Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Infect Immun ; 91(1): e0032222, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36533917

ABSTRACT

Helicobacter pylori colonizes half of the world's population and is responsible for a significant disease burden by causing gastritis, peptic ulcers, and gastric cancer. The development of host inflammation drives these diseases, but there are still open questions in the field about how H. pylori controls this process. We characterized H. pylori inflammation using an 8-month mouse infection time course and comparison of the wild type (WT) and a previously identified mutant lacking the TlpA chemoreceptor that causes elevated inflammation. Our work shows that H. pylori chronic-stage corpus inflammation undergoes surprising fluctuations, with changes in Th17 and eosinophil numbers. The H. pylori tlpA mutant changed the inflammation temporal characteristics, resulting in different inflammation from the wild type at some time points. tlpA mutants have equivalent total and gland colonization in late-stage infections. During early infection, in contrast, they show elevated gland and total colonization compared to those by WT. Our results suggest the chronic inflammation setting is dynamic and may be influenced by colonization properties of early infection.


Subject(s)
Gastritis , Helicobacter Infections , Helicobacter pylori , Animals , Mice , Helicobacter pylori/genetics , Chemotaxis , Bacterial Proteins/genetics , Inflammation , Gastric Mucosa
2.
Elife ; 102021 05 28.
Article in English | MEDLINE | ID: mdl-34047695

ABSTRACT

Determining the layers of gene regulation within the innate immune response is critical to our understanding of the cellular responses to infection and dysregulation in disease. We identified a conserved mechanism of gene regulation in human and mouse via changes in alternative first exon (AFE) usage following inflammation, resulting in changes to the isoforms produced. Of these AFE events, we identified 95 unannotated transcription start sites in mice using a de novo transcriptome generated by long-read native RNA-sequencing, one of which is in the cytosolic receptor for dsDNA and known inflammatory inducible gene, Aim2. We show that this unannotated AFE isoform of Aim2 is the predominant isoform expressed during inflammation and contains an iron-responsive element in its 5'UTR enabling mRNA translation to be regulated by iron levels. This work highlights the importance of examining alternative isoform changes and translational regulation in the innate immune response and uncovers novel regulatory mechanisms of Aim2.


Subject(s)
Alternative Splicing , DNA-Binding Proteins/genetics , Exons , Immunity, Innate/genetics , Inflammation/genetics , Macrophages/metabolism , 5' Untranslated Regions , Animals , Cells, Cultured , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Humans , Inflammation/immunology , Inflammation/metabolism , Macrophages/immunology , Mice , Promoter Regions, Genetic , Transcriptome
3.
Cell Rep ; 33(13): 108541, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33378675

ABSTRACT

Macrophages are critical effector cells of the immune system, and understanding genes involved in their viability and function is essential for gaining insights into immune system dysregulation during disease. We use a high-throughput, pooled-based CRISPR-Cas screening approach to identify essential genes required for macrophage viability. In addition, we target 3' UTRs to gain insights into previously unidentified cis-regulatory regions that control these essential genes. Next, using our recently generated nuclear factor κB (NF-κB) reporter line, we perform a fluorescence-activated cell sorting (FACS)-based high-throughput genetic screen and discover a number of previously unidentified positive and negative regulators of the NF-κB pathway. We unravel complexities of the TNF signaling cascade, showing that it can function in an autocrine manner in macrophages to negatively regulate the pathway. Utilizing a single complex library design, we are capable of interrogating various aspects of macrophage biology, thus generating a resource for future studies.


Subject(s)
Flow Cytometry/methods , High-Throughput Screening Assays/methods , Inflammation/genetics , Inflammation/metabolism , Macrophages/physiology , NF-kappa B/physiology , Tumor Necrosis Factor-alpha/physiology , 3' Untranslated Regions , Animals , CRISPR-Cas Systems , Cell Line , Cell Survival , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Expression Regulation , HEK293 Cells , Humans , Mice , RNA, Guide, Kinetoplastida/genetics , Signal Transduction
4.
Biochim Biophys Acta Gene Regul Mech ; 1863(4): 194419, 2020 04.
Article in English | MEDLINE | ID: mdl-31487549

ABSTRACT

Next-generation sequencing has provided a more complete picture of the composition of the human transcriptome indicating that much of the "blueprint" is a vastness of poorly understood non-protein-coding transcripts. This includes a newly identified class of genes called long noncoding RNAs (lncRNAs). The lack of sequence conservation for lncRNAs across species meant that their biological importance was initially met with some skepticism. LncRNAs mediate their functions through interactions with proteins, RNA, DNA, or a combination of these. Their functions can often be dictated by their localization, sequence, and/or secondary structure. Here we provide a review of the approaches typically adopted to study the complexity of these genes with an emphasis on recent discoveries within the innate immune field. Finally, we discuss the challenges, as well as the emergence of new technologies that will continue to move this field forward and provide greater insight into the biological importance of this class of genes. This article is part of a Special Issue entitled: ncRNA in control of gene expression edited by Kotb Abdelmohsen.


Subject(s)
Immunity, Innate/genetics , RNA, Long Noncoding/metabolism , Animals , Gene Expression Regulation , Humans , Mice , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/genetics
5.
Nat Commun ; 10(1): 338, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30659183

ABSTRACT

Macrophages undergo metabolic changes during activation that are coupled to functional responses. The gram negative bacterial product lipopolysaccharide (LPS) is especially potent at driving metabolic reprogramming, enhancing glycolysis and altering the Krebs cycle. Here we describe a role for the citrate-derived metabolite malonyl-CoA in the effect of LPS in macrophages. Malonylation of a wide variety of proteins occurs in response to LPS. We focused on one of these, glyceraldehyde-3-phosphate dehydrogenase (GAPDH). In resting macrophages, GAPDH binds to and suppresses translation of several inflammatory mRNAs, including that encoding TNFα. Upon LPS stimulation, GAPDH undergoes malonylation on lysine 213, leading to its dissociation from TNFα mRNA, promoting translation. We therefore identify for the first time malonylation as a signal, regulating GAPDH mRNA binding to promote inflammation.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Inflammation Mediators/pharmacology , Inflammation/metabolism , Macrophages/drug effects , Macrophages/metabolism , Animals , Cytokines/metabolism , HEK293 Cells , Humans , Lipopolysaccharides/pharmacology , Lysine/metabolism , Malonyl Coenzyme A/metabolism , Mice, Inbred C57BL , Mutagenesis , Polyribosomes , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Cell Rep ; 25(6): 1511-1524.e6, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30404006

ABSTRACT

An inducible gene expression program is a hallmark of the host inflammatory response. Recently, long intergenic non-coding RNAs (lincRNAs) have been shown to regulate the magnitude, duration, and resolution of these responses. Among these is lincRNA-Cox2, a dynamically regulated gene that broadly controls immune gene expression. To evaluate the in vivo functions of this lincRNA, we characterized multiple models of lincRNA-Cox2-deficient mice. LincRNA-Cox2-deficient macrophages and murine tissues had altered expression of inflammatory genes. Transcriptomic studies from various tissues revealed that deletion of the lincRNA-Cox2 locus also strongly impaired the basal and inducible expression of the neighboring gene prostaglandin-endoperoxide synthase (Ptgs2), encoding cyclooxygenase-2, a key enzyme in the prostaglandin biosynthesis pathway. By utilizing different genetic manipulations in vitro and in vivo, we found that lincRNA-Cox2 functions through an enhancer RNA mechanism to regulate Ptgs2. More importantly, lincRNA-Cox2 also functions in trans, independently of Ptgs2, to regulate critical innate immune genes in vivo.


Subject(s)
Cyclooxygenase 2/metabolism , Immunity/genetics , Models, Genetic , RNA, Long Noncoding/metabolism , Animals , Enhancer Elements, Genetic/genetics , Gene Deletion , Gene Expression Regulation , HEK293 Cells , Humans , Lipopolysaccharides/pharmacology , Lung/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , RNA/metabolism , RNA Splicing/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spleen/metabolism , Transcription, Genetic
7.
J Biol Chem ; 292(51): 20911-20920, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29051223

ABSTRACT

The innate immune system protects against infections by initiating an inducible inflammatory response. NF-κB is one of the critical transcription factors controlling this complex response, but some aspects of its regulation remain unclear. For example, although long non-coding RNAs (lncRNAs) have been shown to critically regulate gene expression, only a fraction of these have been functionally characterized, and the extent to which lncRNAs control NF-κB expression is unknown. Here, we describe the generation of a GFP-based NF-κB reporter system in immortalized murine bone marrow-derived macrophages (iBMDM). Activation of this reporter, using Toll-like receptor ligands, resulted in GFP expression, which could be monitored by flow cytometry. We also established a CRISPR/Cas9 gene deletion system in this NF-κB reporter line, enabling us to screen for genes that regulate NF-κB signaling. Our deletion-based approach identified two long intergenic non-coding(linc)RNAs, lincRNA-Cox2 and lincRNA-AK170409, that control NF-κB signaling. We demonstrate a potential novel role for lincRNA-Cox2 in promoting IκBα degradation in the cytoplasm. For lincRNA-AK170409, we provide evidence that this nuclearly-localized lincRNA regulates a number of inflammation-related genes. In conclusion, we have established an NF-κB-GFP iBMDM reporter cell line and a line that stably expresses Cas9. Our approach enabled the identification of lincRNA-Cox2 and lincRNA-AK170409 as NF-κB regulators, and this tool will be useful for identifying additional genes involved in regulating this transcription factor critical for immune function.


Subject(s)
Macrophages/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , RNA, Long Noncoding/genetics , Animals , CRISPR-Cas Systems , Cells, Cultured , Cyclooxygenase 2/genetics , Gene Knockout Techniques , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunity, Innate/genetics , Macrophages/immunology , Mice , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...