The NF-κB-dependent and -independent transcriptome and chromatin landscapes of human coronavirus 229E-infected cells

التفاصيل البيبلوغرافية
العنوان: The NF-κB-dependent and -independent transcriptome and chromatin landscapes of human coronavirus 229E-infected cells
المؤلفون: Marek Bartkuhn, Michael Kracht, Sabin Bhuju, Nadja Karl, Knut Beuerlein, John Ziebuhr, Michael Poppe, Helmut Müller, Liane Jurida, M. Lienhard Schmitz, Sascha Wittig, Jochen Wilhelm
المساهمون: Helmholtz-Zentrum für Infektionsforschung GmbH, Inhoffenstr.7, 38124 Braunschweig, Germany.
المصدر: PLoS Pathogens, Vol 13, Iss 3, p e1006286 (2017)
PLOS Pathogens
PLoS Pathogens
بيانات النشر: Public Library of Science (PLoS), 2017.
سنة النشر: 2017
مصطلحات موضوعية: 0301 basic medicine, Gene Expression, Biochemistry, Polymerase Chain Reaction, 0302 clinical medicine, Coronavirus 229E, Human, Gene expression, Transcriptional regulation, Post-Translational Modification, Phosphorylation, Biology (General), Oligonucleotide Array Sequence Analysis, Regulation of gene expression, biology, Chromosome Biology, Microbial Genetics, NF-kappa B, Chromatin, 030220 oncology & carcinogenesis, Host cell cytoplasm, Viral Genetics, Epigenetics, Coronavirus Infections, Research Article, Chromatin Immunoprecipitation, Human coronavirus 229E, QH301-705.5, Immunology, DNA transcription, Immunoblotting, Laser Capture Microdissection, Microbiology, Cell Line, 03 medical and health sciences, Virology, Genetics, Humans, Gene Regulation, Enhancer, Molecular Biology, Transcription factor, Host Cells, Biology and Life Sciences, Proteins, Cell Biology, RC581-607, biology.organism_classification, Molecular biology, Viral Replication, 030104 developmental biology, Viral Gene Expression, Gene Expression Regulation, Microscopy, Fluorescence, Parasitology, Immunologic diseases. Allergy, Transcriptome, Viral Transmission and Infection
الوصف: Coronavirus replication takes place in the host cell cytoplasm and triggers inflammatory gene expression by poorly characterized mechanisms. To obtain more insight into the signals and molecular events that coordinate global host responses in the nucleus of coronavirus-infected cells, first, transcriptome dynamics was studied in human coronavirus 229E (HCoV-229E)-infected A549 and HuH7 cells, respectively, revealing a core signature of upregulated genes in these cells. Compared to treatment with the prototypical inflammatory cytokine interleukin(IL)-1, HCoV-229E replication was found to attenuate the inducible activity of the transcription factor (TF) NF-κB and to restrict the nuclear concentration of NF-κB subunits by (i) an unusual mechanism involving partial degradation of IKKβ, NEMO and IκBα and (ii) upregulation of TNFAIP3 (A20), although constitutive IKK activity and basal TNFAIP3 expression levels were shown to be required for efficient virus replication. Second, we characterized actively transcribed genomic regions and enhancers in HCoV-229E-infected cells and systematically correlated the genome-wide gene expression changes with the recruitment of Ser5-phosphorylated RNA polymerase II and prototypical histone modifications (H3K9ac, H3K36ac, H4K5ac, H3K27ac, H3K4me1). The data revealed that, in HCoV-infected (but not IL-1-treated) cells, an extensive set of genes was activated without inducible p65 NF-κB being recruited. Furthermore, both HCoV-229E replication and IL-1 were shown to upregulate a small set of genes encoding immunomodulatory factors that bind p65 at promoters and require IKKβ activity and p65 for expression. Also, HCoV-229E and IL-1 activated a common set of 440 p65-bound enhancers that differed from another 992 HCoV-229E-specific enhancer regions by distinct TF-binding motif combinations. Taken together, the study shows that cytoplasmic RNA viruses fine-tune NF-κB signaling at multiple levels and profoundly reprogram the host cellular chromatin landscape, thereby orchestrating the timely coordinated expression of genes involved in multiple signaling, immunoregulatory and metabolic processes.
Author summary Coronaviruses are major human and animal pathogens. They belong to a family of plus-strand RNA viruses that have extremely large genomes and encode a variety of proteins involved in virus-host interactions. The four common coronaviruses (HCoV-229E, NL63, OC43, HKU1) cause mainly upper respiratory tract infections, while zoonotic coronaviruses (SARS-CoV and MERS-CoV) cause severe lung disease, including acute respiratory distress syndrome (ARDS). The molecular basis for this fundamentally different pathology is incompletely understood. Our study provides a genome-wide investigation of epigenetic changes occurring in response to HCoV-229E. We identify at high resolution a large number of regulatory regions in the genome of infected cells that coordinate de novo gene transcription. Many of these genes have immunomodulatory functions and, most likely, contribute to limiting viral replication, while other factors may promote viral replication. The study provides an intriguing example of a virus that completes its entire life cycle in the cytoplasm while sending multiple signals to the nuclear chromatin compartment to adjust the host cell repertoire of transcribed genes. The approach taken in this study is expected to provide a suitable framework for future studies aimed at dissecting and comparing host responses to representative coronaviruses with different pathogenic potential in humans.
اللغة: English
تدمد: 1553-7374
1553-7366
URL الوصول: https://explore.openaire.eu/search/publication?articleId=doi_dedup___::e4b9e241c3a888a88fd684c8bddaca1d
https://doaj.org/article/5360acc7ff904d7fbc39af9d12d4e187
حقوق: OPEN
رقم الأكسشن: edsair.doi.dedup.....e4b9e241c3a888a88fd684c8bddaca1d
قاعدة البيانات: OpenAIRE