University of Oxford, South Parks Road, OX1 3RE Oxford, United Kingdom
| | - Gernot Längst
- />Department of Biochemistry III, University of Regensburg, Universität Strasse 31, 93053 Regensburg, Germany
| | - Argyris Papantonis
- />Centre for Molecular Medicine, University of Cologne, Robert-Koch-Strasse 21, 50931 Cologne, Germany
- />Sir William Dunn School of Pathology, University of Oxford, South Parks Road, OX1 3RE Oxford, United Kingdom
| |
Collapse
|
41
|
IKK phosphorylates RelB to modulate its promoter specificity and promote fibroblast migration downstream of TNF receptors. Proc Natl Acad Sci U S A 2014; 111:14794-9. [PMID: 25267645 DOI: 10.1073/pnas.1410124111] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
TNFα is a potent cytokine that plays a critical role in numerous cellular processes, particularly immune and inflammatory responses, programmed cell death, angiogenesis, and cell migration. Thus, understanding the molecular mechanisms that mediate TNFα-induced cellular responses is a crucial issue. It is generally accepted that global DNA binding activity of the NF-κB avian reticuloendotheliosis viral (v-rel) oncogene related B (RelB) subunit is not induced upon TNFα treatment in fibroblasts, despite its TNFα-induced nuclear accumulation. Here, we demonstrate that RelB plays a critical role in promoting fibroblast migration upon prolonged TNFα treatment. We identified the two kinases IκB kinase α (IKKα) and IκB kinase β (IKKβ) as RelB interacting partners whose activation by TNFα promotes RelB phosphorylation at serine 472. Once phosphorylated on serine 472, nuclear RelB dissociates from its interaction with the inhibitory protein IκBα and binds to the promoter of critical migration-associated genes, such as the matrix metallopeptidase 3 (MMP3). Further, we show that RelB serine 472 phosphorylation status controls MMP3 expression and promigration activity downstream of TNF receptors. Our findings provide new insights into the regulation of RelB activity and reveal a novel link between selective NF-κB target gene expression and cellular response in response to TNFα.
Collapse
|
42
|
Saliba DG, Heger A, Eames HL, Oikonomopoulos S, Teixeira A, Blazek K, Androulidaki A, Wong D, Goh FG, Weiss M, Byrne A, Pasparakis M, Ragoussis J, Udalova IA. IRF5:RelA interaction targets inflammatory genes in macrophages. Cell Rep 2014; 8:1308-17. [PMID: 25159141 PMCID: PMC4471814 DOI: 10.1016/j.celrep.2014.07.034] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 01/10/2014] [Accepted: 07/22/2014] [Indexed: 12/23/2022] Open
Abstract
Interferon Regulatory Factor 5 (IRF5) plays a major role in setting up an inflammatory macrophage phenotype, but the molecular basis of its transcriptional activity is not fully understood. In this study, we conduct a comprehensive genome-wide analysis of IRF5 recruitment in macrophages stimulated with bacterial lipopolysaccharide and discover that IRF5 binds to regulatory elements of highly transcribed genes. Analysis of protein:DNA microarrays demonstrates that IRF5 recognizes the canonical IRF-binding (interferon-stimulated response element [ISRE]) motif in vitro. However, IRF5 binding in vivo appears to rely on its interactions with other proteins. IRF5 binds to a noncanonical composite PU.1:ISRE motif, and its recruitment is aided by RelA. Global gene expression analysis in macrophages deficient in IRF5 and RelA highlights the direct role of the RelA:IRF5 cistrome in regulation of a subset of key inflammatory genes. We map the RelA:IRF5 interaction domain and suggest that interfering with it would offer selective targeting of macrophage inflammatory activities. Genome-wide function of IRF5 in LPS-stimulated macrophages was analyzed IRF5 cistrome overlaps with RelA cistrome at multiple loci IRF5 targets regulatory elements of highly inducible inflammatory genes IRF5 recruitment to key inflammatory loci is assisted by RelA
Collapse
Affiliation(s)
- David G Saliba
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX37FY, UK
| | - Andreas Heger
- CGAT, MRC Functional Genomics Unit, University of Oxford, South Parks Road, Oxford OX13PT, UK
| | - Hayley L Eames
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX37FY, UK
| | - Spyros Oikonomopoulos
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Ana Teixeira
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Katrina Blazek
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX37FY, UK
| | - Ariadne Androulidaki
- Institute for Genetics, University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne 50931, Germany
| | - Daniel Wong
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Fui G Goh
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX37FY, UK
| | - Miriam Weiss
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX37FY, UK
| | - Adam Byrne
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX37FY, UK
| | - Manolis Pasparakis
- Institute for Genetics, University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne 50931, Germany
| | - Jiannis Ragoussis
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Irina A Udalova
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX37FY, UK.
| |
Collapse
|
43
|
Gallagher SJ, Mijatov B, Gunatilake D, Gowrishankar K, Tiffen J, James W, Jin L, Pupo G, Cullinane C, McArthur GA, Tummino PJ, Rizos H, Hersey P. Control of NF-kB activity in human melanoma by bromodomain and extra-terminal protein inhibitor I-BET151. Pigment Cell Melanoma Res 2014; 27:1126-37. [PMID: 24924589 DOI: 10.1111/pcmr.12282] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Accepted: 06/03/2014] [Indexed: 01/18/2023]
Abstract
The transcription factor NF-kappaB (NF-kB) is a key regulator of cytokine and chemokine production in melanoma and is responsible for symptoms such as anorexia, fatigue, and weight loss. In addition, NF-kB is believed to contribute to progression of the disease by upregulation of cell cycle and anti-apoptotic genes and to contribute to resistance against targeted therapies and immunotherapy. In this study, we have examined the ability of the bromodomain and extra-terminal (BET) protein inhibitor I-BET151 to inhibit NF-kB in melanoma cells. We show that I-BET151 is a potent, selective inhibitor of a number of NF-kB target genes involved in induction of inflammation and cell cycle regulation and downregulates production of cytokines such as IL-6 and IL-8. SiRNA studies indicate that BRD2 is the main BET protein involved in regulation of NF-kB and that I-BET151 caused transcriptional downregulation of the NF-kB subunit p105/p50. These results suggest that BET inhibitors may have an important role in treatment of melanoma where activation of NF-kB may have a key pathogenic role.
Collapse
Affiliation(s)
- Stuart J Gallagher
- Melanoma Research Group, Kolling Institute of Medical Research, University of Sydney, St Leonards, NSW, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
The NF-κB family of inducible transcription factors is activated in response to a variety of stimuli. Amongst the best-characterized inducers of NF-κB are members of the TNF family of cytokines. Research on NF-κB and TNF have been tightly intertwined for more than 25 years. Perhaps the most compelling examples of the interconnectedness of NF-κB and the TNF have come from analysis of knock-out mice that are unable to activate NF-κB. Such mice die embryonically, however, deletion of TNF or TNFR1 can rescue the lethality thereby illustrating the important role of NF-κB as the key regulator of transcriptional responses to TNF. The physiological connections between NF-κB and TNF cytokines are numerous and best explored in articles focusing on a single TNF family member. Instead, in this review, we explore general mechanisms of TNF cytokine signaling, with a focus on the upstream signaling events leading to activation of the so-called canonical and noncanonical NF-κB pathways by TNFR1 and CD40, respectively.
Collapse
Affiliation(s)
- Matthew S Hayden
- Department of Microbiology and Immunology, Columbia University, College of Physicians & Surgeons, New York, NY 10032, USA; Department of Dermatology, Columbia University, College of Physicians & Surgeons, New York, NY 10032, USA.
| | - Sankar Ghosh
- Department of Microbiology and Immunology, Columbia University, College of Physicians & Surgeons, New York, NY 10032, USA.
| |
Collapse
|
45
|
Chang TP, Kim M, Vancurova I. Analysis of TGFβ1 and IL-10 transcriptional regulation in CTCL cells by chromatin immunoprecipitation. Methods Mol Biol 2014; 1172:329-41. [PMID: 24908319 DOI: 10.1007/978-1-4939-0928-5_30] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The immunosuppressive cytokines transforming growth factor β1 (TGFβ1) and interleukin-10 (IL-10) regulate a variety of biological processes including differentiation, proliferation, tissue repair, tumorigenesis, inflammation, and host defense. Aberrant expression of TGFβ1 and IL-10 has been associated with many types of autoimmune and inflammatory disorders, as well as with many types of cancer and leukemia. Patients with cutaneous T cell lymphoma (CTCL) have high levels of malignant CD4+ T cells expressing IL-10 and TGFβ1 that suppress the immune system and diminish the antitumor responses. The transcriptional regulation of TGFβ1 and IL-10 expression is orchestrated by several transcription factors, including NFκB. However, while the transcriptional regulation of pro-inflammatory and anti-apoptotic genes by NFκB has been studied extensively, much less is known about the NFκB regulation of immunosuppressive genes. In this chapter, we describe a protocol that uses chromatin immunoprecipitation (ChIP) to analyze the transcriptional regulation of TGFβ1 and IL-10 by measuring recruitment of NFκB p65, p50, c-Rel, Rel-B, and p52 subunits to TGFβ1 and IL-10 promoters in human CTCL Hut-78 cells.
Collapse
Affiliation(s)
- Tzu-Pei Chang
- Department of Biology, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | | | | |
Collapse
|
46
|
Singha B, Gatla HR, Manna S, Chang TP, Sanacora S, Poltoratsky V, Vancura A, Vancurova I. Proteasome inhibition increases recruitment of IκB kinase β (IKKβ), S536P-p65, and transcription factor EGR1 to interleukin-8 (IL-8) promoter, resulting in increased IL-8 production in ovarian cancer cells. J Biol Chem 2013; 289:2687-700. [PMID: 24337575 DOI: 10.1074/jbc.m113.502641] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Proinflammatory and pro-angiogenic chemokine interleukin-8 (IL-8, CXCL8) contributes to ovarian cancer progression through its induction of tumor cell proliferation, survival, angiogenesis, and metastasis. Proteasome inhibition by bortezomib, which has been used as a frontline therapy in multiple myeloma, has shown only limited effectiveness in ovarian cancer and other solid tumors. However, the responsible mechanisms remain elusive. Here, we show that proteasome inhibition dramatically increases the IL-8 expression and release in ovarian cancer cells. The responsible mechanism involves an increased nuclear accumulation of IκB kinase β (IKKβ) and an increased recruitment of the nuclear IKKβ, p65-phosphorylated at Ser-536, and the transcription factor early growth response-1 (EGR-1) to the endogenous IL-8 promoter. Coimmunoprecipitation studies identified the nuclear EGR-1 associated with IKKβ and with p65, with preferential binding to S536P-p65. Both IKKβ activity and EGR-1 expression are required for the increased IL-8 expression induced by proteasome inhibition in ovarian cancer cells. Interestingly, in multiple myeloma cells the IL-8 release is not increased by bortezomib. Together, these data indicate that the increased IL-8 release may represent one of the underlying mechanisms responsible for the decreased effectiveness of proteasome inhibition in ovarian cancer treatment and identify IKKβ and EGR-1 as potential new targets in ovarian cancer combination therapies.
Collapse
|
47
|
Walker T, Adamson A, Jackson DA. BCL-3 attenuation of TNFA expression involves an incoherent feed-forward loop regulated by chromatin structure. PLoS One 2013; 8:e77015. [PMID: 24130828 PMCID: PMC3794926 DOI: 10.1371/journal.pone.0077015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 08/29/2013] [Indexed: 11/23/2022] Open
Abstract
Induction of genes is rarely an isolated event; more typically occurring as part of a web of parallel interactions, or motifs, which act to refine and control gene expression. Here, we define an Incoherent Feed-forward Loop motif in which TNFα-induced NF-κB signalling activates expression of the TNFA gene itself and also controls synthesis of the negative regulator BCL-3. While sharing a common inductive signal, the two genes have distinct temporal expression profiles. Notably, while the TNFA gene promoter is primed to respond immediately to activated NF-κB in the nucleus, induction of BCL3 expression only occurs after a time delay of about 1h. We show that this time delay is defined by remodelling of the BCL3 gene promoter, which is required to activate gene expression, and characterise the chromatin delayed induction of BCL3 expression using mathematical models. The models show how a delay in inhibitor production effectively uncouples the rate of response to inflammatory cues from the final magnitude of inhibition. Hence, within this regulatory motif, a delayed (incoherent) feed-forward loop together with differential rates of TNFA (fast) and BCL3 (slow) mRNA turnover provide robust, pulsatile expression of TNFα . We propose that the structure of the BCL-3-dependent regulatory motif has a beneficial role in modulating expression dynamics and the inflammatory response while minimising the risk of pathological hyper-inflammation.
Collapse
Affiliation(s)
- Thomas Walker
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Antony Adamson
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Dean A. Jackson
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
48
|
Lone IN, Shukla MS, Charles Richard JL, Peshev ZY, Dimitrov S, Angelov D. Binding of NF-κB to nucleosomes: effect of translational positioning, nucleosome remodeling and linker histone H1. PLoS Genet 2013; 9:e1003830. [PMID: 24086160 PMCID: PMC3784511 DOI: 10.1371/journal.pgen.1003830] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 08/12/2013] [Indexed: 01/29/2023] Open
Abstract
NF-κB is a key transcription factor regulating the expression of inflammatory responsive genes. How NF-κB binds to naked DNA templates is well documented, but how it interacts with chromatin is far from being clear. Here we used a combination of UV laser footprinting, hydroxyl footprinting and electrophoretic mobility shift assay to investigate the binding of NF-κB to nucleosomal templates. We show that NF-κB p50 homodimer is able to bind to its recognition sequence, when it is localized at the edge of the core particle, but not when the recognition sequence is at the interior of the nucleosome. Remodeling of the nucleosome by the chromatin remodeling machine RSC was not sufficient to allow binding of NF-κB to its recognition sequence located in the vicinity of the nucleosome dyad, but RSC-induced histone octamer sliding allowed clearly detectable binding of NF-κB with the slid particle. Importantly, nucleosome dilution-driven removal of H2A–H2B dimer led to complete accessibility of the site located close to the dyad to NF-κB. Finally, we found that NF-κB was able to displace histone H1 and prevent its binding to nucleosome. These data provide important insight on the role of chromatin structure in the regulation of transcription of NF-κB dependent genes. In eukaryotes DNA is hierarchically packaged into chromatin by histones. The fundamental subunit of chromatin is the nucleosome. The packaging of DNA into nucleosomes not only restricts DNA accessibility for regulatory proteins but also provides opportunities to regulate DNA based processes. Accessibility of transcription factor NF-κB to their recognition sequences embedded in nucleosomes is highly controversial. On one hand in vivo studies have suggested that packaging of DNA into chromatin plays an important role in regulating the expression of NF-κB dependent genes, and on the other hand some in vitro studies reported that NF-κB can bind by itself to its recognition sequences embedded in the nucleosome. In this study, we show that NF-κB can specifically bind to its recognition sequences placed at the end of the nucleosome but not when placed inside the nucleosome core. We then demonstrate that disruption of nucleosome is necessary for the productive binding of NF-κB. Finally, we show that the presence of histone H1 does not affect the specific binding of NF-κB to its cognate sequence, when its binding region overlaps with the binding site of NF-κB. We propose that histone eviction is needed for NF-κB to bind specifically to its recognition sequence embedded in the nucleosome.
Collapse
Affiliation(s)
- Imtiaz Nisar Lone
- Université de Lyon, Laboratoire de Biologie Moléculaire de la Cellule, CNRS-UMR 5239, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Manu Shubhdarshan Shukla
- Université de Lyon, Laboratoire de Biologie Moléculaire de la Cellule, CNRS-UMR 5239, Ecole Normale Supérieure de Lyon, Lyon, France
- Université Joseph Fourier - Grenoble 1, INSERM Institut Albert Bonniot, U823, Site Santé-BP 170, Grenoble, France
| | - John Lalith Charles Richard
- Université de Lyon, Laboratoire de Biologie Moléculaire de la Cellule, CNRS-UMR 5239, Ecole Normale Supérieure de Lyon, Lyon, France
- Université Joseph Fourier - Grenoble 1, INSERM Institut Albert Bonniot, U823, Site Santé-BP 170, Grenoble, France
| | - Zahary Yordanov Peshev
- Université de Lyon, Laboratoire de Biologie Moléculaire de la Cellule, CNRS-UMR 5239, Ecole Normale Supérieure de Lyon, Lyon, France
- Institute of Electronics, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Stefan Dimitrov
- Université Joseph Fourier - Grenoble 1, INSERM Institut Albert Bonniot, U823, Site Santé-BP 170, Grenoble, France
- * E-mail: (SD); (DA)
| | - Dimitar Angelov
- Université de Lyon, Laboratoire de Biologie Moléculaire de la Cellule, CNRS-UMR 5239, Ecole Normale Supérieure de Lyon, Lyon, France
- * E-mail: (SD); (DA)
| |
Collapse
|
49
|
Li Y, Liu H, Xu QS, Du YG, Xu J. Chitosan oligosaccharides block LPS-induced O-GlcNAcylation of NF-κB and endothelial inflammatory response. Carbohydr Polym 2013; 99:568-78. [PMID: 24274545 DOI: 10.1016/j.carbpol.2013.08.082] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 08/23/2013] [Accepted: 08/24/2013] [Indexed: 02/06/2023]
Abstract
It is known that chitosan oligosaccharides (COS) suppress LPS-induced vascular endothelial inflammatory response by mechanism involving NF-κB blockade. It remains unknown how COS inhibit NF-κB. We provided evidence both in cultured endothelial cells and mouse model supporting a new mechanism. Regardless of the endothelial cell types, the LPS-induced NF-κB-dependent inflammatory gene expression was suppressed by COS, which was associated with reduced NF-κB nucleus translocation. LPS enhanced O-GlcNAc modification of NF-κB/p65 and activated NF-κB pathway, which could be prevented either by siRNA knockdown of O-GlcNAc transferase (OGT) or pretreatment with COS. Inhibition of either mitogen-activated protein kinase or superoxide generation abolishes LPS-induced NF-κB O-GlcNAcylation. Consistently, aortic tissues from LPS-treated mice presented enhanced NF-κB/p65 O-GlcNAcylation in association with upregulated gene expression of inflammatory cytokines in vascular tissues; however, pre-administration of COS prevented these responses. In conclusion, COS decreased OGT-dependent O-GlcNAcylation of NF-κB and thereby attenuated LPS-induced vascular endothelial inflammatory response.
Collapse
Affiliation(s)
- Yu Li
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; University of Chinese Academy of Sciences, Beijing 100049, China; Department of Medicine, University of Oklahoma Health Sciences Center, 941 Stanton L. Young Boulevard, Oklahoma City, OK 73104, USA
| | | | | | | | | |
Collapse
|
50
|
Adamik J, Wang KZQ, Unlu S, Su AJA, Tannahill GM, Galson DL, O’Neill LA, Auron PE. Distinct mechanisms for induction and tolerance regulate the immediate early genes encoding interleukin 1β and tumor necrosis factor α. PLoS One 2013; 8:e70622. [PMID: 23936458 PMCID: PMC3731334 DOI: 10.1371/journal.pone.0070622] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 06/19/2013] [Indexed: 12/16/2022] Open
Abstract
Interleukin-1β and Tumor Necrosis Factor α play related, but distinct, roles in immunity and disease. Our study revealed major mechanistic distinctions in the Toll-like receptor (TLR) signaling-dependent induction for the rapidly expressed genes (IL1B and TNF) coding for these two cytokines. Prior to induction, TNF exhibited pre-bound TATA Binding Protein (TBP) and paused RNA Polymerase II (Pol II), hallmarks of poised immediate-early (IE) genes. In contrast, unstimulated IL1B displayed very low levels of both TBP and paused Pol II, requiring the lineage-specific Spi-1/PU.1 (Spi1) transcription factor as an anchor for induction-dependent interaction with two TLR-activated transcription factors, C/EBPβ and NF-κB. Activation and DNA binding of these two pre-expressed factors resulted in de novo recruitment of TBP and Pol II to IL1B in concert with a permissive state for elongation mediated by the recruitment of elongation factor P-TEFb. This Spi1-dependent mechanism for IL1B transcription, which is unique for a rapidly-induced/poised IE gene, was more dependent upon P-TEFb than was the case for the TNF gene. Furthermore, the dependence on phosphoinositide 3-kinase for P-TEFb recruitment to IL1B paralleled a greater sensitivity to the metabolic state of the cell and a lower sensitivity to the phenomenon of endotoxin tolerance than was evident for TNF. Such differences in induction mechanisms argue against the prevailing paradigm that all IE genes possess paused Pol II and may further delineate the specific roles played by each of these rapidly expressed immune modulators.
Collapse
Affiliation(s)
- Juraj Adamik
- Department of Biological Sciences, Duquesne University, Pittsburgh, Pennsylvania, United States of America
| | - Kent Z. Q. Wang
- Department of Biological Sciences, Duquesne University, Pittsburgh, Pennsylvania, United States of America
| | - Sebnem Unlu
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - An-Jey A. Su
- Department of Biological Sciences, Duquesne University, Pittsburgh, Pennsylvania, United States of America
| | | | - Deborah L. Galson
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Luke A. O’Neill
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Philip E. Auron
- Department of Biological Sciences, Duquesne University, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|