We have previously shown that Mkp-1deficient mice produce elevated TNF-a, IL-6, and IL-10 following systemic Escherichia coli infection, and they exhibited increased mortality, elevated bacterial burden, and profound metabolic alterations. To understand the function of Mkp-1 during bacterial infection, we performed RNA-sequencing analysis to compare the global gene expression between E. coliinfected wild-type and Mkp-1 2/2 mice. A large number of IFN-stimulated genes were more robustly expressed in E. coliinfected Mkp-1 2/2 mice than in wild-type mice. Multiplex analysis of the serum cytokine levels revealed profound increases in IFN-b, IFN-g, TNF-a, IL-1a and b, IL-6, IL-10, IL-17A, IL-27, and GMSF levels in E. coliinfected Mkp-1 2/2 mice relative to wildtype mice. Administration of a neutralizing Ab against the receptor for type I IFN to Mkp-1 2/2 mice prior to E. coli infection augmented mortality and disease severity. Mkp-1 2/2 bone marrowderived macrophages (BMDM) produced higher levels of IFN-b mRNA and protein than did wild-type BMDM upon treatment with LPS, E. coli, polyinosinic:polycytidylic acid, and herring sperm DNA. Augmented IFN-b induction in Mkp-1 2/2 BMDM was blocked by a p38 inhibitor but not by an JNK inhibitor. Enhanced Mkp-1 expression abolished IFN-b induction by both LPS and E. coli but had little effect on the IFN-b promoter activity in LPSstimulated RAW264.7 cells. Mkp-1 deficiency did not have an overt effect on IRF3/7 phosphorylation or IKK activation but modestly enhanced IFN-b mRNA stability in LPS-stimulated BMDM. Our results suggest that Mkp-1 regulates IFN-b production primarily through a p38-mediated mechanism and that IFN-b plays a beneficial role in E. coliinduced sepsis.
Mkp-1 is a critical negative regulator of the inflammatory response and plays a crucial role for bacterial clearance in an animal model of sepsis. Mkp-1−/− mice exhibit augmented inflammation, increased bacterial burden, elevated organ damage, and increased mortality relative to Mkp-1+/+ mice following systemic E. coli infection. To understand the immunological functions of Mkp-1, we performed RNA-seq analysis with mouse livers. Under normal conditions, 357 genes were differentially expressed between Mkp-1+/+ and Mkp-1−/− mice. However, following E. coli infection > 5,400 genes displayed differential expression between the two groups. Among genes upregulated by E. coli infection and exacerbated by Mkp-1 knockout are IL-6, IL-10, as well as a large number of IFN-inducible genes including Mx1/2, Gbp2/3, Isg15, Usp18, Ifitm1-3, STAT3, and immune checkpoint protein PD-L1. qRT-PCR and Western blot analyses verified E. coli infection-induced PD-L1 expression and further exacerbation in Mkp-1−/− mice. Examination of the RNA-seq dataset also revealed a clear interferon signature. ELISA analyses on the serum revealed low IFN-β levels in uninfected Mkp-1+/+ and Mkp-1−/− mice, increased IFN-β levels in E. coli-infected Mkp-1+/+ mice, and further augmented IFN-β levels in E. coli-infected Mkp-1−/− mice. Similarly, Mkp-1−/− bone marrow-derived macrophages also produced more IFN-β than Mkp-1+/+ macrophages following stimulation with either LPS or heat-killed E. coli. Analysis of the signaling pathways critical for IFN-β expression indicates a marked increase in TBK1 activity. Our studies suggest that exaggerated expression of type I interferons and PD-L1 contribute to the phenotypes of Mkp-1−/− mice in the model of sepsis.
Pde3B hydrolyzes both cAMP and cGMP, important second messengers in a variety of cellular processes. Pde3B has been shown to regulate NLRP3 inflammasome activity in adipose tissue. To understand whether Pde3B plays a role in the macrophage response to bacterial infection, we derived macrophages from the bone marrow of wildtype and Pde3B knockout mice. Macrophages were then stimulated with LPS for 6 h or left untreated, and total RNA was prepared for RNA-seq analyses. Our data indicate that Pde3B expression was inhibited by LPS in macrophages. Knockout of Pde3B resulted in significant changes in the expression of a large group of genes: 427 genes were up-regulated by at least 2-fold while 428 genes were down-regulated by more than 50% in LPS stimulated macrophages as the result of Pde3B deficiency. Gene profiling indicates that a number of biological processes are affected by Pde3B knockout, including cellular ion/cation homeostasis, immune system processes, and chemotaxis. Interestingly, Pde3B deficiency disturbed the expression of several chemokines: Cxcl-1, -2, and -3 expression were decreased by 96, 60, and 75%, respectively. In contrast, Ccl-4, -7, -12, -22, Cxcl-9 and -l11 expression were enhanced 1.8, 1.5, 1.9, 2, 3, and 3-fold, respectively. Differential expression of dual specificity phosphatase (Dusp)-1, -2, -3, -4, -5, -6, -10, -11, and -16 were also seen between LPS-stimulated wildtype and Pde3B-deficient macrophages. Since the Dusps are important regulators of MAP kinases that control the expression of chemokines, alterations in the expression of Dusps provides a plausible explanation for the changes in chemokine expression. Our studies suggest that Pde3B may regulate chemokine expression via modulating MAP kinases.
scite is a Brooklyn-based organization that helps researchers better discover and understand research articles through Smart Citations–citations that display the context of the citation and describe whether the article provides supporting or contrasting evidence. scite is used by students and researchers from around the world and is funded in part by the National Science Foundation and the National Institute on Drug Abuse of the National Institutes of Health.
customersupport@researchsolutions.com
10624 S. Eastern Ave., Ste. A-614
Henderson, NV 89052, USA
This site is protected by reCAPTCHA and the Google Privacy Policy and Terms of Service apply.
Copyright © 2024 scite LLC. All rights reserved.
Made with 💙 for researchers
Part of the Research Solutions Family.