1
|
Sigutova V, Xiang W, Regensburger M, Winner B, Prots I. Alpha-synuclein fine-tunes neuronal response to pro-inflammatory cytokines. Brain Behav Immun 2024; 122:216-230. [PMID: 39128571 DOI: 10.1016/j.bbi.2024.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/30/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024] Open
Abstract
Pro-inflammatory cytokines are emerging as neuroinflammatory mediators in Parkinson's disease (PD) due to their ability to act through neuronal cytokine receptors. Critical questions persist regarding the role of cytokines in neuronal dysfunction and their contribution to PD pathology. Specifically, the potential synergy of the hallmark PD protein alpha-synuclein (α-syn) with cytokines is of interest. We therefore investigated the direct impact of pro-inflammatory cytokines on neurons and hypothesized that α-syn pathology exacerbates cytokine-induced neuronal deficits in PD. iPSC-derived cortical neurons (CNs) from healthy controls and patients with α-syn gene locus duplication (SNCA dupl) were stimulated with IL-17A, TNF-α, IFN-γ, or a combination thereof. For rescue experiments, CNs were pre-treated with α-syn anti-oligomerisation compound NPT100-18A prior to IL-17A stimulation. Cytokine receptor expression, microtubule cytoskeleton, axonal transport and neuronal activity were assessed. SNCA dupl CNs displayed an increased IL-17A receptor expression and impaired IL-17A-mediated cytokine receptor regulation. Cytokines exacerbated the altered distribution of tubulin post-translational modifications in SNCA dupl neurites, with SNCA dupl-specific IL-17A effects. Tau pathology in SNCA dupl CNs was also aggravated by IL-17A and cytokine mix. Cytokines slowed down mitochondrial axonal transport, with IL-17A-mediated retrograde slowing in SNCA dupl only. The pre-treatment of SNCA dupl CNs with NPT100-18A prevented the IL-17A-induced functional impairments in axonal transport and neural activity. Our work elucidates the detrimental effects of pro-inflammatory cytokines, particularly IL-17A, on human neuronal structure and function in the context of α-syn pathology, suggesting that cytokine-mediated inflammation represents a second hit to neurons in PD which is amenable to disease modifying therapies that are currently in clinical trials.
Collapse
Affiliation(s)
- Veronika Sigutova
- Department of Stem Cell Biology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany; Dental Clinic 1, Department of Operative Dentistry and Periodontology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Regensburger
- Department of Stem Cell Biology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany; Department of Molecular Neurology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Erlangen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Erlangen, Germany; Center for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Iryna Prots
- Department of Stem Cell Biology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany; Dental Clinic 1, Department of Operative Dentistry and Periodontology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
2
|
Grabowska AD, Wątroba M, Witkowska J, Mikulska A, Sepúlveda N, Szukiewicz D. Interplay between Systemic Glycemia and Neuroprotective Activity of Resveratrol in Modulating Astrocyte SIRT1 Response to Neuroinflammation. Int J Mol Sci 2023; 24:11640. [PMID: 37511397 PMCID: PMC10380505 DOI: 10.3390/ijms241411640] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
The flow of substances between the blood and the central nervous system is precisely regulated by the blood-brain barrier (BBB). Its disruption due to unbalanced blood glucose levels (hyper- and hypoglycemia) occurring in metabolic disorders, such as type 2 diabetes, can lead to neuroinflammation, and increase the risk of developing neurodegenerative diseases. One of the most studied natural anti-diabetic, anti-inflammatory, and neuroprotective compounds is resveratrol (RSV). It activates sirtuin 1 (SIRT1), a key metabolism regulator dependent on cell energy status. The aim of this study was to assess the astrocyte SIRT1 response to neuroinflammation and subsequent RSV treatment, depending on systemic glycemia. For this purpose, we used an optimized in vitro model of the BBB consisting of endothelial cells and astrocytes, representing microvascular and brain compartments (MC and BC), in different glycemic backgrounds. Astrocyte-secreted SIRT1 reached the highest concentration in hypo-, the lowest in normo-, and the lowest in hyperglycemic backgrounds. Lipopolysaccharide (LPS)-induced neuroinflammation caused a substantial decrease in SIRT1 in all glycemic backgrounds, as observed earliest in hyperglycemia. RSV partially counterbalanced the effect of LPS on SIRT1 secretion, most remarkably in normoglycemia. Our results suggest that abnormal glycemic states have a worse prognosis for RSV-therapy effectiveness compared to normoglycemia.
Collapse
Affiliation(s)
- Anna D. Grabowska
- Laboratory of the Blood-Brain Barrier, Department of Biophysics, Physiology and Pathophysiology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (M.W.); (J.W.); (A.M.); (D.S.)
| | - Mateusz Wątroba
- Laboratory of the Blood-Brain Barrier, Department of Biophysics, Physiology and Pathophysiology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (M.W.); (J.W.); (A.M.); (D.S.)
| | - Joanna Witkowska
- Laboratory of the Blood-Brain Barrier, Department of Biophysics, Physiology and Pathophysiology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (M.W.); (J.W.); (A.M.); (D.S.)
| | - Agnieszka Mikulska
- Laboratory of the Blood-Brain Barrier, Department of Biophysics, Physiology and Pathophysiology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (M.W.); (J.W.); (A.M.); (D.S.)
| | - Nuno Sepúlveda
- Faculty of Mathematics and Information Science, Warsaw University of Technology, Koszykowa 75, 00-662 Warsaw, Poland
- CEAUL—Centro de Estatística e Aplicações da Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Dariusz Szukiewicz
- Laboratory of the Blood-Brain Barrier, Department of Biophysics, Physiology and Pathophysiology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (M.W.); (J.W.); (A.M.); (D.S.)
| |
Collapse
|
3
|
Tran VL, Liu P, Katsumura KR, Kim E, Schoff BM, Johnson KD, Bresnick EH. Restricting genomic actions of innate immune mediators on fetal hematopoietic progenitor cells. iScience 2023; 26:106297. [PMID: 36950124 PMCID: PMC10025987 DOI: 10.1016/j.isci.2023.106297] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/03/2023] [Accepted: 02/24/2023] [Indexed: 03/15/2023] Open
Abstract
Innate immune signaling protects against pathogens, controls hematopoietic development, and functions in oncogenesis, yet the relationship between these mechanisms is undefined. Downregulating the GATA2 transcription factor in fetal hematopoietic progenitor cells upregulates genes encoding innate immune regulators, increases Interferon-γ (IFNγ) signaling, and disrupts differentiation. We demonstrate that deletion of an enhancer that confers GATA2 expression in fetal progenitors elevated Toll-like receptor (TLR) TLR1/2 and TLR2/6 expression and signaling. Rescue by expressing GATA2 downregulated elevated TLR signaling. IFNγ amplified TLR1/2 and TLR2/6 signaling in GATA2-deficient progenitors, synergistically activating cytokine/chemokine genes and elevating cytokine/chemokine production in myeloid cell progeny. Genomic analysis of how innate immune signaling remodels the GATA2-deficient progenitor transcriptome revealed hypersensitive responses at innate immune genes harboring motifs for signal-dependent transcription factors and factors not linked to these mechanisms. As GATA2 establishes a transcriptome that constrains innate immune signaling, insufficient GATA2 renders fetal progenitor cells hypersensitive to innate immune signaling.
Collapse
Affiliation(s)
- Vu L. Tran
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Peng Liu
- Department of Biostatistics and Biomedical Informatics, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Koichi R. Katsumura
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Erin Kim
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Bjorn M. Schoff
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Kirby D. Johnson
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Emery H. Bresnick
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
4
|
Mobley JA, Molyvdas A, Kojima K, Ahmad I, Jilling T, Li JL, Garantziotis S, Matalon S. The SARS-CoV-2 spike S1 protein induces global proteomic changes in ATII-like rat L2 cells that are attenuated by hyaluronan. Am J Physiol Lung Cell Mol Physiol 2023; 324:L413-L432. [PMID: 36719087 PMCID: PMC10042596 DOI: 10.1152/ajplung.00282.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/29/2022] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
The COVID-19 pandemic continues to impose a major impact on global health and economy since its identification in early 2020, causing significant morbidity and mortality worldwide. Caused by the SARS-CoV-2 virus, along with a growing number of variants, COVID-19 has led to 651,918,402 confirmed cases and 6,656,601 deaths worldwide (as of December 27, 2022; https://covid19.who.int/). Despite advances in our understanding of COVID-19 pathogenesis, the precise mechanism by which SARS-CoV2 causes epithelial injury is incompletely understood. In this current study, robust application of global-discovery proteomics identified highly significant induced changes by the Spike S1 protein of SARS-CoV-2 in the proteome of alveolar type II (ATII)-like rat L2 cells that lack ACE2 receptors. Systems biology analysis revealed that the S1-induced proteomics changes were associated with three significant network hubs: E2F1, CREB1/RelA, and ROCK2/RhoA. We also found that pretreatment of L2 cells with high molecular weight hyaluronan (HMW-HA) greatly attenuated the S1 effects on the proteome. Western blotting analysis and cell cycle measurements confirmed the S1 upregulation of E2F1 and ROCK2/RhoA in L2 cells and the protective effects of HMW-HA. Taken as a whole, our studies revealed profound and novel biological changes that contribute to our current understanding of both S1 and hyaluronan biology. These data show that the S1 protein may contribute to epithelial injury induced by SARS-CoV-2. In addition, our work supports the potential benefit of HMW-HA in ameliorating SARS CoV-2-induced cell injury.
Collapse
Affiliation(s)
- James A Mobley
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Adam Molyvdas
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Kyoko Kojima
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Israr Ahmad
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Tamas Jilling
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Jian-Liang Li
- National Institute of Environmental Health Sciences, Durham, North Carolina, United States
| | - Stavros Garantziotis
- National Institute of Environmental Health Sciences, Durham, North Carolina, United States
| | - Sadis Matalon
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
5
|
Hernandez JC, Chen CL, Machida T, Uthaya Kumar DB, Tahara SM, Montana J, Sher L, Liang J, Jung JU, Tsukamoto H, Machida K. LIN28 and histone H3K4 methylase induce TLR4 to generate tumor-initiating stem-like cells. iScience 2023; 26:106254. [PMID: 36949755 PMCID: PMC10025994 DOI: 10.1016/j.isci.2023.106254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/09/2022] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Chemoresistance and plasticity of tumor-initiating stem-like cells (TICs) promote tumor recurrence and metastasis. The gut-originating endotoxin-TLR4-NANOG oncogenic axis is responsible for the genesis of TICs. This study investigated mechanisms as to how TICs arise through transcriptional, epigenetic, and post-transcriptional activation of oncogenic TLR4 pathways. Here, we expressed constitutively active TLR4 (caTLR4) in mice carrying pLAP-tTA or pAlb-tTA, under a tetracycline withdrawal-inducible system. Liver progenitor cell induction accelerated liver tumor development in caTLR4-expressing mice. Lentiviral shRNA library screening identified histone H3K4 methylase SETD7 as central to activation of TLR4. SETD7 combined with hypoxia induced TLR4 through HIF2 and NOTCH. LIN28 post-transcriptionally stabilized TLR4 mRNA via de-repression of let-7 microRNA. These results supported a LIN28-TLR4 pathway for the development of HCCs in a hypoxic microenvironment. These findings not only advance our understanding of molecular mechanisms responsible for TIC generation in HCC, but also represent new therapeutic targets for the treatment of HCC.
Collapse
Affiliation(s)
- Juan Carlos Hernandez
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
- MS Biotechnology Program, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Chia-Lin Chen
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
- Department of Life Sciences & Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 110, Taiwan
| | - Tatsuya Machida
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Dinesh Babu Uthaya Kumar
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Stanley M. Tahara
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Jared Montana
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Linda Sher
- Department of Surgery, University of Southern California, Los Angeles, CA 90033, USA
| | | | - Jae U. Jung
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Hidekazu Tsukamoto
- Department of Pathology, University of Southern California, Los Angeles, CA 90033, USA
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA
- Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Keigo Machida
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA
| |
Collapse
|
6
|
Pan W, Meshcheryakov VA, Li T, Wang Y, Ghosh G, Wang VYF. Structures of NF-κB p52 homodimer-DNA complexes rationalize binding mechanisms and transcription activation. eLife 2023; 12:e86258. [PMID: 36779700 PMCID: PMC9991059 DOI: 10.7554/elife.86258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/07/2023] [Indexed: 02/14/2023] Open
Abstract
The mammalian NF-κB p52:p52 homodimer together with its cofactor Bcl3 activates transcription of κB sites with a central G/C base pair (bp), while it is inactive toward κB sites with a central A/T bp. To understand the molecular basis for this unique property of p52, we have determined the crystal structures of recombinant human p52 protein in complex with a P-selectin(PSel)-κB DNA (5'-GGGGTGACCCC-3') (central bp is underlined) and variants changing the central bp to A/T or swapping the flanking bp. The structures reveal a nearly two-fold widened minor groove in the central region of the DNA as compared to all other currently available NF-κB-DNA complex structures, which have a central A/T bp. Microsecond molecular dynamics (MD) simulations of free DNAs and p52 bound complexes reveal that free DNAs exhibit distinct preferred conformations, and p52:p52 homodimer induces the least amount of DNA conformational changes when bound to the more transcriptionally active natural G/C-centric PSel-κB, but adopts closed conformation when bound to the mutant A/T and swap DNAs due to their narrowed minor grooves. Our binding assays further demonstrate that the fast kinetics favored by entropy is correlated with higher transcriptional activity. Overall, our studies have revealed a novel conformation for κB DNA in complex with NF-κB and pinpoint the importance of binding kinetics, dictated by DNA conformational and dynamic states, in controlling transcriptional activation for NF-κB.
Collapse
Affiliation(s)
- Wenfei Pan
- Faculty of Health Sciences, University of MacauTaipaChina
| | | | - Tianjie Li
- Department of Physics, Chinese University of Hong KongShatinHong Kong
| | - Yi Wang
- Department of Physics, Chinese University of Hong KongShatinHong Kong
| | - Gourisankar Ghosh
- Department of Chemistry and Biochemistry, University of California, San DiegoLa JollaUnited States
| | - Vivien Ya-Fan Wang
- Faculty of Health Sciences, University of MacauTaipaChina
- MoE Frontiers Science Center for Precision Oncology, University of MacauTaipaMacao
- Cancer Centre, Faculty of Health Sciences, University of MacauTaipaChina
| |
Collapse
|
7
|
Song Z, Liu N, He Y, Chen J, Li J, Wang F, Wu Z. Knockout of ICAT in Adipose Tissue Alleviates Fibro-inflammation in Obese Mice. Inflammation 2023; 46:404-417. [PMID: 36181623 DOI: 10.1007/s10753-022-01742-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022]
Abstract
The E2 promoter binding factor 1 (E2F1) and the Wnt/β-catenin signaling are crucial in regulating metabolic homeostasis including obesity. The β-catenin interacting protein 1 (CTNNBIP1), also known as the inhibitor of β-catenin and TCF4 (ICAT), is required for E2F1 to inhibit the activity of β-catenin. However, the role of ICAT in E2F1 regulating obesity-related metabolic disorders remains unknown. In the present study, male adipose tissue-specific ICAT knockout (ICATadi-/-) C57BL/6 J mice and control littermates aged 6-8 weeks were fed with high-fat diet (HFD) for 12 weeks to explore the effect of ICAT on lipid metabolism and obesity-related disorders. Results showed that the adipose tissue-specific ICAT knockout had negligible effect on lipid metabolism, reflected by no difference in body weight, fat mass, and the expression of proteins involved in lipid metabolism in white adipose tissue (WAT) and the liver between the ICATadi-/- mice and the control littermate (ICATfl/fl) mice. However, the knockout of ICAT reduced inflammatory response in WAT and the liver. Additionally, Sirius red staining results showed that deletion of ICAT attenuated fibrosis and reduced mRNA levels of transforming growth factor β1(TGF-β1), matrix metallopeptidase 2 (Mmp2), Mmp3, and collagen, type V, alpha 1 (Col5a1) in WAT and the liver. These results suggested that knockout of ICAT improved the metabolic abnormalities of obese mice through attenuating adipose tissue and the liver inflammation as well as fibrosis. Our findings may provide a new insight to understand the role of ICAT in inflammation and fibrosis.
Collapse
Affiliation(s)
- Zhuan Song
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Ning Liu
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, China
| | - Yu He
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Jingqing Chen
- Laboratory Animal Center of the Academy of Military Medical Sciences, Beijing, 100193, China
| | - Jun Li
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Fengchao Wang
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Zhenlong Wu
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China. .,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
8
|
Masyutina AM, Maximchik PV, Chkadua GZ, Pashenkov MV. Inhibition of specific signaling pathways rather than epigenetic silencing of effector genes is the leading mechanism of innate tolerance. Front Immunol 2023; 14:1006002. [PMID: 36776861 PMCID: PMC9909295 DOI: 10.3389/fimmu.2023.1006002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Introduction Macrophages activated through a pattern-recognition receptor (PRR) enter a transient state of tolerance characterized by diminished responsiveness to restimulation of the same receptor. Signaling-based and epigenetic mechanisms are invoked to explain this innate tolerance. However, these two groups of mechanisms should result in different outcomes. The epigenetic scenario (silencing of effector genes) predicts that activation of a PRR should broadly cross-tolerize to agonists of unrelated PRRs, whereas in the signaling-based scenario (inhibition of signaling pathways downstream of specific PRRs), cross-tolerization should occur only between agonists utilizing the same PRR and/or signaling pathway. Also, the so-called non-tolerizeable genes have been described, which acquire distinct epigenetic marks and increased responsiveness to rechallenge with the same agonist. The existence of such genes is well explained by epigenetic mechanisms but difficult to explain solely by signaling mechanisms. Methods To evaluate contribution of signaling and epigenetic mechanisms to innate tolerance, we tolerized human macrophages with agonists of TLR4 or NOD1 receptors, which signal via distinct pathways, and assessed responses of tolerized cells to homologous restimulation and to cross-stimulation using different signaling, metabolic and transcriptomic read-outs. We developed a transcriptomics-based approach to distinguish responses to secondary stimulation from continuing responses to primary stimulation. Results We found that macrophages tolerized with a NOD1 agonist lack responses to homologous restimulation, whereas LPS-tolerized macrophages partially retain the ability to activate NF-κB pathway upon LPS rechallenge, which allows to sustain low-level expression of a subset of pro-inflammatory genes. Contributing to LPS tolerance is blockade of signaling pathways required for IFN-β production, resulting in 'pseudo-tolerization' of IFN-regulated genes. Many genes in NOD1- or TLR4-tolerized macrophages are upregulated as the result of primary stimulation (due to continuing transcription and/or high mRNA stability), but do not respond to homologous restimulation. Hyperresponsiveness of genes to homologous rechallenge is a rare and inconsistent phenomenon. However, most genes that have become unresponsive to homologous stimuli show unchanged or elevated responses to agonists of PRRs signaling via distinct pathways. Discussion Thus, inhibition of specific signaling pathways rather than epigenetic silencing is the dominant mechanism of innate tolerance.
Collapse
Affiliation(s)
- Anna M. Masyutina
- Laboratory of Clinical Immunology, National Research Center “Institute of Immunology” of the Federal Medical-Biological Agency of Russia, Moscow, Russia,Biological Faculty, Lomonosov Moscow State University, Moscow, Russia
| | - Polina V. Maximchik
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Georgy Z. Chkadua
- Laboratory of experimental diagnostics and biotherapy of tumors, N.N.Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Mikhail V. Pashenkov
- Laboratory of Clinical Immunology, National Research Center “Institute of Immunology” of the Federal Medical-Biological Agency of Russia, Moscow, Russia,*Correspondence: Mikhail V. Pashenkov,
| |
Collapse
|
9
|
An Expanded Interplay Network between NF-κB p65 (RelA) and E2F1 Transcription Factors: Roles in Physiology and Pathology. Cancers (Basel) 2022; 14:cancers14205047. [PMID: 36291831 PMCID: PMC9600032 DOI: 10.3390/cancers14205047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/30/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
Transcription Factors (TFs) are the main regulators of gene expression, controlling among others cell homeostasis, identity, and fate. TFs may either act synergistically or antagonistically on nearby regulatory elements and their interplay may activate or repress gene expression. The family of NF-κB TFs is among the most important TFs in the regulation of inflammation, immunity, and stress-like responses, while they also control cell growth and survival, and are involved in inflammatory diseases and cancer. The family of E2F TFs are major regulators of cell cycle progression in most cell types. Several studies have suggested the interplay between these two TFs in the regulation of numerous genes controlling several biological processes. In the present study, we compared the genomic binding landscape of NF-κB RelA/p65 subunit and E2F1 TFs, based on high throughput ChIP-seq and RNA-seq data in different cell types. We confirmed that RelA/p65 has a binding profile with a high preference for distal enhancers bearing active chromatin marks which is distinct to that of E2F1, which mostly generates promoter-specific binding. Moreover, the RelA/p65 subunit and E2F1 cistromes have limited overlap and tend to bind chromatin that is in an active state even prior to immunogenic stimulation. Finally, we found that a fraction of the E2F1 cistrome is recruited by NF-κΒ near pro-inflammatory genes following LPS stimulation in immune cell types.
Collapse
|
10
|
Baughman HER, Narang D, Chen W, Villagrán Suárez AC, Lee J, Bachochin MJ, Gunther TR, Wolynes PG, Komives EA. An intrinsically disordered transcription activation domain increases the DNA binding affinity and reduces the specificity of NFκB p50/RelA. J Biol Chem 2022; 298:102349. [PMID: 35934050 PMCID: PMC9440430 DOI: 10.1016/j.jbc.2022.102349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/03/2022] Open
Abstract
Many transcription factors contain intrinsically disordered transcription activation domains (TADs), which mediate interactions with coactivators to activate transcription. Historically, DNA-binding domains and TADs have been considered as modular units, but recent studies have shown that TADs can influence DNA binding. Whether these results can be generalized to more TADs is not clear. Here, we biophysically characterized the NFκB p50/RelA heterodimer including the RelA TAD and investigated the TAD's influence on NFκB-DNA interactions. In solution, we show the RelA TAD is disordered but compact, with helical tendency in two regions that interact with coactivators. We determined that the presence of the TAD increased the stoichiometry of NFκB-DNA complexes containing promoter DNA sequences with tandem κB recognition motifs by promoting the binding of NFκB dimers in excess of the number of κB sites. In addition, we measured the binding affinity of p50/RelA for DNA containing tandem κB sites and single κB sites. While the presence of the TAD enhanced the binding affinity of p50/RelA for all κB sequences tested, it also increased the affinity for nonspecific DNA sequences by over 10-fold, leading to an overall decrease in specificity for κB DNA sequences. In contrast, previous studies have generally reported that TADs decrease DNA-binding affinity and increase sequence specificity. Our results reveal a novel function of the RelA TAD in promoting binding to nonconsensus DNA, which sheds light on previous observations of extensive nonconsensus DNA binding by NFκB in vivo in response to strong inflammatory signals.
Collapse
Affiliation(s)
- Hannah E R Baughman
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, USA
| | - Dominic Narang
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, USA
| | - Wei Chen
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, USA
| | - Amalia C Villagrán Suárez
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, USA
| | - Joan Lee
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, USA
| | - Maxwell J Bachochin
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, USA
| | - Tristan R Gunther
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, USA
| | - Peter G Wolynes
- Department of Chemistry and Center for Theoretical Biological Physics, Rice University, Houston, Texas, USA
| | - Elizabeth A Komives
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, USA.
| |
Collapse
|
11
|
Mobley JA, Molyvdas A, Kojima K, Jilling T, Li JL, Garantziotis S, Matalon S. The SARS-CoV-2 Spike S1 Protein Induces Global Proteomic Changes in ATII-Like Rat L2 Cells that are Attenuated by Hyaluronan. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.08.31.506023. [PMID: 36093347 PMCID: PMC9460966 DOI: 10.1101/2022.08.31.506023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The COVID-19 pandemic continues to impose a major impact on global health and economy since its identification in early 2020, causing significant morbidity and mortality worldwide. Caused by the SARS-CoV-2 virus, along with a growing number of variants that have been characterized to date, COVID-19 has led to 571,198,904 confirmed cases, and 6,387,863 deaths worldwide (as of July 15 th , 2022). Despite tremendous advances in our understanding of COVID19 pathogenesis, the precise mechanism by which SARS-CoV2 causes epithelial injury is incompletely understood. In this current study, robust application of global-discovery proteomics applications combined with systems biology analysis identified highly significant induced changes by the Spike S1 protein of SARS-CoV-2 in an ATII-like Rat L2 cells that include three significant network hubs: E2F1, CREB1/ RelA, and ROCK2/ RhoA. Separately, we found that pre-treatment with High Molecular Weight Hyaluronan (HMW-HA), greatly attenuated the S1 effects. Immuno-targeted studies carried out on E2F1 and Rock2/ RhoA induction and kinase-mediated activation, in addition to cell cycle measurements, validated these observations. Taken as a whole, our discovery proteomics and systems analysis workflow, combined with standard immuno-targeted and cell cycle measurements revealed profound and novel biological changes that contribute to our current understanding of both Spike S1 and Hyaluronan biology. This data shows that the Spike S1 protein may contribute to epithelial injury induced by SARS-CoV-2. In addition, our work supports the potential benefit of HMW-HA in ameliorating SARS CoV2 induced cell injury.
Collapse
|
12
|
Budke B, Zhong A, Sullivan K, Park C, Gittin DI, Kountz TS, Connell PP. Noncanonical NF-κB factor p100/p52 regulates homologous recombination and modulates sensitivity to DNA-damaging therapy. Nucleic Acids Res 2022; 50:6251-6263. [PMID: 35689636 PMCID: PMC9226503 DOI: 10.1093/nar/gkac491] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/17/2022] [Accepted: 05/25/2022] [Indexed: 11/14/2022] Open
Abstract
Homologous recombination (HR) serves multiple roles in DNA repair that are essential for maintaining genomic stability, including double-strand DNA break (DSB) repair. The central HR protein, RAD51, is frequently overexpressed in human malignancies, thereby elevating HR proficiency and promoting resistance to DNA-damaging therapies. Here, we find that the non-canonical NF-κB factors p100/52, but not RelB, control the expression of RAD51 in various human cancer subtypes. While p100/p52 depletion inhibits HR function in human tumor cells, it does not significantly influence the proficiency of non-homologous end joining, the other key mechanism of DSB repair. Clonogenic survival assays were performed using a pair DLD-1 cell lines that differ only in their expression of the key HR protein BRCA2. Targeted silencing of p100/p52 sensitizes the HR-competent cells to camptothecin, while sensitization is absent in HR-deficient control cells. These results suggest that p100/p52-dependent signaling specifically controls HR activity in cancer cells. Since non-canonical NF-κB signaling is known to be activated after various forms of genomic crisis, compensatory HR upregulation may represent a natural consequence of DNA damage. We propose that p100/p52-dependent signaling represents a promising oncologic target in combination with DNA-damaging treatments.
Collapse
Affiliation(s)
- Brian Budke
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Alison Zhong
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Katherine Sullivan
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Chanyoung Park
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - David I Gittin
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Timothy S Kountz
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Philip P Connell
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
13
|
Jones K, Ramirez-Perez S, Niu S, Gangishetti U, Drissi H, Bhattaram P. SOX4 and RELA Function as Transcriptional Partners to Regulate the Expression of TNF- Responsive Genes in Fibroblast-Like Synoviocytes. Front Immunol 2022; 13:789349. [PMID: 35529852 PMCID: PMC9074688 DOI: 10.3389/fimmu.2022.789349] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
SOX4 belongs to the group C of the SOX transcription factor family. It is a critical mediator of tumor necrosis factor alpha (TNF)-induced transformation of fibroblast-like s-ynoviocytes (FLS) in arthritis. In this study we investigated the genome wide association between the DNA binding and transcriptional activities of SOX4 and the NF-kappaB signaling transcription factor RELA/p65 downstream of TNF signaling. We used ChIP-seq assays in mouse FLS to compare the global DNA binding profiles of SOX4 and RELA. RNA-seq of TNF-induced wildtype and SoxC-knockout FLS was used to identify the SOX4-dependent and independent aspects of the TNF-regulated transcriptome. We found that SOX4 and RELA physically interact with each other on the chromatin. Interestingly, ChIP-seq assays revealed that 70.4% of SOX4 peak summits were within 50bp of the RELA peak summits suggesting that both proteins bind in close-proximity on regulatory sequences, enabling them to co-operatively regulate gene expression. By integrating the ChIP-seq results with RNA-seq from SoxC-knockout FLS we identified a set of TNF-responsive genes that are targets of the RELA-SOX4 transcriptional complex. These TNF-responsive and RELA-SOX4-depenedent genes included inflammation mediators, histone remodeling enzymes and components of the AP-1 signaling pathway. We also identified an autoregulatory mode of SoxC gene expression that involves a TNF-mediated switch from RELA binding to SOX4 binding in the 3' UTR of Sox4 and Sox11 genes. In conclusion, our results show that SOX4 and RELA together orchestrate a multimodal regulation of gene expression downstream of TNF signaling. Their interdependent activities play a pivotal role in the transformation of FLS in arthritis and in the inflammatory pathology of diverse tissues where RELA and SOX4 are co-expressed.
Collapse
Affiliation(s)
- Kyle Jones
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, United States
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Sergio Ramirez-Perez
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, United States
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Sean Niu
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, United States
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Umesh Gangishetti
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, United States
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, United States
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Veterans Affairs, Atlanta VA Medical Center, Decatur, GA, United States
| | - Pallavi Bhattaram
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, United States
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
14
|
Cai J, Wu J, Fang S, Liu S, Wang T, Li Y, Zou J, Shi R, Wang Z, Yang L, Ma Y. Cultured bear bile powder ameliorates acute liver injury in cholestatic mice via inhibition of hepatic inflammation and apoptosis. JOURNAL OF ETHNOPHARMACOLOGY 2022; 284:114829. [PMID: 34763041 DOI: 10.1016/j.jep.2021.114829] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/23/2021] [Accepted: 11/06/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Natural bear bile powder (NBBP) is a traditional Chinese medicine used for treating liver dysfunction. Cultured bear bile powder (CBBP), which is produced using biotransformation of chicken bile, acts as an appropriate substitute for NBBP when treating cholestatic liver injury. AIM OF THE STUDY To investigate the molecular mechanisms underlying the hepatoprotective effects of CBBP in an α-naphthylisothiocyanate (ANIT)-induced cholestatic mouse model. MATERIALS AND METHODS Cholestatic mice were pretreated with CBBP or NBBP via oral gavage once a day for two weeks. Their blood biochemistry and liver histopathology were then evaluated using standard protocols. Western blot analyses, real-time polymerase chain reaction, and immunohistochemistry were used to evaluate changes in the protein levels and gene expression profiles of factors associated with hepatic inflammation and apoptosis in cholestatic mice. RESULTS CBBP significantly decreased the serum indices of liver injury, and ameliorated neutrophil infiltration and hepatocyte necrosis within liver tissue of cholestatic mice. Expression of the inflammatory factors, such as tumor necrosis factor-α, interleukin-1β (IL-1β), IL-6, monocyte chemoattractant protein-1, and intercellular adhesion molecule 1, was significantly reduced in CBBP-treated cholestatic mice. Moreover, proteins involved in the toll-like receptor 4/myeloid differentiation factor 88/nuclear factor-kappa B (TLR4/Myd88/NF-κB) signaling pathway, such as CD14, TLR4, Myd88, and NF-κB, that were increased in cholestatic mice, were downregulated by CBBP. Meanwhile, increased expression of the apoptosis-related proteins, caspase-3 and Bax, in cholestatic mice was reversed by CBBP treatment. CONCLUSION CBBP treatment alleviates ANIT-induced cholestasis and liver injury by reducing hepatocyte inflammation and apoptosis.
Collapse
Affiliation(s)
- Jingyi Cai
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jiasheng Wu
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Su Fang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shaoyong Liu
- Shanghai Kai Bao Pharmaceutical CO. Ltd., Shanghai, 201401, China
| | - Tianming Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuanyuan Li
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Juan Zou
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Rong Shi
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhengtao Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li Yang
- Center for Traditional Chinese Medicine of Complexity Systems, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yueming Ma
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Key Laboratory of Compound Chinese Medicines, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
15
|
Mejhert N, Gabriel KR, Frendo-Cumbo S, Krahmer N, Song J, Kuruvilla L, Chitraju C, Boland S, Jang DK, von Grotthuss M, Costanzo MC, Rydén M, Olzmann JA, Flannick J, Burtt NP, Farese RV, Walther TC. The Lipid Droplet Knowledge Portal: A resource for systematic analyses of lipid droplet biology. Dev Cell 2022; 57:387-397.e4. [PMID: 35134345 PMCID: PMC9129885 DOI: 10.1016/j.devcel.2022.01.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/16/2021] [Accepted: 12/31/2021] [Indexed: 12/15/2022]
Abstract
Lipid droplets (LDs) are organelles of cellular lipid storage with fundamental roles in energy metabolism and cell membrane homeostasis. There has been an explosion of research into the biology of LDs, in part due to their relevance in diseases of lipid storage, such as atherosclerosis, obesity, type 2 diabetes, and hepatic steatosis. Consequently, there is an increasing need for a resource that combines datasets from systematic analyses of LD biology. Here, we integrate high-confidence, systematically generated human, mouse, and fly data from studies on LDs in the framework of an online platform named the "Lipid Droplet Knowledge Portal" (https://lipiddroplet.org/). This scalable and interactive portal includes comprehensive datasets, across a variety of cell types, for LD biology, including transcriptional profiles of induced lipid storage, organellar proteomics, genome-wide screen phenotypes, and ties to human genetics. This resource is a powerful platform that can be utilized to identify determinants of lipid storage.
Collapse
Affiliation(s)
- Niklas Mejhert
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Department of Medicine (H7), Karolinska Institutet, Huddinge, 141 86 Stockholm, Sweden
| | - Katlyn R Gabriel
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA
| | - Scott Frendo-Cumbo
- Department of Medicine (H7), Karolinska Institutet, Huddinge, 141 86 Stockholm, Sweden
| | - Natalie Krahmer
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, 85764 Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany
| | - Jiunn Song
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Leena Kuruvilla
- Primary Pharmacology Group, Discovery Sciences, Pfizer Inc., Groton, CT 06340, USA
| | - Chandramohan Chitraju
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sebastian Boland
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Dong-Keun Jang
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Marcin von Grotthuss
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Maria C Costanzo
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, Huddinge, 141 86 Stockholm, Sweden
| | - James A Olzmann
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, CA 94720, USA; Miller Institute for Basic Research in Science, University of California Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Jason Flannick
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Noël P Burtt
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Robert V Farese
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center on the Causes and Prevention of Cardiovascular Disease (CAP-CVD), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| | - Tobias C Walther
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center on the Causes and Prevention of Cardiovascular Disease (CAP-CVD), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
16
|
Chen Y, Gong W, Dai W, Jiang H, Xu X. E2F1/2/4 mRNA is associated with immune infiltration and are potential biomarkers for the prognosis of human gastric carcinoma. Transl Cancer Res 2022; 10:2801-2811. [PMID: 35116590 PMCID: PMC8797903 DOI: 10.21037/tcr-21-45] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/27/2021] [Indexed: 12/19/2022]
Abstract
Background E2Fs are genes that regulate DNA synthesis and the cell cycle by encoding a family of transcription factors. Increasing experimental evidence has revealed that E2Fs play key roles in tumor progression in various types of cancer. Methods We investigated the survival, expression and transcriptional data of E2F1/2/4 in gastric cancer (GC) patients using the immunohistochemistry assay, Kaplan-Meier Plotter, cBioPortal, String, and GEPIA databases. The plasma of GC patients was analyzed using the real-time reverse transcription polymerase chain reaction (RT-PCR) assay. The correlation between E2F1/2/4 expression and clinical features was analyzed using the quartile method. As well, the correlation between E2F1/2/4 and GC immune infiltration was also investigated using the TIMER database. Database of Immune Cell Expression (DICE) was also used to analyze correlations between SOX4 and immune responses. Results RT-PCR and tissue immunohistochemistry confirmed that E2F1/2/4 was highly expressed in serum and GC tissue samples of GC patients, the expression of which was not affected by patient age and gender. Also, the survival analysis revealed that low levels of E2F1/2/4 expression were significantly associated with a longer overall survival (OS) in GC patients. E2F1/2/4 was correlated with patient prognosis and immune cell infiltration, including B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils, and DCs in GC. Our findings indicated that E2F1/2/4 could be used as a prognostic biomarker and indicator of immune infiltration in GC. Conclusions This study revealed that E2F1/2/4 could be a promising indicator for tumor-associated immune infiltration and prognosis in GC patients.
Collapse
Affiliation(s)
- Yongyi Chen
- Department of Clinical Lab, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Wangang Gong
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China.,Zhejiang Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Wumin Dai
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China.,Zhejiang Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Huifen Jiang
- Department of Clinical Lab, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xiaohong Xu
- Department of Clinical Lab, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
17
|
Ghosh G, Wang VYF. Origin of the Functional Distinctiveness of NF-κB/p52. Front Cell Dev Biol 2021; 9:764164. [PMID: 34888310 PMCID: PMC8650618 DOI: 10.3389/fcell.2021.764164] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
The transcription regulators of the NF-κB family have emerged as a critical factor affecting the function of various adult tissues. The NF-κB family transcription factors are homo- and heterodimers made up of five monomers (p50, p52, RelA, cRel and RelB). The family is distinguished by sequence homology in their DNA binding and dimerization domains, which enables them to bind similar DNA response elements and participate in similar biological programs through transcriptional activation and repression of hundreds of genes. Even though the family members are closely related in terms of sequence and function, they all display distinct activities. In this review, we discuss the sequence characteristics, protein and DNA interactions, and pathogenic involvement of one member of family, NF-κB/p52, relative to that of other members. We pinpoint the small sequence variations within the conserved region that are mostly responsible for its distinct functional properties.
Collapse
Affiliation(s)
- Gourisankar Ghosh
- Department of Biochemistry, University of California, San Diego, San Diego, CA, United States
| | - Vivien Ya-Fan Wang
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.,Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| |
Collapse
|
18
|
Meier-Soelch J, Mayr-Buro C, Juli J, Leib L, Linne U, Dreute J, Papantonis A, Schmitz ML, Kracht M. Monitoring the Levels of Cellular NF-κB Activation States. Cancers (Basel) 2021; 13:5351. [PMID: 34771516 PMCID: PMC8582385 DOI: 10.3390/cancers13215351] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
The NF-κB signaling system plays an important regulatory role in the control of many biological processes. The activities of NF-κB signaling networks and the expression of their target genes are frequently elevated in pathophysiological situations including inflammation, infection, and cancer. In these conditions, the outcome of NF-κB activity can vary according to (i) differential activation states, (ii) the pattern of genomic recruitment of the NF-κB subunits, and (iii) cellular heterogeneity. Additionally, the cytosolic NF-κB activation steps leading to the liberation of DNA-binding dimers need to be distinguished from the less understood nuclear pathways that are ultimately responsible for NF-κB target gene specificity. This raises the need to more precisely determine the NF-κB activation status not only for the purpose of basic research, but also in (future) clinical applications. Here we review a compendium of different methods that have been developed to assess the NF-κB activation status in vitro and in vivo. We also discuss recent advances that allow the assessment of several NF-κB features simultaneously at the single cell level.
Collapse
Affiliation(s)
- Johanna Meier-Soelch
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392 Giessen, Germany; (J.M.-S.); (C.M.-B.); (J.J.); (L.L.)
| | - Christin Mayr-Buro
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392 Giessen, Germany; (J.M.-S.); (C.M.-B.); (J.J.); (L.L.)
| | - Jana Juli
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392 Giessen, Germany; (J.M.-S.); (C.M.-B.); (J.J.); (L.L.)
| | - Lisa Leib
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392 Giessen, Germany; (J.M.-S.); (C.M.-B.); (J.J.); (L.L.)
| | - Uwe Linne
- Mass Spectrometry Facility of the Department of Chemistry, Philipps University, 35032 Marburg, Germany;
| | - Jan Dreute
- Institute of Biochemistry, Justus Liebig University, 35392 Giessen, Germany;
| | - Argyris Papantonis
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany;
| | - M. Lienhard Schmitz
- Institute of Biochemistry, Justus Liebig University, 35392 Giessen, Germany;
| | - Michael Kracht
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392 Giessen, Germany; (J.M.-S.); (C.M.-B.); (J.J.); (L.L.)
| |
Collapse
|
19
|
Li B, Zhang L, Zhu L, Cao Y, Dou Z, Yu Q. HDAC5 promotes intestinal sepsis via the Ghrelin/E2F1/NF-κB axis. FASEB J 2021; 35:e21368. [PMID: 34125448 DOI: 10.1096/fj.202001584r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 12/20/2020] [Accepted: 12/28/2020] [Indexed: 01/24/2023]
Abstract
In the current study, we sought to determine the roles of histone deacetylase 5 (HDAC5) on the promotion of intestinal sepsis in a mouse model. Dual luciferase reporter gene assay was used to determine the binding relationship between HDAC5 and Ghrelin. Cecal ligation and puncture (CLP) was used as an animal model of intestinal sepsis. The roles of HDAC5 on intestinal sepsis were determined by HDAC5 knockdown, overexpression, and inhibitor (LMK-235) in vivo. Mice intestinal permeability and intestinal epithelial damage were evaluated, and HE staining was used to evaluate the intestinal mucosal injury index. Lipopolysaccharide (LPS)-treated intestinal-derived macrophages served as a cell model of sepsis, followed by the loss-of-function and gain-of-function assays. ELISA was used to determine the levels of inflammatory factors, and TUNEL staining was used to detect intestinal cell apoptosis. HDAC5 was upregulated in the intestine of sepsis patients. This increased HDAC5 expression was positively correlated with the expression of inflammatory factors TNF-α, IL-1β, IL-6, and HMGB1, as well as the intestinal dysfunction-related factors IFABP. In sepsis mice, the expression of inflammatory factors was reduced by HDAC5 knockdown. HDAC5 knockdown also improved survival, morphology of intestinal tissue, intestinal permeability, and epithelial damage. Ghrelin was bound and inhibited by HDAC5, but E2F1 expression was increased by Ghrelin overexpression, leading to inhibition of the NF-κB pathway. Ghrelin and E2F1 expression were increased by the treatment with HDAC5 inhibitor LMK-235, which inhibited the NF-κB pathway to improve intestinal dysfunction in the sepsis model. In conclusion, HDAC5 inhibits Ghrelin to reduce E2F1 and thus activate the NF-κB pathway, thereby promoting intestinal sepsis.
Collapse
Affiliation(s)
- Bin Li
- Department of Critical Medicine, the First Hospital of Lanzhou University, Lanzhou, P.R. China
| | - Lei Zhang
- Department of Critical Medicine, the First Hospital of Lanzhou University, Lanzhou, P.R. China
| | - Lei Zhu
- Department of Critical Medicine, the First Hospital of Lanzhou University, Lanzhou, P.R. China
| | - Yongqiang Cao
- Department of Critical Medicine, the First Hospital of Lanzhou University, Lanzhou, P.R. China
| | - Zhimin Dou
- Department of Critical Medicine, the First Hospital of Lanzhou University, Lanzhou, P.R. China
| | - Qin Yu
- Department of Respiratory, the First Hospital of Lanzhou University (the First School of Clinical Medicine), Lanzhou, P.R. China
| |
Collapse
|
20
|
Song M, He J, Pan QZ, Yang J, Zhao J, Zhang YJ, Huang Y, Tang Y, Wang Q, He J, Gu J, Li Y, Chen S, Zeng J, Zhou ZQ, Yang C, Han Y, Chen H, Xiang T, Weng DS, Xia JC. Cancer-Associated Fibroblast-Mediated Cellular Crosstalk Supports Hepatocellular Carcinoma Progression. Hepatology 2021; 73:1717-1735. [PMID: 33682185 DOI: 10.1002/hep.31792] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 01/14/2021] [Accepted: 02/01/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND AIMS Cancer-associated fibroblasts (CAFs) are key players in multicellular, stromal-dependent alterations leading to HCC pathogenesis. However, the intricate crosstalk between CAFs and other components in the tumor microenvironment (TME) remains unclear. This study aimed to investigate the cellular crosstalk among CAFs, tumor cells, and tumor-associated neutrophils (TANs) during different stages of HCC pathogenesis. APPROACH AND RESULTS In the HCC-TME, CAF-derived cardiotrophin-like cytokine factor 1 (CLCF1) increased chemokine (C-X-C motif) ligand 6 (CXCL6) and TGF-β secretion in tumor cells, which subsequently promoted tumor cell stemness in an autocrine manner and TAN infiltration and polarization in a paracrine manner. Moreover, CXCL6 and TGF-β secreted by HCC cells activated extracellular signal-regulated kinase (ERK) 1/2 signaling of CAFs to produce more CLCF1, thus forming a positive feedback loop to accelerate HCC progression. Inhibition of ERK1/2 or CLCF1/ciliary neurotrophic factor receptor signaling efficiently impaired CLCF1-mediated crosstalk among CAFs, tumor cells, and TANs both in vitro and in vivo. In clinical samples, up-regulation of the CLCF1-CXCL6/TGF-β axis exhibited a marked correlation with increased cancer stem cells, "N2"-polarized TANs, tumor stage, and poor prognosis. CONCLUSIONS This study reveals a cytokine-mediated cellular crosstalk and clinical network involving the CLCF1-CXCL6/TGF-β axis, which regulates the positive feedback loop among CAFs, tumor stemness, and TANs, HCC progression, and patient prognosis. These results may support the CLCF1 cascade as a potential prognostic biomarker and suggest that selective blockade of CLCF1/ciliary neurotrophic factor receptor or ERK1/2 signaling could provide an effective therapeutic target for patients with HCC.
Collapse
Affiliation(s)
- Mengjia Song
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Junyi He
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Qiu-Zhong Pan
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jieying Yang
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jingjing Zhao
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yao-Jun Zhang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yue Huang
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yan Tang
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Qijing Wang
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jia He
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jiamei Gu
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yongqiang Li
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Shiping Chen
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jianxiong Zeng
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Zi-Qi Zhou
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Chaopin Yang
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yulong Han
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Hao Chen
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Tong Xiang
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - De-Sheng Weng
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jian-Chuan Xia
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| |
Collapse
|
21
|
Regulation of Transcription Factor NF-κB in Its Natural Habitat: The Nucleus. Cells 2021; 10:cells10040753. [PMID: 33805563 PMCID: PMC8066257 DOI: 10.3390/cells10040753] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/24/2021] [Accepted: 03/24/2021] [Indexed: 01/11/2023] Open
Abstract
Activation of the transcription factor NF-κB elicits an individually tailored transcriptional response in order to meet the particular requirements of specific cell types, tissues, or organs. Control of the induction kinetics, amplitude, and termination of gene expression involves multiple layers of NF-κB regulation in the nucleus. Here we discuss some recent advances in our understanding of the mutual relations between NF-κB and chromatin regulators also in the context of different levels of genome organization. Changes in the 3D folding of the genome, as they occur during senescence or in cancer cells, can causally contribute to sustained increases in NF-κB activity. We also highlight the participation of NF-κB in the formation of hierarchically organized super enhancers, which enable the coordinated expression of co-regulated sets of NF-κB target genes. The identification of mechanisms allowing the specific regulation of NF-κB target gene clusters could potentially enable targeted therapeutic interventions, allowing selective interference with subsets of the NF-κB response without a complete inactivation of this key signaling system.
Collapse
|
22
|
Su H, Fan S, Zhang L, Qi H. TMAO Aggregates Neurological Damage Following Ischemic Stroke by Promoting Reactive Astrocytosis and Glial Scar Formation via the Smurf2/ALK5 Axis. Front Cell Neurosci 2021; 15:569424. [PMID: 33815059 PMCID: PMC8012716 DOI: 10.3389/fncel.2021.569424] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 01/06/2021] [Indexed: 11/15/2022] Open
Abstract
Ischemic stroke has been reported to cause significant changes to memory, thinking, and behavior. Intriguingly, recently reported studies have indicated the association of Trimethylamine N-oxide (TMAO) with the acute phase of ischemic stroke. However, the comprehensive underlying mechanism remained unknown. The objective of the present study was to investigate the association between TMAO and recovery of neurological function after ischemic stroke. For this purpose, a middle cerebral artery occlusion/reperfusion (MCAO/R) rat model was established and treated with TMAO or/and sh-ALK5, followed by the neurological function evaluation. Behaviors of rats were observed through staircase and cylinder tests. Moreover, the expression of Smurf2 and ALK5 was detected by immunohistochemistry while expression of GFAP, Neurocan, and Phosphacan in brain tissues was determined by immunofluorescence. Thereafter, gain- and loss-of-function assays in astrocytes, the proliferation, viability, and migration were evaluated by the EdU, CCK-8, and Transwell assays. Besides, Smurf2 mRNA expression was determined by the RT-qPCR, whereas, Smurf2, ALK5, GFAP, Neurocan, and Phosphacan expression was evaluated by the Western blotting. Finally, the interaction of Smurf2 with ALK5 and ALK5 ubiquitination was assessed by the co-immunoprecipitation. Notably, our results showed that TMAO promoted the proliferation of reactive astrocyte and formation of glial scar in MCAO/R rats. However, this effect was abolished by the Smurf2 overexpression or ALK5 silencing. We further found that TMAO upregulated the ALK5 expression by inhibiting the ubiquitination role of Smurf2. Overexpression of ALK5 reversed the inhibitory effect of Smurf2 on astrocyte proliferation, migration, and viability. Collectively, our work identifies the evolutionarily TMAO/Smurf2/ALK5 signaling as a major genetic factor in the control of reactive astrocyte proliferation and glial scar formation in ischemic stroke, thus laying a theoretical foundation for the identification of ischemic stroke.
Collapse
Affiliation(s)
- Haibo Su
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Shaoping Fan
- Department of Neurosurgery, The People's Hospital of Longhua District, Shenzhen, China
| | - Lingqiong Zhang
- Department of Hand Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Hui Qi
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
23
|
Wang X, Chen Y, Dong K, Ma Y, Jin Q, Yin S, Zhu X, Wang S. Effects of FER1L4-miR-106a-5p/miR-372-5p-E2F1 regulatory axis on drug resistance in liver cancer chemotherapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 24:449-461. [PMID: 33868788 PMCID: PMC8040129 DOI: 10.1016/j.omtn.2021.02.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 02/03/2021] [Indexed: 12/22/2022]
Abstract
Liver cancer presents a challenge in today's healthcare system. This study aimed at investigating the effects of Fer-1 like family member 4 (FER1L4) on chemotherapy resistance and liver cancer development by using clinically collected liver cancer tissues and commercially purchased human liver cancer cisplatin-resistant cell line HUH-7/DDP. Bioinformatics analysis, dual luciferase reporter gene assay, and RNA pull-down were applied to predict and verify the possible binding relationships. The expressions of FER1L4, E2F transcription factor 1 (E2F1), microRNA-106a-5p (miR-106a-5p), or miR-372-5p were altered in the cells, followed by flow cytometry, Cell Counting Kit-8 (CCK-8), and Transwell assays to evaluate apoptotic, proliferative, and invasive abilities in vitro and nude mice xenografts to observe tumor growth in vivo. FER1L4 was highly expressed and miR-106-5p and miR-372-5p were poorly expressed in tumor cells and tissues. FER1L4 knockdown or the overexpression of miR-106-5p and miR-372-5p inhibited the cancerous cell proliferation and invasion while promoting apoptosis. FERIL4 silencing increased the miR-106-5p/miR-372-5p expression to inhibit the E2F1-activated nuclear factor κB (NF-κB) pathway. Besides, overexpressing FER1L4 led to an increased tumor growth in nude mice, which was reversed by the NF-κB inhibitor pyrollidine dithiocarbamate (PDTC). In conclusion, the results indicated that FER1L4 could inhibit the expression of miR-106a-5p/miR-372-5p, to activate E2F1-mediated NF-κB pathway, leading to drug resistance in liver cancer.
Collapse
Affiliation(s)
- Xu Wang
- The Second Ward of Hepatobiliary Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China and Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, P.R. China
| | - Yunfei Chen
- Organ Transplant Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China and Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, P.R. China.,The Third Ward of Hepatobiliary Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China and Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, P.R. China
| | - Ke Dong
- The Second Ward of Hepatobiliary Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China and Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, P.R. China
| | - Yujing Ma
- The Second Ward of Hepatobiliary Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China and Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, P.R. China
| | - Qizhi Jin
- The Second Ward of Hepatobiliary Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China and Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, P.R. China
| | - Shujun Yin
- The Second Ward of Hepatobiliary Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China and Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, P.R. China
| | - Xiaoshi Zhu
- Pediatric Intensive Care Unit, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China and Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, P.R. China
| | - Shan Wang
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China and Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, P.R. China
| |
Collapse
|
24
|
Zarate MA, Nguyen LM, De Dios RK, Zheng L, Wright CJ. Maturation of the Acute Hepatic TLR4/NF-κB Mediated Innate Immune Response Is p65 Dependent in Mice. Front Immunol 2020; 11:1892. [PMID: 32973783 PMCID: PMC7472845 DOI: 10.3389/fimmu.2020.01892] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/14/2020] [Indexed: 12/27/2022] Open
Abstract
Compared to adults, neonates are at increased risk of infection. There is a growing recognition that dynamic qualitative and quantitative differences in immunity over development contribute to these observations. The liver plays a key role as an immunologic organ, but whether its contribution to the acute innate immune response changes over lifetime is unknown. We hypothesized that the liver would activate a developmentally-regulated acute innate immune response to intraperitoneal lipopolysaccharide (LPS). We first assessed the hepatic expression and activity of the NF-κB, a key regulator of the innate immune response, at different developmental ages (p0, p3, p7, p35, and adult). Ontogeny of the NF-κB subunits (p65/p50) revealed a reduction in Rela (p65) and Nfkb1 (p105, precursor to p50) gene expression (p0) and p65 subunit protein levels (p0 and p3) vs. older ages. The acute hepatic innate immune response to LPS was associated by the degradation of the NF-κB inhibitory proteins (IκBα and IκBβ), and nuclear translocation of the NF-κB subunit p50 in all ages, whereas nuclear translocation of the NF-κB subunit p65 was only observed in the p35 and adult mouse. Consistent with these findings, we detected NF-κB subunit p65 nuclear staining exclusively in the LPS-exposed adult liver compared with p7 mouse. We next interrogated the LPS-induced hepatic expression of pro-inflammatory genes (Tnf, Icam1, Ccl3, and Traf1), and observed a gradually increase in gene expression starting from p0. Confirming our results, hepatic NF-κB subunit p65 nuclear translocation was associated with up-regulation of the Icam1 gene in the adult, and was not detected in the p7 mouse. Thus, an inflammatory challenge induces an NF-κB-mediated hepatic innate immune response activation across all developmental ages, but nuclear translocation of the NF-κB subunit p65 and associated induction of pro-inflammatory genes occurred only after the first month of life. Our results demonstrate that the LPS-induced hepatic innate immune response is developmentally regulated by the NF-κB subunit p65 in the mouse.
Collapse
Affiliation(s)
- Miguel A Zarate
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
| | - Leanna M Nguyen
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
| | - Robyn K De Dios
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
| | - Lijun Zheng
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
25
|
p53 destabilizing protein skews asymmetric division and enhances NOTCH activation to direct self-renewal of TICs. Nat Commun 2020; 11:3084. [PMID: 32555153 PMCID: PMC7299990 DOI: 10.1038/s41467-020-16616-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 04/28/2020] [Indexed: 01/11/2023] Open
Abstract
Tumor-initiating stem-like cells (TICs) are defective in maintaining asymmetric cell division and responsible for tumor recurrence. Cell-fate-determinant molecule NUMB-interacting protein (TBC1D15) is overexpressed and contributes to p53 degradation in TICs. Here we identify TBC1D15-mediated oncogenic mechanisms and tested the tumorigenic roles of TBC1D15 in vivo. We examined hepatocellular carcinoma (HCC) development in alcohol Western diet-fed hepatitis C virus NS5A Tg mice with hepatocyte-specific TBC1D15 deficiency or expression of non-phosphorylatable NUMB mutations. Liver-specific TBC1D15 deficiency or non-p-NUMB expression reduced TIC numbers and HCC development. TBC1D15–NuMA1 association impaired asymmetric division machinery by hijacking NuMA from LGN binding, thereby favoring TIC self-renewal. TBC1D15–NOTCH1 interaction activated and stabilized NOTCH1 which upregulated transcription of NANOG essential for TIC expansion. TBC1D15 activated three novel oncogenic pathways to promote self-renewal, p53 loss, and Nanog transcription in TICs. Thus, this central regulator could serve as a potential therapeutic target for treatment of HCC. Normal stem cells are maintained by asymmetric cell division, but this process is dysregulated in tumour initiating stem-like cells (TICs). Here, the authors show that TBC1D15 impairs the asymmetric division machinery and activates NOTCH pathway for TIC self-renewal and expansion to promote liver tumorigenesis.
Collapse
|
26
|
Ameur LB, Marie P, Thenoz M, Giraud G, Combe E, Claude JB, Lemaire S, Fontrodona N, Polveche H, Bastien M, Gessain A, Wattel E, Bourgeois CF, Auboeuf D, Mortreux F. Intragenic recruitment of NF-κB drives splicing modifications upon activation by the oncogene Tax of HTLV-1. Nat Commun 2020; 11:3045. [PMID: 32546717 PMCID: PMC7298006 DOI: 10.1038/s41467-020-16853-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Chronic NF-κB activation in inflammation and cancer has long been linked to persistent activation of NF-κB–responsive gene promoters. However, NF-κB factors also massively bind to gene bodies. Here, we demonstrate that recruitment of the NF-κB factor RELA to intragenic regions regulates alternative splicing upon NF-κB activation by the viral oncogene Tax of HTLV-1. Integrative analyses of RNA splicing and chromatin occupancy, combined with chromatin tethering assays, demonstrate that DNA-bound RELA interacts with and recruits the splicing regulator DDX17, in an NF-κB activation-dependent manner. This leads to alternative splicing of target exons due to the RNA helicase activity of DDX17. Similar results were obtained upon Tax-independent NF-κB activation, indicating that Tax likely exacerbates a physiological process where RELA provides splice target specificity. Collectively, our results demonstrate a physical and direct involvement of NF-κB in alternative splicing regulation, which significantly revisits our knowledge of HTLV-1 pathogenesis and other NF-κB-related diseases. The nuclear factors κB (NF-κB) is a transcription factor involved in immune functions, inflammation, and cancer. Here, the authors show that the NF-κB factor RELA regulates splicing of target genes by recruiting DDX17 on chromatin upon expression of the viral oncogene Tax.
Collapse
Affiliation(s)
- Lamya Ben Ameur
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie Site Jacques Monod, 69007, Lyon, France
| | - Paul Marie
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie Site Jacques Monod, 69007, Lyon, France
| | - Morgan Thenoz
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie Site Jacques Monod, 69007, Lyon, France.,Department of Pediatrics and Medical Genetics, Faculty of Medicine and Health Sciences, 9000, Gent, Belgium
| | - Guillaume Giraud
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie Site Jacques Monod, 69007, Lyon, France
| | - Emmanuel Combe
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie Site Jacques Monod, 69007, Lyon, France
| | - Jean-Baptiste Claude
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie Site Jacques Monod, 69007, Lyon, France
| | - Sebastien Lemaire
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie Site Jacques Monod, 69007, Lyon, France
| | - Nicolas Fontrodona
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie Site Jacques Monod, 69007, Lyon, France
| | | | - Marine Bastien
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie Site Jacques Monod, 69007, Lyon, France.,School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Antoine Gessain
- Unité d'Epidémiologie et Physiopathologie des Virus Oncogénes, Institut Pasteur, Paris, France
| | - Eric Wattel
- Université Lyon 1, CNRS UMR5239, Oncovirologie et Biothérapies, Faculté de Médecine Lyon Sud, ENS - HCL, Pierre Bénite, France.,Université Lyon 1, Service d'Hématologie, Pavillon Marcel Bérard, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| | - Cyril F Bourgeois
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie Site Jacques Monod, 69007, Lyon, France
| | - Didier Auboeuf
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie Site Jacques Monod, 69007, Lyon, France.
| | - Franck Mortreux
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie Site Jacques Monod, 69007, Lyon, France.
| |
Collapse
|
27
|
de Jesús TJ, Ramakrishnan P. NF-κB c-Rel Dictates the Inflammatory Threshold by Acting as a Transcriptional Repressor. iScience 2020; 23:100876. [PMID: 32062419 PMCID: PMC7031323 DOI: 10.1016/j.isci.2020.100876] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 01/11/2020] [Accepted: 01/28/2020] [Indexed: 12/14/2022] Open
Abstract
NF-κB/Rel family of transcription factors plays a central role in initiation and resolution of inflammatory responses. Here, we identified a function of the NF-κB subunit c-Rel as a transcriptional repressor of inflammatory genes. Genetic deletion of c-Rel substantially potentiates the expression of several TNF-α-induced RelA-dependent mediators of inflammation. v-Rel, the viral homologue of c-Rel, but not RelB, also possesses this repressive function. Mechanistically, we found that c-Rel selectively binds to the co-repressor HDAC1 and competitively binds to the DNA mediating HDAC1 recruitment to the promoters of inflammatory genes. A specific point mutation at tyrosine25 in c-Rel's DNA-binding domain, for which a missense single nucleotide variation (Y25H) exists in humans, completely abrogated its ability to bind DNA and repress TNF-α-induced, RelA-mediated transcription. Our findings reveal that the transactivator NF-κB subunit c-Rel also plays a role as a transcriptional repressor in the maintenance of inflammatory homeostasis.
Collapse
Affiliation(s)
- Tristan James de Jesús
- Department of Pathology, School of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, 6526, Wolstein Research Building, 2103 Cornell Road, Cleveland, OH 44106, USA
| | - Parameswaran Ramakrishnan
- Department of Pathology, School of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, 6526, Wolstein Research Building, 2103 Cornell Road, Cleveland, OH 44106, USA; Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; The Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
28
|
Upregulation of CASP9 through NF-κB and Its Target MiR-1276 Contributed to TNFα-promoted Apoptosis of Cancer Cells Induced by Doxorubicin. Int J Mol Sci 2020; 21:ijms21072290. [PMID: 32225068 PMCID: PMC7177739 DOI: 10.3390/ijms21072290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/17/2020] [Accepted: 03/23/2020] [Indexed: 12/18/2022] Open
Abstract
Under some conditions, nuclear factor-κB (NF-κB) has a pro-apoptotic role, but the mechanisms underlying this function remain unclear. This study demonstrated that NF-κB directly binds to CASP9 and miR1276 in tumor necrosis factor α (TNFα)-treated HeLa and HepG2 cells. NF-κB upregulated CASP9 expression, whereas downregulated miR1276 expression in the TNFα-treated cells. The miR1276 repressed CASP9 expression in both cells. As a result, a typical NF-κB-mediated coherent feed-forward loop was formed in the TNFα-treated cells. It was proposed that the NF-κB-mediated loop may contribute to cell apoptosis under certain conditions. This opinion was supported by the following evidence: TNFα promoted the apoptosis of HeLa and HepG2 cells induced by doxorubicin (DOX). CASP9 was significantly upregulated and activated by TNFα in the DOX-induced cells. Moreover, a known inhibitor of CASP9 activation significantly repressed the TNFα promotion of apoptosis induced by DOX. These findings indicate that CASP9 is a new mediator of the NF-κB pro-apoptotic pathway, at least in such conditions. This study therefore provides new insights into the pro-apoptotic role of NF-κB. The results also shed new light on the molecular mechanism underlying TNFα-promotion of cancer cells apoptosis induced by some anticancer drugs such as DOX.
Collapse
|
29
|
Ren X, Chen C, Luo Y, Liu M, Li Y, Zheng S, Ye H, Fu Z, Li M, Li Z, Chen R. lncRNA-PLACT1 sustains activation of NF-κB pathway through a positive feedback loop with IκBα/E2F1 axis in pancreatic cancer. Mol Cancer 2020; 19:35. [PMID: 32085715 PMCID: PMC7033942 DOI: 10.1186/s12943-020-01153-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 02/13/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The activation of NF-κB signaling pathway is regarded as the dominant process that correlates with tumorigenesis. Recently, increasing evidence shows that long noncoding RNAs (lncRNAs) play crucial roles in sustaining the NF-κB signaling pathway. However, the underlying mechanisms have not yet been elucidated. METHODS The expression and clinical features of PLACT1 were analyzed in a 166-case cohort of PDAC by qRT-PCR and in situ hybridization. The functional role of PLACT1 was evaluated by both in vitro and in vivo experiments. Chromatin isolation by RNA purification assays were utilized to examine the interaction of PLACT1 with IκBα promoter. RESULTS We identified a novel lncRNA-PLACT1, which was significantly upregulated in tumor tissues and correlated with progression and poor survival in PDAC patients. Moreover, PLACT1 promoted the proliferation and invasion of PDAC cells in vitro. Consistently, PLACT1 overexpression fostered the progression of PDAC both in orthotopic and lung metastasis mice models. Mechanistically, PLACT1 suppressed IκBα expression by recruiting hnRNPA1 to IκBα promoter, which led to increased H3K27me3 that decreased the transcriptional level of IκBα. Furthermore, E2F1-mediated overexpression of PLACT1 modulated the progression of PDAC by sustained activation of NF-κB signaling pathway through forming a positive feedback loop with IκBα. Importantly, administration of the NF-κB signaling pathway inhibitor significantly suppressed PLACT1-induced sustained activation of NF-κB signaling pathway, leading to reduced tumorigenesis in vivo. CONCLUSIONS Our findings suggest that PLACT1 provides a novel epigenetic mechanism involved in constitutive activation of NF-κB signaling pathway and may represent a new therapeutic target of PDAC.
Collapse
Affiliation(s)
- Xiaofan Ren
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, Guangdong province, 510120, People's Republic of China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China
| | - Changhao Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China. .,Department of Urology, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, Guangdong province, 510120, People's Republic of China.
| | - Yuming Luo
- Department of Hepatopancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China
| | - Mingyang Liu
- Department of Medicine, Department of Surgery, the University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1262A, Oklahoma City, OK, 73104, USA
| | - Yuting Li
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, Guangdong province, 510120, People's Republic of China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China
| | - Shangyou Zheng
- Department of Hepatopancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China
| | - Huilin Ye
- Department of Hepatopancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China
| | - Zhiqiang Fu
- Department of Hepatopancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China
| | - Min Li
- Department of Medicine, Department of Surgery, the University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1262A, Oklahoma City, OK, 73104, USA.
| | - Zhihua Li
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, Guangdong province, 510120, People's Republic of China. .,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, 510120, Guangdong province, People's Republic of China.
| | - Rufu Chen
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106th of 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province, 510080, People's Republic of China.
| |
Collapse
|
30
|
Methane-Rich Saline Counteracts Cholestasis-Induced Liver Damage via Regulating the TLR4/NF- κB/NLRP3 Inflammasome Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6565283. [PMID: 31827690 PMCID: PMC6885841 DOI: 10.1155/2019/6565283] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 10/10/2019] [Indexed: 12/17/2022]
Abstract
Cholestatic liver injury, due to obstruction of the biliary tract or genetic defects, is often accompanied by progressive inflammation and liver fibrosis. Methane-rich saline (MRS) has anti-inflammatory properties. However, whether MRS can provide protective effect in cholestatic liver injury is still unclear. In this study, Sprague-Dawley rats received bile duct ligation (BDL) to generate a cholestatic model followed by MRS treatment (10 mL/kg, ip treatment) every 12 h after the operation to explore the potential protective mechanism of MRS in cholestatic liver injury. We found that MRS effectively improved liver function, alleviated liver pathological damage, and localized infiltration of inflammatory cells. MRS treatment decreased the expression of hepatic fibrosis-associated proteins to alleviate liver fibrosis. Furthermore, MRS treatment suppressed the TLR4/NF-κB pathway and further reduced the levels of proinflammatory factors. Downregulation of NF-κB subsequently reduced the NLRP3 expression to inhibit pyroptosis. Our data indicated that methane treatment prevented cholestatic liver injury via anti-inflammatory properties that involved the TLR4/NF-κB/NLRP3 signaling pathway.
Collapse
|
31
|
Mulero MC, Wang VYF, Huxford T, Ghosh G. Genome reading by the NF-κB transcription factors. Nucleic Acids Res 2019; 47:9967-9989. [PMID: 31501881 PMCID: PMC6821244 DOI: 10.1093/nar/gkz739] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/05/2019] [Accepted: 08/21/2019] [Indexed: 12/25/2022] Open
Abstract
The NF-κB family of dimeric transcription factors regulates transcription by selectively binding to DNA response elements present within promoters or enhancers of target genes. The DNA response elements, collectively known as κB sites or κB DNA, share the consensus 5'-GGGRNNNYCC-3' (where R, Y and N are purine, pyrimidine and any nucleotide base, respectively). In addition, several DNA sequences that deviate significantly from the consensus have been shown to accommodate binding by NF-κB dimers. X-ray crystal structures of NF-κB in complex with diverse κB DNA have helped elucidate the chemical principles that underlie target selection in vitro. However, NF-κB dimers encounter additional impediments to selective DNA binding in vivo. Work carried out during the past decades has identified some of the barriers to sequence selective DNA target binding within the context of chromatin and suggests possible mechanisms by which NF-κB might overcome these obstacles. In this review, we first highlight structural features of NF-κB:DNA complexes and how distinctive features of NF-κB proteins and DNA sequences contribute to specific complex formation. We then discuss how native NF-κB dimers identify DNA binding targets in the nucleus with support from additional factors and how post-translational modifications enable NF-κB to selectively bind κB sites in vivo.
Collapse
Affiliation(s)
- Maria Carmen Mulero
- Department of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Vivien Ya-Fan Wang
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| | - Tom Huxford
- Structural Biochemistry Laboratory, Department of Chemistry and Biochemistry, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Gourisankar Ghosh
- Department of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
32
|
Mishra N, Schwerdtner L, Sams K, Mondal S, Ahmad F, Schmidt RE, Coonrod SA, Thompson PR, Lerch MM, Bossaller L. Cutting Edge: Protein Arginine Deiminase 2 and 4 Regulate NLRP3 Inflammasome-Dependent IL-1β Maturation and ASC Speck Formation in Macrophages. THE JOURNAL OF IMMUNOLOGY 2019; 203:795-800. [PMID: 31292215 DOI: 10.4049/jimmunol.1800720] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 06/15/2019] [Indexed: 01/22/2023]
Abstract
Protein arginine deiminase (PAD) enzymes catalyze the conversion of protein-bound arginine into citrulline, an irreversible posttranslational modification with loss of a positive charge that can influence protein-protein interactions and protein structure. Protein arginine deiminase activity depends on high intracellular calcium concentrations occurring in dying cells. In this study, we demonstrate that protein citrullination is common during pyroptotic cell death in macrophages and that inhibition of PAD enzyme activity by Cl-amidine, a pan-PAD inhibitor, blocks NLRP3 inflammasome assembly and proinflammatory IL-1β release in macrophages. Genetic deficiency of either PAD2 or PAD4 alone in murine macrophages does not impair IL-1β release; however, pharmacological inhibition or small interfering RNA knockdown of PAD2 within PAD4-/- macrophages does. Our results suggest that PAD2 and 4 activity in macrophages is required for optimal inflammasome assembly and IL-1β release, a finding of importance for autoimmune diseases and inflammation.
Collapse
Affiliation(s)
- Neha Mishra
- Section of Rheumatology, Department of Medicine A, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Lidja Schwerdtner
- Section of Rheumatology, Department of Medicine A, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Kelly Sams
- Department of Biomedical Sciences, Baker Institute for Animal Health, Cornell University, Ithaca, NY 14853
| | - Santanu Mondal
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Fareed Ahmad
- Department of Dermatology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Reinhold E Schmidt
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, 30625 Hannover, Germany; and
| | - Scott A Coonrod
- Department of Biomedical Sciences, Baker Institute for Animal Health, Cornell University, Ithaca, NY 14853
| | - Paul R Thompson
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Markus M Lerch
- Department of Medicine A, University Medicine Greifswald, 1745 Greifswald, Germany
| | - Lukas Bossaller
- Section of Rheumatology, Department of Medicine A, University Medicine Greifswald, 17475 Greifswald, Germany;
| |
Collapse
|
33
|
Lee WJ, Chen LC, Lin JH, Cheng TC, Kuo CC, Wu CH, Chang HW, Tu SH, Ho YS. Melatonin promotes neuroblastoma cell differentiation by activating hyaluronan synthase 3-induced mitophagy. Cancer Med 2019; 8:4821-4835. [PMID: 31274246 PMCID: PMC6712479 DOI: 10.1002/cam4.2389] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 12/20/2022] Open
Abstract
Neuroblastoma is the second most common pediatric malignancy and has a high rate of spontaneous remission. Uncovering the mechanisms underlying neuroblastoma cell differentiation is critical for therapeutic purposes. A neuroblastoma cell line (N2a) treated with either serum withdrawal (<2.5%) or melatonin (>0.1 nmol/L) for 24 hours was used as a cell differentiation research model. Interestingly, the hyaluronan synthase 3 (HAS3) protein was induced in differentiated N2a cells. N2a-allografted nude mice received an intraperitoneal injection of melatonin (40 or 80 mg/kg/day for 3 weeks). The mean tumor volume in mice treated with 80 mg/kg melatonin was smaller than that in PBS-treated mice (1416.3 and 3041.3 mm3 , respectively, difference = 1625 mm3 , *P = 0.0003, n = 7 per group). Compared with the vector control group, N2a cells with forced HAS3 overexpression showed significantly increased neuron length (*P = 0.00082) and neurite outgrowth (*P = 0.00059). Intracellular changes in autophagy, including distorted mitochondria with abnormal circular inner membranes, were detected by transmission electron microscopy (TEM). Our study demonstrated that HAS3-mediated signaling activated by physiological concentrations of melatonin (>0.1 nmol/L) triggered significant N2a cell differentiation. These results provide molecular data with potential clinical relevance for therapeutic drug development.
Collapse
Affiliation(s)
- Wen-Jui Lee
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan
| | - Li-Ching Chen
- Division of Breast Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan.,Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Juo-Han Lin
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan
| | - Tzu-Chun Cheng
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Ching-Chuan Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Chih-Hsiung Wu
- Department of Surgery, En Chun Kong Hospital, New Taipei City, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hui-Wen Chang
- Department of Laboratory Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Shih-Hsin Tu
- Division of Breast Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan.,Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yuan-Soon Ho
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.,School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Department of Laboratory Medicine, Taipei Medical University Hospital, Taipei, Taiwan.,Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
34
|
Piaszyk-Borychowska A, Széles L, Csermely A, Chiang HC, Wesoły J, Lee CK, Nagy L, Bluyssen HAR. Signal Integration of IFN-I and IFN-II With TLR4 Involves Sequential Recruitment of STAT1-Complexes and NFκB to Enhance Pro-inflammatory Transcription. Front Immunol 2019; 10:1253. [PMID: 31231385 PMCID: PMC6558219 DOI: 10.3389/fimmu.2019.01253] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 05/17/2019] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the blood vessels, characterized by atherosclerotic lesion formation. Vascular Smooth Muscle Cells (VSMC), macrophages (MΦ), and dendritic cells (DC) play a crucial role in vascular inflammation and atherosclerosis. Interferon (IFN)α, IFNγ, and Toll-like receptor (TLR)4 activate pro-inflammatory gene expression and are pro-atherogenic. Gene expression regulation of many pro-inflammatory genes has shown to rely on Signal Integration (SI) between IFNs and TLR4 through combinatorial actions of the Signal Transducer and Activator of Transcription (STAT)1 complexes ISGF3 and γ-activated factor (GAF), and Nuclear Factor-κB (NFκB). Thus, IFN pre-treatment (“priming”) followed by LPS stimulation leads to enhanced transcriptional responses as compared to the individual stimuli. To characterize the mechanism of priming-induced IFNα + LPS- and IFNγ + LPS-dependent SI in vascular cells as compared to immune cells, we performed a comprehensive genome-wide analysis of mouse VSMC, MΦ, and DC in response to IFNα, IFNγ, and/or LPS. Thus, we identified IFNα + LPS or IFNγ + LPS induced genes commonly expressed in these cell types that bound STAT1 and p65 at comparable γ-activated sequence (GAS), Interferon-stimulated response element (ISRE), or NFκB sites in promoter proximal and distal regions. Comparison of the relatively high number of overlapping ISRE sites in these genes unraveled a novel role of ISGF3 and possibly STAT1/IRF9 in IFNγ responses. In addition, similar STAT1-p65 co-binding modes were detected for IFNα + LPS and IFNγ + LPS up-regulated genes, which involved recruitment of STAT1 complexes preceding p65 to closely located GAS/NFκB or ISRE/NFκB composite sites already upon IFNα or IFNγ treatment. This STAT1-p65 co-binding significantly increased after subsequent LPS exposure and correlated with histone acetylation, PolII recruitment, and amplified target gene transcription in a STAT1-p65 co-bound dependent manner. Thus, co-binding of STAT1-containing transcription factor complexes and NFκB, activated by IFN-I or IFN-II together with LPS, provides a platform for robust transcriptional activation of pro-inflammatory genes. Moreover, our data offer an explanation for the comparable effects of IFNα or IFNγ priming on TLR4-induced activation in vascular and immune cells, with important implications in atherosclerosis.
Collapse
Affiliation(s)
| | - Lajos Széles
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Attila Csermely
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Hsin-Chien Chiang
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Joanna Wesoły
- Laboratory of High Throughput Technologies, Adam Mickiewicz University, Poznan, Poland
| | - Chien-Kuo Lee
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Laszlo Nagy
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary.,Departments of Medicine and Biological Chemistry, Johns Hopkins All Children's Hospital, Johns Hopkins University School of Medicine, St. Petersburg, FL, United States
| | - Hans A R Bluyssen
- Department of Human Molecular Genetics, Adam Mickiewicz University, Poznan, Poland
| |
Collapse
|
35
|
Yuan G, Xu L, Cai T, Hua B, Sun N, Yan Z, Lu C, Qian R. Clock mutant promotes osteoarthritis by inhibiting the acetylation of NFκB. Osteoarthritis Cartilage 2019; 27:922-931. [PMID: 30716535 DOI: 10.1016/j.joca.2019.01.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/04/2018] [Accepted: 01/20/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVES To examine the effect of the circadian gene Clock on posttranscriptional function and pro-inflammatory mechanisms in osteoarthritis (OA). METHODS The cartilage from Clock mutant mice was assessed using histology, (OA) score, and real-time polymerase chain reaction (PCR) quantification of key pro-inflammatory genes. Nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) translocation, posttranslational state and expression levels during day and night conditions were assessed using immunoblot and IP. The regulation of transcription by Clock in cartilage tissue was assessed by using chromatin immunoprecipitation (ChIP) and luciferase assays. Total acetylation level and pattern over 24 h were quantified using immunoblot and real-time PCR. Finally, the effects of exogenous Clock nanoparticle treatment were quantified by histology and immunoblot. RESULTS The Clock mutation significantly promoted the degradation of cartilage and the expression of the key pro-inflammatory mediators, IL-1β, IL-6 and MCP-1. The Clock mutation significantly promoted NFκB nuclear translocation. The circadian protein CLOCK positively regulates NFκB at the transcriptional level by binding the E-box domain. The Clock mutation significantly inhibited the total lysine acetylation level in cartilage and inhibited NFκB acetylation at the Lys310 residue but promoted phosphorylation at the Ser276 residue. The forced expression of Clock in vivo inhibited NFκB activation by increasing acetylation and decreasing phosphorylation levels and by decreasing cartilage damage and inflammation. CONCLUSIONS This study demonstrates the mutation of Clock promotes inflammatory activity by mediating the posttranscriptional regulation of NFκB in OA pathogenesis.
Collapse
Affiliation(s)
- G Yuan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Research Center on Aging and Medicine, Fudan University, Shanghai 200032, China
| | - L Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - T Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - B Hua
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - N Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Research Center on Aging and Medicine, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Z Yan
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - C Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Research Center on Aging and Medicine, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China.
| | - R Qian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Research Center on Aging and Medicine, Fudan University, Shanghai 200032, China.
| |
Collapse
|
36
|
Borghini L, Png E, Binder A, Wright VJ, Pinnock E, de Groot R, Hazelzet J, Emonts M, Van der Flier M, Schlapbach LJ, Anderson S, Secka F, Salas A, Fink C, Carrol ED, Pollard AJ, Coin LJ, Kuijpers TW, Martinon-Torres F, Zenz W, Levin M, Hibberd ML, Davila S. Identification of regulatory variants associated with genetic susceptibility to meningococcal disease. Sci Rep 2019; 9:6966. [PMID: 31061469 PMCID: PMC6502852 DOI: 10.1038/s41598-019-43292-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 04/17/2019] [Indexed: 12/30/2022] Open
Abstract
Non-coding genetic variants play an important role in driving susceptibility to complex diseases but their characterization remains challenging. Here, we employed a novel approach to interrogate the genetic risk of such polymorphisms in a more systematic way by targeting specific regulatory regions relevant for the phenotype studied. We applied this method to meningococcal disease susceptibility, using the DNA binding pattern of RELA – a NF-kB subunit, master regulator of the response to infection – under bacterial stimuli in nasopharyngeal epithelial cells. We designed a custom panel to cover these RELA binding sites and used it for targeted sequencing in cases and controls. Variant calling and association analysis were performed followed by validation of candidate polymorphisms by genotyping in three independent cohorts. We identified two new polymorphisms, rs4823231 and rs11913168, showing signs of association with meningococcal disease susceptibility. In addition, using our genomic data as well as publicly available resources, we found evidences for these SNPs to have potential regulatory effects on ATXN10 and LIF genes respectively. The variants and related candidate genes are relevant for infectious diseases and may have important contribution for meningococcal disease pathology. Finally, we described a novel genetic association approach that could be applied to other phenotypes.
Collapse
Affiliation(s)
- Lisa Borghini
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore. .,Infectious diseases, Genome Institute of Singapore, Singapore, Singapore.
| | - Eileen Png
- Infectious diseases, Genome Institute of Singapore, Singapore, Singapore
| | - Alexander Binder
- Department of General Pediatrics, Medical University of Graz, Graz, Austria
| | - Victoria J Wright
- Section for Paediatrics, Division of Infectious Diseases, Department of Medicine, Imperial College London, London, UK
| | - Ellie Pinnock
- Micropathology Ltd, University of Warwick, Warwick, UK
| | - Ronald de Groot
- Department of Pediatrics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan Hazelzet
- Department of Pediatrics, Erasmus Medical Center-Sophia Children's hospital, University Medical Center, Rotterdam, The Netherlands
| | - Marieke Emonts
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Paediatric Infectious Diseases and Immunology Department, Newcastle upon Tyne Hospitals Foundation Trust, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Michiel Van der Flier
- Department of Pediatrics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Luregn J Schlapbach
- Faculty of Medicine, The University of Queensland, Brisbane, Australia.,Paediatric Critical Care Research Group, Mater Research Institute, University of Queensland, Brisbane, Australia.,Paediatric Intensive Care Unit, Lady Cilento Children's Hospital, Brisbane, Australia.,Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Fatou Secka
- Medical Research Council Unit Gambia, Banjul, The Gambia
| | - Antonio Salas
- Unidade de Xenética, Departamento de Anatomía Patolóxica e Ciencias Forenses, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigaciones Sanitarias (IDIS), Hospital Clínico Universitario de Santiago, Galicia, Spain
| | - Colin Fink
- Micropathology Ltd, University of Warwick, Warwick, UK
| | - Enitan D Carrol
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Lachlan J Coin
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, 4072, Australia
| | - Taco W Kuijpers
- Division of Pediatric Hematology, Immunology and Infectious diseases, Emma Children's Hospital Academic Medical Center, Amsterdam, The Netherlands
| | - Federico Martinon-Torres
- Translational Pediatrics and Infectious Diseases, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain.,GENVIP Research Group (www.genvip.org), Instituto de Investigación Sanitaria de Santiago, Galicia, Spain
| | - Werner Zenz
- Department of General Pediatrics, Medical University of Graz, Graz, Austria
| | - Michael Levin
- Section for Paediatrics, Division of Infectious Diseases, Department of Medicine, Imperial College London, London, UK
| | - Martin L Hibberd
- Infectious diseases, Genome Institute of Singapore, Singapore, Singapore.,Infectious and Tropical Disease, London School of Hygiene & Tropical Medicine, London, UK
| | - Sonia Davila
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore. .,SingHealth Duke-NUS Institute of Precision Medicine (PRISM), Singapore, Singapore. .,Duke-NUS Medical School, Singapore, Singapore.
| | | |
Collapse
|
37
|
Dantoft W, Robertson KA, Watkins WJ, Strobl B, Ghazal P. Metabolic Regulators Nampt and Sirt6 Serially Participate in the Macrophage Interferon Antiviral Cascade. Front Microbiol 2019; 10:355. [PMID: 30886604 PMCID: PMC6409323 DOI: 10.3389/fmicb.2019.00355] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/11/2019] [Indexed: 11/13/2022] Open
Abstract
Molecular determinants underlying interferon (IFN)-macrophage biology can help delineate enzyme systems, pathways and mechanisms for enabling host-directed therapeutic approaches against infection. Notably, while the IFN antiviral response is known to be directly coupled to mevalonate-sterol biosynthesis, mechanistic insight for providing host pathway-therapeutic targets remain incomplete. Here, we show that Nampt and Sirt6 are coordinately regulated upon immune activation of macrophages and contribute to the IFN-sterol antiviral response. In silico analysis of the Nampt and Sirt6 promoter regions identified multiple core immune gene-regulatory transcription factor sites, including Stat1, implicating a molecular link to IFN control. Experimentally, we show using a range of genetically IFN-defective macrophages that the expression of Nampt is stringently regulated by the Jak/Stat-pathway while Sirt6 activation is temporally displaced in a partial IFN-dependent manner. We further show that pharmacological inhibition of Nampt and small interfering RNA (siRNA)-mediated inhibition of Nampt and Sirt6 promotes viral growth of cytomegalovirus in both fibroblasts and macrophages. Our results support the notion of pharmacologically exploiting immune regulated enzyme systems of macrophages for use as an adjuvant-based therapy for augmenting host protective pathway responses to infection.
Collapse
Affiliation(s)
- Widad Dantoft
- Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Kevin A Robertson
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - W John Watkins
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, Department for Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Peter Ghazal
- Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Division of Infection and Pathway Medicine, School of Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
38
|
Riedlinger T, Liefke R, Meier-Soelch J, Jurida L, Nist A, Stiewe T, Kracht M, Schmitz ML. NF-κB p65 dimerization and DNA-binding is important for inflammatory gene expression. FASEB J 2018; 33:4188-4202. [PMID: 30526044 PMCID: PMC6404571 DOI: 10.1096/fj.201801638r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Increasing evidence shows that many transcription factors execute important biologic functions independent from their DNA-binding capacity. The NF-κB p65 (RELA) subunit is a central regulator of innate immunity. Here, we investigated the relative functional contribution of p65 DNA-binding and dimerization in p65-deficient human and murine cells reconstituted with single amino acid mutants preventing either DNA-binding (p65 E/I) or dimerization (p65 FL/DD). DNA-binding of p65 was required for RelB-dependent stabilization of the NF-κB p100 protein. The antiapoptotic function of p65 and expression of the majority of TNF-α–induced genes were dependent on p65’s ability to bind DNA and to dimerize. Chromatin immunoprecipitation with massively parallel DNA sequencing experiments revealed that impaired DNA-binding and dimerization strongly diminish the chromatin association of p65. However, there were also p65-independent TNF-α–inducible genes and a subgroup of p65 binding sites still allowed some residual chromatin association of the mutants. These sites were enriched in activator protein 1 (AP-1) binding motifs and showed increased chromatin accessibility and basal transcription. This suggests a mechanism of assisted p65 chromatin association that can be in part facilitated by chromatin priming and cooperativity with other transcription factors such as AP-1.—Riedlinger, T., Liefke, R., Meier-Soelch, J., Jurida, L., Nist, A., Stiewe, T., Kracht, M., Schmitz, M. L. NF-κB p65 dimerization and DNA-binding is important for inflammatory gene expression.
Collapse
Affiliation(s)
- Tabea Riedlinger
- Institute of Biochemistry, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany
| | - Robert Liefke
- Institute of Biochemistry, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany.,Institute of Molecular Biology and Tumor Research (IMT), Philipps University Marburg, Marburg, Germany
| | - Johanna Meier-Soelch
- Rudolf-Buchheim-Institute of Pharmacology, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany; and
| | - Liane Jurida
- Rudolf-Buchheim-Institute of Pharmacology, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany; and
| | - Andrea Nist
- Genomics Core Facility-Institute of Molecular Oncology, Philipps University Marburg, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility-Institute of Molecular Oncology, Philipps University Marburg, Marburg, Germany
| | - Michael Kracht
- Rudolf-Buchheim-Institute of Pharmacology, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany; and
| | - M Lienhard Schmitz
- Institute of Biochemistry, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
39
|
Kalita CA, Brown CD, Freiman A, Isherwood J, Wen X, Pique-Regi R, Luca F. High-throughput characterization of genetic effects on DNA-protein binding and gene transcription. Genome Res 2018; 28:1701-1708. [PMID: 30254052 PMCID: PMC6211638 DOI: 10.1101/gr.237354.118] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 09/20/2018] [Indexed: 12/29/2022]
Abstract
Many variants associated with complex traits are in noncoding regions and contribute to phenotypes by disrupting regulatory sequences. To characterize these variants, we developed a streamlined protocol for a high-throughput reporter assay, Biallelic Targeted STARR-seq (BiT-STARR-seq), that identifies allele-specific expression (ASE) while accounting for PCR duplicates through unique molecular identifiers. We tested 75,501 oligos (43,500 SNPs) and identified 2720 SNPs with significant ASE (FDR < 10%). To validate disruption of binding as one of the mechanisms underlying ASE, we developed a new high-throughput allele-specific binding assay for NFKB1. We identified 2684 SNPs with allele-specific binding (ASB) (FDR < 10%); 256 of these SNPs also had ASE (OR = 1.97, P-value = 0.0006). Of variants associated with complex traits, 1531 resulted in ASE, and 1662 showed ASB. For example, we characterized that the Crohn's disease risk variant for rs3810936 increases NFKB1 binding and results in altered gene expression.
Collapse
Affiliation(s)
- Cynthia A Kalita
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan 48202, USA
| | - Christopher D Brown
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Andrew Freiman
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan 48202, USA
| | - Jenna Isherwood
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan 48202, USA
| | - Xiaoquan Wen
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Roger Pique-Regi
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan 48202, USA.,Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan 48202, USA
| | - Francesca Luca
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan 48202, USA.,Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan 48202, USA
| |
Collapse
|
40
|
Vandenbon A, Kumagai Y, Lin M, Suzuki Y, Nakai K. Waves of chromatin modifications in mouse dendritic cells in response to LPS stimulation. Genome Biol 2018; 19:138. [PMID: 30231913 PMCID: PMC6146659 DOI: 10.1186/s13059-018-1524-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/04/2018] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The importance of transcription factors (TFs) and epigenetic modifications in the control of gene expression is widely accepted. However, causal relationships between changes in TF binding, histone modifications, and gene expression during the response to extracellular stimuli are not well understood. Here, we analyze the ordering of these events on a genome-wide scale in dendritic cells in response to lipopolysaccharide (LPS) stimulation. RESULTS Using a ChIP-seq time series dataset, we find that the LPS-induced accumulation of different histone modifications follows clearly distinct patterns. Increases in H3K4me3 appear to coincide with transcriptional activation. In contrast, H3K9K14ac accumulates early after stimulation, and H3K36me3 at later time points. Integrative analysis with TF binding data reveals potential links between TF activation and dynamics in histone modifications. Especially, LPS-induced increases in H3K9K14ac and H3K4me3 are associated with binding by STAT1/2 and were severely impaired in Stat1-/- cells. CONCLUSIONS While the timing of short-term changes of some histone modifications coincides with changes in transcriptional activity, this is not the case for others. In the latter case, dynamics in modifications more likely reflect strict regulation by stimulus-induced TFs and their interactions with chromatin modifiers.
Collapse
Affiliation(s)
- Alexis Vandenbon
- Laboratory of Infection and Prevention, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan.
- Institute for Liberal Arts and Sciences, Kyoto University, Kyoto, 606-8507, Japan.
| | - Yutaro Kumagai
- Quantitative Immunology Research Unit, Immunology Frontier Research Center (IFReC), Osaka University, Suita, 565-0871, Japan
- Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, 305-8565, Japan
| | - Mengjie Lin
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, 277-8561, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, 277-8561, Japan
| | - Kenta Nakai
- Laboratory of Functional Analysis in silico, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, 108-8639, Japan.
| |
Collapse
|
41
|
Zhao M, Joy J, Zhou W, De S, Wood WH, Becker KG, Ji H, Sen R. Transcriptional outcomes and kinetic patterning of gene expression in response to NF-κB activation. PLoS Biol 2018; 16:e2006347. [PMID: 30199532 PMCID: PMC6147668 DOI: 10.1371/journal.pbio.2006347] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 09/20/2018] [Accepted: 08/23/2018] [Indexed: 11/26/2022] Open
Abstract
Transcription factor nuclear factor kappa B (NF-κB) regulates cellular responses to environmental cues. Many stimuli induce NF-κB transiently, making time-dependent transcriptional outputs a fundamental feature of NF-κB activation. Here we show that NF-κB target genes have distinct kinetic patterns in activated B lymphoma cells. By combining RELA binding, RNA polymerase II (Pol II) recruitment, and perturbation of NF-κB activation, we demonstrate that kinetic differences amongst early- and late-activated RELA target genes can be understood based on chromatin configuration prior to cell activation and RELA-dependent priming, respectively. We also identified genes that were repressed by RELA activation and others that responded to RELA-activated transcription factors. Cumulatively, our studies define an NF-κB-responsive inducible gene cascade in activated B cells. The nuclear factor kappa B (NF-κB) family of transcription factors regulates cellular responses to a wide variety of environmental cues. These could be extracellular stimuli that activate cell surface receptors, such as pathogens, or intracellular stress signals such as DNA damage or oxidative stress. In response to these triggers, NF-κB proteins accumulate in the cell nucleus, bind to specific DNA sequences in the genome, and thereby modulate gene transcription. Because of the diversity of signals that activate NF-κB and the ubiquity of this pathway in most cell types, cellular outcomes via NF-κB activation must be finely tuned to respond to the initiating stimulus. One mechanism by which NF-κB-dependent gene expression is regulated is by varying the duration of nuclear NF-κB; some signals lead to persistent nuclear NF-κB, while others lead to transient nuclear NF-κB. Consequently, time dependency of transcriptional responses is a unique signature of the initiating stimulus. Here we probed mechanisms that generate kinetic patterns of NF-κB-dependent gene expression in B lymphoma cells responding to a transient NF-κB-activating stimulus. By genetically manipulating NF-κB induction, we identified direct targets of RELA, a member of the NF-κB family, and provide evidence that kinetic patterns are established by a combination of factors that include the chromatin state of genes prior to cell activation and cofactors that work with RELA.
Collapse
Affiliation(s)
- Mingming Zhao
- Gene Regulation Section, Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Jaimy Joy
- Gene Regulation Section, Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Weiqiang Zhou
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Supriyo De
- Gene Expression and Genomics Unit, Laboratory of Genetics and Genomics, National Institute on Aging, Baltimore, Maryland, United States of America
| | - William H. Wood
- Gene Expression and Genomics Unit, Laboratory of Genetics and Genomics, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Kevin G. Becker
- Gene Expression and Genomics Unit, Laboratory of Genetics and Genomics, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Ranjan Sen
- Gene Regulation Section, Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
42
|
Nikopoulou C, Panagopoulos G, Sianidis G, Psarra E, Ford E, Thanos D. The Transcription Factor ThPOK Orchestrates Stochastic Interchromosomal Interactions Required for IFNB1 Virus-Inducible Gene Expression. Mol Cell 2018; 71:352-361.e5. [PMID: 30017585 DOI: 10.1016/j.molcel.2018.06.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 04/30/2018] [Accepted: 06/07/2018] [Indexed: 02/01/2023]
Abstract
Virus infection induces stochastic activation of the interferon-β gene. Three previously identified Alu-like DNA elements called NRCs (NF-κB reception centers) function by capturing and delivering NF-κB to the IFNB1 enhancer via stochastic interchromosomal interactions. We show that the transcription factor ThPOK binds cooperatively with NF-κB to NRCs and mediates their physical proximity with the IFNB1 gene via its ability to oligomerize when bound to DNA. ThPOK knockdown significantly decreased the frequency of interchromosomal interactions, NF-κB DNA binding to the IFNB1 enhancer, and virus-induced IFNB1 gene activation. We also demonstrate that cooperative DNA binding between ThPOK and NF-κB on the same face of the double DNA helix is required for interchromosomal interactions and distinguishes NRCs from various other Alu elements bearing κB sites. These studies show how DNA binding cooperativity of stereospecifically aligned transcription factors provides the necessary ultrasensitivity for the all-or-none stochastic cell responses to virus infection.
Collapse
Affiliation(s)
- Chrysa Nikopoulou
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Efessiou Street, Athens 11527, Greece
| | - Giorgos Panagopoulos
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Efessiou Street, Athens 11527, Greece
| | - Georgios Sianidis
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Efessiou Street, Athens 11527, Greece
| | - Eleni Psarra
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Efessiou Street, Athens 11527, Greece
| | - Ethan Ford
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Efessiou Street, Athens 11527, Greece
| | - Dimitris Thanos
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Efessiou Street, Athens 11527, Greece.
| |
Collapse
|
43
|
Borghini L, Lu J, Hibberd M, Davila S. Variation in Genome-Wide NF-κB RELA Binding Sites upon Microbial Stimuli and Identification of a Virus Response Profile. THE JOURNAL OF IMMUNOLOGY 2018; 201:1295-1305. [PMID: 29959281 DOI: 10.4049/jimmunol.1800246] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 06/04/2018] [Indexed: 12/22/2022]
Abstract
NF-κB transcription factors are master regulators of the innate immune response. Activated downstream of pathogen recognition receptors, they regulate the expression of genes to help fight infections as well as recruit the adaptive immune system. NF-κB responds to a wide variety of signals, but the processes by which stimulus specificity is attained remain unclear. In this article, we characterized the response of one NF-κB member, RELA, to four stimuli mimicking infection in human nasopharyngeal epithelial cells. Comparing genome-wide RELA binding, we observed stimulus-specific sites, although most sites overlapped across stimuli. Specifically, the response to poly I:C (mimicking viral dsRNA and signaling through TLR3) induced a distinct RELA profile, binding in the vicinity of antiviral genes and correlating with corresponding gene expression. This group of binding sites was also enriched in IFN regulatory factor motifs and showed overlapping with IFN regulatory factor binding sites. A novel NF-κB target, OASL, was further validated and showed TLR3-specific activation. This work showed that some RELA DNA binding sites varied in activation response following different stimulations and that interaction with more specialized factors could help achieve this stimulus-specific activity. Our data provide a genomic view of regulated host response to different pathogen stimuli.
Collapse
Affiliation(s)
- Lisa Borghini
- Department of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore; .,Department of Infectious Diseases, Genome Institute of Singapore, Singapore 138672, Singapore; and
| | - Jinhua Lu
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine and Immunology Program, National University of Singapore, Singapore 119077
| | - Martin Hibberd
- Department of Infectious Diseases, Genome Institute of Singapore, Singapore 138672, Singapore; and
| | - Sonia Davila
- Department of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore;
| |
Collapse
|
44
|
Mulero MC, Shahabi S, Ko MS, Schiffer JM, Huang DB, Wang VYF, Amaro RE, Huxford T, Ghosh G. Protein Cofactors Are Essential for High-Affinity DNA Binding by the Nuclear Factor κB RelA Subunit. Biochemistry 2018; 57:2943-2957. [PMID: 29708732 DOI: 10.1021/acs.biochem.8b00158] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Transcription activator proteins typically contain two functional domains: a DNA binding domain (DBD) that binds to DNA with sequence specificity and an activation domain (AD) whose established function is to recruit RNA polymerase. In this report, we show that purified recombinant nuclear factor κB (NF-κB) RelA dimers bind specific κB DNA sites with an affinity significantly lower than that of the same dimers from nuclear extracts of activated cells, suggesting that additional nuclear cofactors might facilitate DNA binding by the RelA dimers. Additionally, recombinant RelA binds DNA with relatively low affinity at a physiological salt concentration in vitro. The addition of p53 or RPS3 (ribosomal protein S3) increases RelA:DNA binding affinity 2- to >50-fold depending on the protein and ionic conditions. These cofactor proteins do not form stable ternary complexes, suggesting that they stabilize the RelA:DNA complex through dynamic interactions. Surprisingly, the RelA-DBD alone fails to bind DNA under the same solution conditions even in the presence of cofactors, suggesting an important role of the RelA-AD in DNA binding. Reduced RelA:DNA binding at a physiological ionic strength suggests that multiple cofactors might be acting simultaneously to mitigate the electrolyte effect and stabilize the RelA:DNA complex in vivo. Overall, our observations suggest that the RelA-AD and multiple cofactor proteins function cooperatively to prime the RelA-DBD and stabilize the RelA:DNA complex in cells. Our study provides a mechanism for nuclear cofactor proteins in NF-κB-dependent gene regulation.
Collapse
Affiliation(s)
- Maria Carmen Mulero
- Department of Chemistry & Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093 , United States
| | - Shandy Shahabi
- Department of Chemistry & Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093 , United States
| | - Myung Soo Ko
- Structural Biochemistry Laboratory, Department of Chemistry & Biochemistry , San Diego State University , 5500 Campanile Drive , San Diego , California 92182 , United States
| | - Jamie M Schiffer
- Department of Chemistry & Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093 , United States
| | - De-Bin Huang
- Department of Chemistry & Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093 , United States
| | - Vivien Ya-Fan Wang
- Faculty of Health Sciences , University of Macau , Avenida da Universidade , Taipa , Macau SAR , China
| | - Rommie E Amaro
- Department of Chemistry & Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093 , United States
| | - Tom Huxford
- Structural Biochemistry Laboratory, Department of Chemistry & Biochemistry , San Diego State University , 5500 Campanile Drive , San Diego , California 92182 , United States
| | - Gourisankar Ghosh
- Department of Chemistry & Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093 , United States
| |
Collapse
|
45
|
Wu J, Wang Q, Dai W, Wang W, Yue M, Wang J. Massive GGAAs in genomic repetitive sequences serve as a nuclear reservoir of NF-κB. J Genet Genomics 2018; 45:193-203. [PMID: 29748061 DOI: 10.1016/j.jgg.2018.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 12/01/2022]
Abstract
Nuclear factor κB (NF-κB) is a DNA-binding transcription factor. Characterizing its genomic binding sites is crucial for understanding its gene regulatory function and mechanism in cells. This study characterized the binding sites of NF-κB RelA/p65 in the tumor neurosis factor-α (TNFα) stimulated HeLa cells by a precise chromatin immunoprecipitation-sequencing (ChIP-seq). The results revealed that NF-κB binds nontraditional motifs (nt-motifs) containing conserved GGAA quadruplet. Moreover, nt-motifs mainly distribute in the peaks nearby centromeres that contain a larger number of repetitive elements such as satellite, simple repeats and short interspersed nuclear elements (SINEs). This intracellular binding pattern was then confirmed by the in vitro detection, indicating that NF-κB dimers can bind the nontraditional κB (nt-κB) sites with low affinity. However, this binding hardly activates transcription. This study thus deduced that NF-κB binding nt-motifs may realize functions other than gene regulation as NF-κB binding traditional motifs (t-motifs). To testify the deduction, many ChIP-seq data of other cell lines were then analyzed. The results indicate that NF-κB binding nt-motifs is also widely present in other cells. The ChIP-seq data analysis also revealed that nt-motifs more widely distribute in the peaks with low-fold enrichment. Importantly, it was also found that NF-κB binding nt-motifs is mainly present in the resting cells, whereas NF-κB binding t-motifs is mainly present in the stimulated cells. Astonishingly, no known function was enriched by the gene annotation of nt-motif peaks. Based on these results, this study proposed that the nt-κB sites that extensively distribute in larger numbers of repeat elements function as a nuclear reservoir of NF-κB. The nuclear NF-κB proteins stored at nt-κB sites in the resting cells may be recruited to the t-κB sites for regulating its target genes upon stimulation.
Collapse
Affiliation(s)
- Jian Wu
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Qiao Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Wei Dai
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Wei Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Ming Yue
- Department of Infectious Diseases, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210002, China
| | - Jinke Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China.
| |
Collapse
|
46
|
Lu HJ, Jin PY, Tang Y, Fan SH, Zhang ZF, Wang F, Wu DM, Lu J, Zheng YL. microRNA-136 inhibits proliferation and promotes apoptosis and radiosensitivity of cervical carcinoma through the NF-κB pathway by targeting E2F1. Life Sci 2018; 199:167-178. [DOI: 10.1016/j.lfs.2018.02.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 02/12/2018] [Accepted: 02/12/2018] [Indexed: 12/25/2022]
|
47
|
Khoyratty TE, Udalova IA. Diverse mechanisms of IRF5 action in inflammatory responses. Int J Biochem Cell Biol 2018; 99:38-42. [PMID: 29578052 DOI: 10.1016/j.biocel.2018.03.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/12/2018] [Accepted: 03/16/2018] [Indexed: 10/25/2022]
Abstract
Interferon regulatory factor 5 (IRF5) is a key signal-dependent transcription factor in myeloid cells. Its expression is induced by granulocyte-macrophage colony stimulating factor and interferon-gamma. IRF5 protein is further activated in response to stimulation, translocating to the nucleus where it mediates inflammatory responses. IRF5 is capable of both the up-regulation of pro-inflammatory genes and repressing anti-inflammatory mediators, thus polarising macrophages to a pro-inflammatory phenotype. We discuss IRF5 interactions with a wide range of transcriptional regulators that give rise to its diverse effects at the level of chromatin.
Collapse
Affiliation(s)
- Tariq E Khoyratty
- The Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, United Kingdom
| | - Irina A Udalova
- The Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, United Kingdom.
| |
Collapse
|
48
|
Riedlinger T, Haas J, Busch J, van de Sluis B, Kracht M, Schmitz ML. The Direct and Indirect Roles of NF-κB in Cancer: Lessons from Oncogenic Fusion Proteins and Knock-in Mice. Biomedicines 2018; 6:biomedicines6010036. [PMID: 29562713 PMCID: PMC5874693 DOI: 10.3390/biomedicines6010036] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 12/24/2022] Open
Abstract
NF-κB signaling pathways play an important role in the regulation of cellular immune and stress responses. Aberrant NF-κB activity has been implicated in almost all the steps of cancer development and many of the direct and indirect contributions of this transcription factor system for oncogenesis were revealed in the recent years. The indirect contributions affect almost all hallmarks and enabling characteristics of cancer, but NF-κB can either promote or antagonize these tumor-supportive functions, thus prohibiting global NF-κB inhibition. The direct effects are due to mutations of members of the NF-κB system itself. These mutations typically occur in upstream components that lead to the activation of NF-κB together with further oncogenesis-promoting signaling pathways. In contrast, mutations of the downstream components, such as the DNA-binding subunits, contribute to oncogenic transformation by affecting NF-κB-driven transcriptional output programs. Here, we discuss the features of recently identified oncogenic RelA fusion proteins and the characterization of pathways that are regulating the transcriptional activity of NF-κB by regulatory phosphorylations. As NF-κB’s central role in human physiology prohibits its global inhibition, these auxiliary or cell type-specific NF-κB regulating pathways are potential therapeutic targets.
Collapse
Affiliation(s)
- Tabea Riedlinger
- Institute of Biochemistry, Justus-Liebig-University, D-35392 Giessen, Germany.
| | - Jana Haas
- Institute of Biochemistry, Justus-Liebig-University, D-35392 Giessen, Germany.
| | - Julia Busch
- Institute of Biochemistry, Justus-Liebig-University, D-35392 Giessen, Germany.
| | - Bart van de Sluis
- Department of Pediatrics, Molecular Genetics Section, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| | - Michael Kracht
- Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University, D-35392 Giessen, Germany.
| | - M Lienhard Schmitz
- Institute of Biochemistry, Justus-Liebig-University, D-35392 Giessen, Germany.
| |
Collapse
|
49
|
Zhang H, Li HS, Hillmer EJ, Zhao Y, Chrisikos TT, Hu H, Wu X, Thompson EJ, Clise-Dwyer K, Millerchip KA, Wei Y, Puebla-Osorio N, Kaushik S, Santos MA, Wang B, Garcia-Manero G, Wang J, Sun SC, Watowich SS. Genetic rescue of lineage-balanced blood cell production reveals a crucial role for STAT3 antiinflammatory activity in hematopoiesis. Proc Natl Acad Sci U S A 2018; 115:E2311-E2319. [PMID: 29463696 PMCID: PMC5878002 DOI: 10.1073/pnas.1713889115] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Blood cell formation must be appropriately maintained throughout life to provide robust immune function, hemostasis, and oxygen delivery to tissues, and to prevent disorders that result from over- or underproduction of critical lineages. Persistent inflammation deregulates hematopoiesis by damaging hematopoietic stem and progenitor cells (HSPCs), leading to elevated myeloid cell output and eventual bone marrow failure. Nonetheless, antiinflammatory mechanisms that protect the hematopoietic system are understudied. The transcriptional regulator STAT3 has myriad roles in HSPC-derived populations and nonhematopoietic tissues, including a potent antiinflammatory function in differentiated myeloid cells. STAT3 antiinflammatory activity is facilitated by STAT3-mediated transcriptional repression of Ube2n, which encodes the E2 ubiquitin-conjugating enzyme Ubc13 involved in proinflammatory signaling. Here we demonstrate a crucial role for STAT3 antiinflammatory activity in preservation of HSPCs and lineage-balanced hematopoiesis. Conditional Stat3 removal from the hematopoietic system led to depletion of the bone marrow lineage- Sca-1+ c-Kit+ CD150+ CD48- HSPC subset (LSK CD150+ CD48- cells), myeloid-skewed hematopoiesis, and accrual of DNA damage in HSPCs. These responses were accompanied by intrinsic transcriptional alterations in HSPCs, including deregulation of inflammatory, survival and developmental pathways. Concomitant Ube2n/Ubc13 deletion from Stat3-deficient hematopoietic cells enabled lineage-balanced hematopoiesis, mitigated depletion of bone marrow LSK CD150+ CD48- cells, alleviated HSPC DNA damage, and corrected a majority of aberrant transcriptional responses. These results indicate an intrinsic protective role for STAT3 in the hematopoietic system, and suggest that this is mediated by STAT3-dependent restraint of excessive proinflammatory signaling via Ubc13 modulation.
Collapse
Affiliation(s)
- Huiyuan Zhang
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Haiyan S Li
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Emily J Hillmer
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Yang Zhao
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Taylor T Chrisikos
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030
| | - Hongbo Hu
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Xiao Wu
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Erika J Thompson
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Karen Clise-Dwyer
- Department of Stem Cell Transplantation Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Karen A Millerchip
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Yue Wei
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Nahum Puebla-Osorio
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Saakshi Kaushik
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Margarida A Santos
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Bin Wang
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | | | - Jing Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Shao-Cong Sun
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030
| | - Stephanie S Watowich
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030;
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030
| |
Collapse
|
50
|
Mulero MC, Huang DB, Nguyen HT, Wang VYF, Li Y, Biswas T, Ghosh G. DNA-binding affinity and transcriptional activity of the RelA homodimer of nuclear factor κB are not correlated. J Biol Chem 2017; 292:18821-18830. [PMID: 28935669 DOI: 10.1074/jbc.m117.813980] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/15/2017] [Indexed: 01/05/2023] Open
Abstract
The nuclear factor κB (NF-κB) transcription factor family regulates genes involved in cell proliferation and inflammation. The promoters of these genes often contain NF-κB-binding sites (κB sites) arranged in tandem. How NF-κB activates transcription through these multiple sites is incompletely understood. We report here an X-ray crystal structure of homodimers comprising the RelA DNA-binding domain containing the Rel homology region (RHR) in NF-κB bound to an E-selectin promoter fragment with tandem κB sites. This structure revealed that two dimers bind asymmetrically to the symmetrically arranged κB sites at which multiple cognate contacts between one dimer to the corresponding DNA are broken. Because simultaneous RelA-RHR dimer binding to tandem sites in solution was anti-cooperative, we inferred that asymmetric RelA-RHR binding with fewer contacts likely indicates a dissociative binding mode. We found that both κB sites are essential for reporter gene activation by full-length RelA homodimer, suggesting that dimers facilitate DNA binding to each other even though their stable co-occupation is not promoted. Promoter variants with altered spacing and orientation of tandem κB sites displayed unexpected reporter activities that were not explained by the solution-binding pattern of RelA-RHR. Remarkably, full-length RelA bound all DNAs with a weaker affinity and specificity. Moreover, the transactivation domain played a negative role in DNA binding. These observations suggest that other nuclear factors influence full-length RelA binding to DNA by neutralizing the transactivation domain negative effect. We propose that DNA binding by NF-κB dimers is highly complex and modulated by facilitated association-dissociation processes.
Collapse
Affiliation(s)
- Maria Carmen Mulero
- From the Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093 and
| | - De-Bin Huang
- From the Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093 and
| | - H Thien Nguyen
- From the Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093 and
| | - Vivien Ya-Fan Wang
- From the Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093 and.,the Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Yidan Li
- From the Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093 and
| | - Tapan Biswas
- From the Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093 and
| | - Gourisankar Ghosh
- From the Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093 and
| |
Collapse
|