1
|
Shen Y, Gong L, Xu F, Wang S, Liu H, Wang Y, Hu L, Zhu L. Insight into the lncRNA-mRNA Co-Expression Profile and ceRNA Network in Lipopolysaccharide-Induced Acute Lung Injury. Curr Issues Mol Biol 2023; 45:6170-6189. [PMID: 37504305 PMCID: PMC10378513 DOI: 10.3390/cimb45070389] [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: 05/29/2023] [Revised: 07/16/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) participate in acute lung injury (ALI). However, their latent biological function and molecular mechanism have not been fully understood. In the present study, the global expression profiles of lncRNAs and mRNAs between the control and lipopolysaccharide (LPS)-stimulated groups of human normal lung epithelial cells (BEAS-2B) were determined using high-throughput sequencing. Overall, a total of 433 lncRNAs and 183 mRNAs were differentially expressed. A lncRNA-mRNA co-expression network was established, and then the top 10 lncRNAs were screened using topological methods. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis results showed that the key lncRNAs targeting mRNAs were mostly enriched in the inflammatory-related biological processes. Gene set variation analysis and Pearson's correlation coefficients confirmed the close correlation for the top 10 lncRNAs with inflammatory responses. A protein-protein interaction network analysis was conducted based on the key lncRNAs targeting mRNAs, where IL-1β, IL-6, and CXCL8 were regarded as the hub genes. A competing endogenous RNA (ceRNA) modulatory network was created with five lncRNAs, thirteen microRNAs, and twelve mRNAs. Finally, real-time quantitative reverse transcription-polymerase chain reaction was employed to verify the expression levels of several key lncRNAs in BEAS-2B cells and human serum samples.
Collapse
Affiliation(s)
- Yue Shen
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Linjing Gong
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fan Xu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Sijiao Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hanhan Liu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yali Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lijuan Hu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lei Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Pulmonary and Critical Care Medicine, Huadong Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
2
|
Clustering ICU patients with sepsis based on the patterns of their circulating biomarkers: A secondary analysis of the CAPTAIN prospective multicenter cohort study. PLoS One 2022; 17:e0267517. [PMID: 36301921 PMCID: PMC9612564 DOI: 10.1371/journal.pone.0267517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 10/11/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Although sepsis is a life-threatening condition, its heterogeneous presentation likely explains the negative results of most trials on adjunctive therapy. This study in patients with sepsis aimed to identify subgroups with similar immune profiles and their clinical and outcome correlates. METHODS A secondary analysis used data of a prospective multicenter cohort that included patients with early assessment of sepsis. They were described using Predisposition, Insult, Response, Organ failure sepsis (PIRO) staging system. Thirty-eight circulating biomarkers (27 proteins, 11 mRNAs) were assessed at sepsis diagnosis, and their patterns were determined through principal component analysis (PCA). Hierarchical clustering was used to group the patients and k-means algorithm was applied to assess the internal validity of the clusters. RESULTS Two hundred and three patients were assessed, of median age 64.5 [52.0-77.0] years and SAPS2 score 55 [49-61] points. Five main patterns of biomarkers and six clusters of patients (including 42%, 21%, 17%, 9%, 5% and 5% of the patients) were evidenced. Clusters were distinguished according to the certainty of the causal infection, inflammation, use of organ support, pro- and anti-inflammatory activity, and adaptive profile markers. CONCLUSIONS In this cohort of patients with suspected sepsis, we individualized clusters which may be described with criteria used to stage sepsis. As these clusters are based on the patterns of circulating biomarkers, whether they might help to predict treatment responsiveness should be addressed in further studies. TRIAL REGISTRATION The CAPTAIN study was registered on clinicaltrials.gov on June 22, 2011, # NCT01378169.
Collapse
|
3
|
Wu J, Wei X, Li J, Gan Y, Zhang R, Han Q, Liang P, Zeng Y, Yang Q. Plasma exosomal IRAK1 can be a potential biomarker for predicting the treatment response to renin-angiotensin system inhibitors in patients with IgA nephropathy. Front Immunol 2022; 13:978315. [PMID: 36091017 PMCID: PMC9459338 DOI: 10.3389/fimmu.2022.978315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/09/2022] [Indexed: 11/25/2022] Open
Abstract
Background Renin-angiotensin system inhibitors (RASi) are the first choice and basic therapy for the treatment of IgA nephropathy (IgAN) with proteinuria. However, approximately 40% of patients have no response to RASi treatment. The aim of this study was to screen potential biomarkers for predicting the treatment response of RASi in patients with IgAN. Methods We included IgAN patients who were treatment-naive. They received supportive treatment, including a maximum tolerant dose of RASi for 3 months. According to the degree of decrease in proteinuria after 3 months of follow-up, these patients were divided into a sensitive group and a resistant group. The plasma of the two groups of patients was collected, and the exosomes were extracted for high-throughput sequencing. The screening of hub genes was performed using a weighted gene co-expression network (WGCNA) and filtering differentially expressed genes (DEGs). We randomly selected 20 patients in the sensitive group and 20 patients in the resistant group for hub gene validation by real-time quantitative polymerase chain reaction (qRT−PCR). A receiver operating characteristic (ROC) curve was used to evaluate hub genes that predicted the efficacy of the RASi response among the 40 validation patients. Results After screening 370 IgAN patients according to the inclusion and exclusion criteria and the RASi treatment response evaluation, there were 38 patients in the sensitive group and 32 patients in the resistant group. IRAK1, ABCD1 and PLXNB3 were identified as hub genes by analyzing the high-throughput sequencing of the plasma exosomes of the two groups through WGCNA and DEGs screening. The sequencing data were consistent with the validation data showing that these three hub genes were upregulated in the resistant group compared with the sensitive group. The ROC curve indicated that IRAK1 was a good biomarker to predict the therapeutic response of RASi in patients with IgAN. Conclusions Plasma exosomal IRAK1 can be a potential biomarker for predicting the treatment response of RASi in patients with IgAN.
Collapse
|
4
|
Qin Y, Livingston DH, Spolarics Z. INTERACTIONS BETWEEN BIOLOGICAL SEX AND THE X-LINKED VARIANT IRAK1 HAPLOTYPE IN MODULATING CLINICAL OUTCOME AND CELLULAR PHENOTYPES AFTER TRAUMA. Shock 2022; 58:179-188. [PMID: 35953456 DOI: 10.1097/shk.0000000000001966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Sex-related outcome differences in trauma remain controversial. The mechanisms causing sex-biased outcomes are likely to have hormonal and genetic components, in which X-linked genetic polymorphisms may play distinct roles because of X-linked inheritance, hemizygosity in males, and X chromosome mosaicism in females. The study aimed to elucidate the contribution of biological sex and the common X-linked IRAK1 haplotype to posttrauma clinical complications, inflammatory cytokine and chemokine production, and polymorphonuclear cell and monocyte activation. Postinjury clinical outcome was tested in 1507 trauma patients (1,110 males, 397 females) after stratification by sex or the variant IRAK1 haplotype. Males showed a three- to fivefold greater frequency of posttrauma sepsis, but similar mortality compared to females. Stratification by the variant IRAK1 haplotype revealed increased pneumonia and urinary tract infection in Wild type (WT) versus variant IRAK1 males, whereas increased respiratory failures in variant versus WT females. Cytokine/chemokine profiles were tested in whole blood from a subset of patients (n = 81) and healthy controls (n = 51), which indicated sex-related differences in ex vivo lipopolysaccharide responsiveness manifesting in a 1.5- to 2-fold increased production rate of tumor necrosis factor α, interleukin-1β (IL-1β), IL-10, Macrophage Inflammatory Protein-1 Alpha, and MIP1β in WT male compared to WT female trauma patients. Variant IRAK1 decreased IL-6, IL-8, and interferon gamma-induced protein 10 production in male trauma subjects compared to WT, whereas cytokine/chemokine responses were similar in variant IRAK1 and WT female trauma subjects. Trauma-induced and lipopolysaccharide-stimulated polymorphonuclear cell and monocyte activation determined by using a set of cluster of differentiation markers and flow cytometry were not influenced by sex or variant IRAK1. These findings suggest that variant IRAK1 is a potential contributor to sex-based outcome differences, but its immunomodulatory impacts are modulated by biological sex.
Collapse
Affiliation(s)
- Yong Qin
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, New Jersey
| | | | | |
Collapse
|
5
|
Brinkworth JF, Shaw JG. On race, human variation, and who gets and dies of sepsis. AMERICAN JOURNAL OF BIOLOGICAL ANTHROPOLOGY 2022. [PMCID: PMC9544695 DOI: 10.1002/ajpa.24527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Jessica F. Brinkworth
- Department of Anthropology University of Illinois Urbana‐Champaign Urbana Illinois USA
- Carl R. Woese Institute for Genomic Biology University of Illinois at Urbana‐Champaign Urbana Illinois USA
- Department of Evolution, Ecology and Behavior University of Illinois Urbana‐Champaign Urbana Illinois USA
| | - J. Grace Shaw
- Department of Anthropology University of Illinois Urbana‐Champaign Urbana Illinois USA
- Carl R. Woese Institute for Genomic Biology University of Illinois at Urbana‐Champaign Urbana Illinois USA
| |
Collapse
|
6
|
Wu X, Xu M, Liu Z, Zhang Z, Liu Y, Luo S, Zheng X, Little PJ, Xu S, Weng J. Pharmacological inhibition of IRAK1 and IRAK4 prevents endothelial inflammation and atherosclerosis in ApoE -/- mice. Pharmacol Res 2022; 175:106043. [PMID: 34954030 DOI: 10.1016/j.phrs.2021.106043] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 02/08/2023]
Abstract
Inflammation associated endothelial dysfunction represents a pivotal contributor to atherosclerosis. Increasingly, evidence has demonstrated that interleukin 1 receptor (IL1-R) / toll-like receptor (TLR) signaling participates in the development of atherosclerosis. Recent large-scale clinical trials have supported the therapeutic potential of anti-inflammatory therapies targeting IL-1β and IL-6 in reducing atherosclerosis. The present study examined the pharmacological effects of IL-1R-associated kinase 1 and 4 inhibitors (IRAK1/4i) in regulating inflammation of the endothelium and atherosclerosis. We demonstrate that dual pharmacological inhibition of IRAK1 and IRAK4 by an IRAK1/4i is more effective against LPS induced endothelial inflammation, compared with IRAK1 inhibitor or IRAK4 inhibitor monotherapy. IRAK1/4i showed little endothelial cell toxicity at concentrations from 1 μM up to 10 μM. Inhibition of IRAK1/4 reduced endothelial activation induced by LPS in vitro as evidenced by attenuated monocyte adhesion to the endothelium. Mechanistically, blockade of IRAK1/4 ameliorated the transcriptional activity of NF-κB. To assess the pharmacological effects of IRAK1/4i on atherosclerosis in vivo, ApoE-/- mice were orally administered IRAK1/4i (20 mg/kg/d) for 8 weeks. We show that IRAK1/4i reduced atherosclerotic lesion size in the aortic sinus and increased hepatic LDLR protein levels as well as lowered LDL-C level, without affecting other lipid parameters or glucose tolerance. Taken together, our findings demonstrate that dual pharmacological inhibition of IRAK1 and IRAK4 attenuates endothelial inflammation, lowers LDL-C levels and reduces atherosclerosis. Our study reinforces the evolving standing of anti-inflammatory approaches in cardiovascular therapeutics.
Collapse
Affiliation(s)
- Xiumei Wu
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, 510000 Guangzhou, China
| | - Mengyun Xu
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zhenghong Liu
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zhidan Zhang
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Yujie Liu
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Sihui Luo
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Xueying Zheng
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia
| | - Suowen Xu
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China.
| | - Jianping Weng
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, 510000 Guangzhou, China; Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|
7
|
Genome-Wide Linkage Analysis of the Risk of Contracting a Bloodstream Infection in 47 Pedigrees Followed for 23 Years Assembled From a Population-Based Cohort (the HUNT Study). Crit Care Med 2021; 48:1580-1586. [PMID: 32885941 DOI: 10.1097/ccm.0000000000004520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Bloodstream infection is an important cause of death worldwide. The main objective of this study was to identify genetic loci linked to risk of contracting a bloodstream infection. DESIGN Genome-wide linkage analysis. SETTING Population-based, Norwegian cohort, followed between 1995 and 2017. SUBJECTS Among 69,423 genotyped subjects, there were 47 families with two or more second-degree relatives with bloodstream infection in the follow-up period. There were 365 subjects in these families, of which 110 were affected. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS The cohort was genotyped using Illumina HumanCoreExome (Illumina, San Diego, CA) arrays. Before linkage analysis, single-nucleotide polymorphisms were pruned and clumped. In nonparametric linkage analysis using an exponential model, we found three loci with a suggestive linkage to bloodstream infection, all on chromosome 4, at 46.6 centimorgan (logarithm of odds, 2.3), 57.7 centimorgan (logarithm of odds, 3.2), and 70.0 centimorgan (logarithm of odds, 2.1). At the peak of the lead region are three genes: TLR10, TLR1, and TLR6. CONCLUSIONS Variations in the TLR10/1/6 locus appear to be linked with the risk of contracting a bloodstream infection.
Collapse
|
8
|
Laserna AKC, Lai Y, Fang G, Ganapathy R, Atan MSBM, Lu J, Wu J, Uttamchandani M, Moochhala SM, Li SFY. Metabolic Profiling of a Porcine Combat Trauma-Injury Model Using NMR and Multi-Mode LC-MS Metabolomics-A Preliminary Study. Metabolites 2020; 10:metabo10090373. [PMID: 32948079 PMCID: PMC7570375 DOI: 10.3390/metabo10090373] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 11/16/2022] Open
Abstract
Profiles of combat injuries worldwide have shown that penetrating trauma is one of the most common injuries sustained during battle. This is usually accompanied by severe bleeding or hemorrhage. If the soldier does not bleed to death, he may eventually succumb to complications arising from trauma hemorrhagic shock (THS). THS occurs when there is a deficiency of oxygen reaching the organs due to excessive blood loss. It can trigger massive metabolic derangements and an overwhelming inflammatory response, which can subsequently lead to the failure of organs and possibly death. A better understanding of the acute metabolic changes occurring after THS can help in the development of interventional strategies, as well as lead to the identification of potential biomarkers for rapid diagnosis of hemorrhagic shock and organ failure. In this preliminary study, a metabolomic approach using the complementary platforms of nuclear magnetic resonance (NMR) spectroscopy and liquid chromatography coupled with mass spectrometry (LC-MS) was used to determine the metabolic changes occurring in a porcine model of combat trauma injury comprising of penetrating trauma to a limb with hemorrhagic shock. Several metabolites associated with the acute-phase reaction, inflammation, energy depletion, oxidative stress, and possible renal dysfunction were identified to be significantly changed after a thirty-minute shock period.
Collapse
Affiliation(s)
- Anna Karen Carrasco Laserna
- Department of Chemistry, Faculty of Science, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore; (A.K.C.L.); (G.F.); (M.U.)
| | - Yiyang Lai
- Defence Medical and Environmental Research Institute, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Singapore; (Y.L.); (R.G.); (J.L.); (J.W.)
| | - Guihua Fang
- Department of Chemistry, Faculty of Science, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore; (A.K.C.L.); (G.F.); (M.U.)
- Forensic Science Division, Health Services Authority, 11 Outram Road, Singapore 169078, Singapore
| | - Rajaseger Ganapathy
- Defence Medical and Environmental Research Institute, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Singapore; (Y.L.); (R.G.); (J.L.); (J.W.)
| | | | - Jia Lu
- Defence Medical and Environmental Research Institute, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Singapore; (Y.L.); (R.G.); (J.L.); (J.W.)
| | - Jian Wu
- Defence Medical and Environmental Research Institute, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Singapore; (Y.L.); (R.G.); (J.L.); (J.W.)
| | - Mahesh Uttamchandani
- Department of Chemistry, Faculty of Science, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore; (A.K.C.L.); (G.F.); (M.U.)
- Defence Medical and Environmental Research Institute, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Singapore; (Y.L.); (R.G.); (J.L.); (J.W.)
| | - Shabbir M. Moochhala
- School of Applied Sciences, Temasek Polytechnic, 21 Tampines Ave 1, Singapore 529757, Singapore;
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Blk MD3, 16 Medical Drive, Singapore 117600, Singapore
- Correspondence: (S.M.M.); (S.F.Y.L.); Tel.: +65-6516-2681 (S.F.Y.L.)
| | - Sam Fong Yau Li
- Department of Chemistry, Faculty of Science, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore; (A.K.C.L.); (G.F.); (M.U.)
- NUS Environmental Research Institute, National University of Singapore, T-Lab Building, 5A Engineering Drive 1, Singapore 117411, Singapore
- Correspondence: (S.M.M.); (S.F.Y.L.); Tel.: +65-6516-2681 (S.F.Y.L.)
| |
Collapse
|
9
|
Zhang Y, Li X, Wang C, Zhang M, Yang H, Lv K. lncRNA AK085865 Promotes Macrophage M2 Polarization in CVB3-Induced VM by Regulating ILF2-ILF3 Complex-Mediated miRNA-192 Biogenesis. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 21:441-451. [PMID: 32668391 PMCID: PMC7358220 DOI: 10.1016/j.omtn.2020.06.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/15/2020] [Accepted: 06/18/2020] [Indexed: 12/15/2022]
Abstract
Accumulating evidence indicates that macrophage polarization plays a crucial role in coxsackievirus B3 (CVB3)-induced viral myocarditis (VM). Our previous study demonstrated that long noncoding ribonucleic acid (lncRNA) AK085865 ablation confers susceptibility to VM by regulating macrophage polarization. However, the detailed molecular mechanisms by which AK085865 regulates macrophage polarization remain to be explored. In this study, we found that AK085865 specifically interacts with interleukin enhancer-binding factor 2 (ILF2) and facilitates M2 macrophage polarization by functioning as a negative regulator in the ILF2-ILF3 complex-mediated microRNA (miRNA or miR) processing pathway. miR-192 was downregulated, whereas the levels of pri-miR-192 were significantly increased in bone marrow-derived macrophages (BMDMs) from AK085865-/- mice compared with the BMDMs from wild-type (WT) mice. Conversely, knockdown of ILF2 resulted in elevated levels of mature miR-192 and decreased expression of pri-miR-192 in BMDMs from AK085865-/- mice. Moreover, miR-192 overexpression promoted macrophage M2 polarization in vitro, and interleukin-1 receptor-associated kinase 1 (IRAK1) was identified as a direct target. miR-192 overexpression effectively rescued mice from lethal myocarditis caused by CVB3 infection and switched myocardial-infiltrating macrophages to a predominant M2 phenotype. Collectively, our findings uncover a critical mechanism of AK085865 in the regulation of macrophage polarization in vitro and in vivo and provide a potential, clinically significant therapeutic target.
Collapse
Affiliation(s)
- Yingying Zhang
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes, Wuhu 241001, China; Department of Laboratory Medicine, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu 241001, China
| | - Xueqin Li
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes, Wuhu 241001, China; Department of Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu 241001, China
| | - Chen Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Mengying Zhang
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes, Wuhu 241001, China; Department of Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu 241001, China
| | - Hui Yang
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes, Wuhu 241001, China; Department of Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu 241001, China
| | - Kun Lv
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes, Wuhu 241001, China; Department of Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu 241001, China.
| |
Collapse
|
10
|
Pan B, Gao J, Chen W, Liu C, Shang L, Xu M, Fu C, Zhu S, Niu M, Xu K. Selective inhibition of interleukin-1 receptor-associated kinase 1 ameliorates lipopolysaccharide-induced sepsis in mice. Int Immunopharmacol 2020; 85:106597. [PMID: 32422509 DOI: 10.1016/j.intimp.2020.106597] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/29/2020] [Accepted: 05/10/2020] [Indexed: 12/26/2022]
Abstract
Interleukin-1 receptor-associated kinases (IRAKs), particularly IRAK1 and IRAK4, are important in transducing signal from Toll-like receptor 4. We interrogated if a selective inhibition of IRAK1 could alleviate lipopolysaccharide (LPS)-induced sepsis. In this study, we tested the impact of a novel selective IRAK1 inhibitor Jh-X-119-01 on LPS-induced sepsis in mice. Survival at day 5 was 13.3% in control group where septic mice were treated by vehicle, while the values were 37.5% (p = 0.046, vs. control) and 56.3% (p = 0.003, vs. control) for 5 mg/kg and 10 mg/kg Jh-X-119-01-treated mice. Jh-X-119-01 alleviated lung injury and reduced production of TNFα and IFNγ in peritoneal macrophages. Jh-X-119-01 decreased phosphorylation of NF-κB and mRNA levels of IL-6 and TNFα in LPS-treated macrophages in vitro. Jh-X-119-01 selectively inhibited IRAK1 phosphorylation comparing with a non-selective IRAK1/4 inhibitor which simultaneously inhibited phosphorylation of IRAK1 and IRAK4. Both Jh-X-119-01 and IRAK1/4 inhibitor increased survival of septic mice, but Jh-X-119-01-treated mice had higher blood CD11b+ cell counts than IRAK1/4 inhibitor-treated ones [24 h: (1.18 ± 0.26) × 106/ml vs. (0.79 ± 0.20) × 106/ml, p = 0.001; 48 h: (1.00 ± 0.30) × 106/ml vs. (0.67 ± 0.23) × 106/ml, p = 0.042]. IRAK1/4 inhibitor induced more apoptosis of macrophages than Jh-X-119-01 did in vitro. IRAK1/4 inhibitor decreased protein levels of anti-apoptotic BCL-2 and MCL-1 in RAW 264.7 and THP-1 cells, an effect not seen in Jh-X-119-01-treated cells. In conclusion, Jh-X-119-01 selectively inhibited activation of IRAK1 and protected mice from LPS-induced sepsis. Jh-X-119-01 showed less toxicity on macrophages comparing with a non-selective IRAK1/4 inhibitor.
Collapse
Affiliation(s)
- Bin Pan
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Jun Gao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Wei Chen
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Cong Liu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Longmei Shang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Mengdi Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Chunling Fu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Shengyun Zhu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Mingshan Niu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
11
|
Barbagelata E, Cillóniz C, Dominedò C, Torres A, Nicolini A, Solidoro P. Gender differences in community-acquired pneumonia. Minerva Med 2020; 111:153-165. [PMID: 32166931 DOI: 10.23736/s0026-4806.20.06448-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Community-acquired pneumonia (CAP) is the most common type of lower respiratory tract infection and a major cause of morbidity and mortality in adults worldwide. Sex and gender play an active role in the incidence and outcomes of major infectious diseases, including CAP. EVIDENCE ACQUISITION We searched the following electronic databases from January 2001 to December 2018: MEDLINE, EMBASE, CINHAIL, CENTRAL (Cochrane Central register of Controlled Trials), DARE (Database of Abstracts of Reviews of Effectiveness), Cochrane Database of Systematic Reviews and ACP Journal Club database. EVIDENCE SYNTHESIS Several studies have reported higher male susceptibility to pulmonary infections and higher risk of death due to sepsis. Biological differences (e.g. hormonal cycles and cellular immune-mediated responses) together with cultural, behavioral and socio-economic differences are important determinants of the course and outcome of CAP. However, gender-related bias in the provision of care and use of hospital resources has been reported among women, resulting in delayed hospital admission and consequently necessary care. CONCLUSIONS CAP is more severe in males than in females, leading to higher mortality in males, especially in older age. To identify gender differences in CAP can guide patient's prognostication and management.
Collapse
Affiliation(s)
- Elena Barbagelata
- Department of Internal Medicine, General Hospital, Sestri Levante, Genoa, Italy
| | - Catia Cillóniz
- Department of Pneumology, Hospital Clinic of Barcelona, August Pi i Sunyer Biomedical Research Institute - IDIBAP S, Biomedical Research Networking Centers in Respiratory Diseases (Ciberes), University of Barcelona, Barcelona, Spain
| | - Cristina Dominedò
- Department of Anesthesiology and Intensive Care Medicine, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
| | - Antoni Torres
- Department of Pneumology, Hospital Clinic of Barcelona, August Pi i Sunyer Biomedical Research Institute - IDIBAP S, Biomedical Research Networking Centers in Respiratory Diseases (Ciberes), University of Barcelona, Barcelona, Spain
| | - Antonello Nicolini
- Unit of Respiratory Diseases, General Hospital, Sestri Levante, Genoa, Italy -
| | - Paolo Solidoro
- Unit of Pneumology U, Cardiovascular and Thoracic Department, Molinette Hospital, Città della Salute e della Scienza, University of Turin, Turin, Italy
| |
Collapse
|
12
|
Morcillo P, Qin Y, Peña G, Mosenthal AC, Livingston DH, Spolarics Z. Directional X Chromosome Skewing of White Blood Cells from Subjects with Heterozygous Mosaicism for the Variant IRAK1 Haplotype. Inflammation 2019; 43:370-381. [PMID: 31748848 DOI: 10.1007/s10753-019-01127-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Random X chromosome (ChrX) inactivation and consequent cellular mosaicism for the active ChrXs in white blood cells (WBCs) is unique to females and may contribute to sex-biased modulation of the innate immune response. Polymorphic differences between the two parental ChrXs may result in ChrX skewing of circulating WBCs (ChrX inactivation-ratio (XCI) > 3) driven by differences in stem cell selection and activity in the bone marrow or WBC trafficking at the periphery. Independent of the mechanism, ChrX skewing may result in genotype-phenotype discrepancies. This study aimed to develop an allele-specific assay and test its applicability in clinical samples to determine the "direction" of ChrX skewing in the variant IRAK1 haplotype, a common X-linked polymorphism with major clinical impacts. Because alternative splice variants of IRAK1 are also produced in the region surrounding the critical single-nucleotide polymorphism (SNP, rs1059703), we also tested the abundance of alternative splice variant IRAK1 transcripts. The expression of splice variants IRAK1-B and IRAK1-C was about 30 and 50% of the full-length (IRAK1-A) in WBCs from healthy subjects (n = 53). IRAK1-A, B, and C showed about 30% lower expression level in males (n = 25) than females (n = 28). By contrast, the expression levels of IRAK1-A, B, and C were not affected by the variant IRAK1 haplotype in either sex. Allele-specific primers generated WT and variant-IRAK1 amplicons with high selectivity, and on average produced about half the expression levels of each transcript in heterozygous IRAK1-mosaic females. Because injury was shown to induce de novo ChrX skewing of WBCs, we tested the directional XCI ratio changes in WBC in a sample of trauma patients heterozygous for the variant IRAK1 haplotype (n = 18). Using the allele-specific assay in combination with the DNA methylation status at the polymorphic HUMARA locus, we found that at admission, about 60% the patients presented XCI ratios skewed toward WBCs with active ChrXs carrying the variant-IRAK1 similar to healthy controls. De novo, trauma-induced XCI ratio changes showed increased extravasation of the more abundant mosaic WBC subset without reversal in the direction of ChrX skewing during the injury course.
Collapse
Affiliation(s)
- Patrick Morcillo
- Department of Surgery, Rutgers-New Jersey Medical School, 185 South Orange Ave., MSB G-578, Newark, NJ, 07103, USA
| | - Yong Qin
- Department of Surgery, Rutgers-New Jersey Medical School, 185 South Orange Ave., MSB G-578, Newark, NJ, 07103, USA
| | - Geber Peña
- Department of Surgery, Rutgers-New Jersey Medical School, 185 South Orange Ave., MSB G-578, Newark, NJ, 07103, USA
| | - Anne C Mosenthal
- Department of Surgery, Rutgers-New Jersey Medical School, 185 South Orange Ave., MSB G-578, Newark, NJ, 07103, USA
| | - David H Livingston
- Department of Surgery, Rutgers-New Jersey Medical School, 185 South Orange Ave., MSB G-578, Newark, NJ, 07103, USA
| | - Zoltan Spolarics
- Department of Surgery, Rutgers-New Jersey Medical School, 185 South Orange Ave., MSB G-578, Newark, NJ, 07103, USA.
| |
Collapse
|
13
|
X-Linked IRAK1 Polymorphism is Associated with Sex-Related Differences in Polymorphonuclear Granulocyte and Monocyte Activation and Response Variabilities. Shock 2019; 53:434-441. [PMID: 31306349 DOI: 10.1097/shk.0000000000001404] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Common X-linked genetic polymorphisms are expected to alter cellular responses affecting males and females differently through sex-linked inheritance pattern as well as X chromosome (ChrX) mosaicism and associated ChrX skewing, which is unique to females. We tested this hypothesis in ex vivo lipopolysaccharide and phorbol ester-stimulated polymorphonuclear granulocytes (PMNs) and monocytes from healthy volunteers (n = 51). Observations were analyzed after stratification by sex alone or the presence of variant IRAK1 haplotype a common X-linked polymorphism with previously demonstrated major clinical impacts. Upon cell activation, CD11b, CD45, CD66b, CD63, and CD14 expression was markedly and similarly elevated in healthy males and females. By contrast, PMN and monocyte activation measured by CD11b, CD66b, and CD63 was increased in variant-IRAK1 subjects as compared with WT. Stratification by IRAK1 genotype and sex showed similar cell activation effect on variant-IRAK1 subjects and an intermediate degree of cell activation in heterozygous mosaic females. The increased membrane expression of these proteins in variant-IRAK1 subjects was associated with similar or increased intersubject but uniformly decreased intrasubject cell response variabilities as compared with WT. We also tested white blood cell ChrX skewing in the healthy cohort as well as in a sample of female trauma patients (n = 201). ChrX inactivation ratios were similar in IRAK1 WT, variant, and heterozygous healthy subjects. Trauma patients showed a trend of blunted ChrX skewing at admission in homozygous variant-IRAK1 and heterozygous mosaic-IRAK1 female subjects as compared with WT. Trauma-induced de novo ChrX skewing was also depressed in variant-IRAK1 and mosaic-IRAK1 female trauma patients as compared with WT. Our study indicates that augmented PMN and monocyte activation in variant-IRAK1 subjects is accompanied by decreased intrasubject cellular variability and blunted de novo ChrX skewing in response to trauma. A more pronounced cell activation of PMNs and monocytes accompanied by decreased response variabilities in variant-IRAK1 subjects may be a contributing mechanism affecting the course of sepsis and trauma and may also impact sex-based outcome differences due to its X-linked inheritance pattern and high prevalence.
Collapse
|
14
|
Vidal C, Moulin F, Nassif X, Galmiche L, Borgel D, Charbit A, Picard C, Mira JP, Lortholary O, Jamet A, Toubiana J. Fulminant arterial vasculitis as an unusual complication of disseminated staphylococcal disease due to the emerging CC1 methicillin-susceptible Staphylococcus aureus clone: a case report. BMC Infect Dis 2019; 19:302. [PMID: 30943907 PMCID: PMC6446405 DOI: 10.1186/s12879-019-3933-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 03/24/2019] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Staphylococcus aureus has emerged as a leading cause of invasive severe diseases with a high rate of morbidity and mortality worldwide. The wide spectrum of clinical manifestations and outcome observed in staphylococcal illness may be a consequence of both microbial factors and variability of the host immune response. CASE PRESENTATION A 14-years old child developed limb ischemia with gangrene following S. aureus bloodstream infection. Histopathology revealed medium-sized arterial vasculitis. The causing strain belonged to the emerging clone CC1-MSSA and numerous pathogenesis-related genes were identified. Patient's genotyping revealed functional variants associated with severe infections. A combination of virulence and host factors might explain this unique severe form of staphylococcal disease. CONCLUSION A combination of virulence and genetic host factors might explain this unique severe form of staphylococcal disease.
Collapse
Affiliation(s)
- Charles Vidal
- Department of Microbiology, Necker Enfants-malades hospital, APHP, Paris Descartes University, Paris, EU, France
| | - Florence Moulin
- Department of Pediatric Intensive Care Unit, Necker Enfants-Malades Hospital, APHP, Paris Descartes University, Paris, EU, France
| | - Xavier Nassif
- Department of Microbiology, Necker Enfants-malades hospital, APHP, Paris Descartes University, Paris, EU, France
| | - Louise Galmiche
- Pathology Department, Necker Enfants-Malades Hospital, APHP, Paris Descartes University, Paris, EU, France
| | - Delphine Borgel
- Department of Hematology, Necker Enfants-Malades Hospital, APHP, Paris Descartes University, Paris, EU, France
| | - Alain Charbit
- Necker-Enfants-Malades Institute, INSERM U1151; CNRS UMR8253, Paris, France
| | - Capucine Picard
- Center for the Study of Primary Immunodeficiencies, Necker Enfants Malades Hospital, APHP, Paris Descartes University, Paris, EU, France.,IHU Imagine, Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Paris, EU, France
| | - Jean-Paul Mira
- Medical Intensive Care Unit, Cochin Hospital, AP-HP, Paris Descartes University, Paris, EU, France.,Department of Infection, Immunity and Inflammation, Institut Cochin, INSERM U1016, Paris, EU, France
| | - Olivier Lortholary
- Department of Infectious Diseases and Tropical Medicine, Necker Enfants-Malades Hospital, Necker-Pasteur Infectious Diseases Center, Université Paris Descartes, IHU Imagine, Paris, EU, France
| | - Anne Jamet
- Department of Microbiology, Necker Enfants-malades hospital, APHP, Paris Descartes University, Paris, EU, France.,Necker-Enfants-Malades Institute, INSERM U1151; CNRS UMR8253, Paris, France
| | - Julie Toubiana
- Department of Infection, Immunity and Inflammation, Institut Cochin, INSERM U1016, Paris, EU, France. .,Department of General Pediatrics and Pediatric Infectious Diseases, Necker Enfants-Malades Hospital, APHP, Paris Descartes University, 149 rue de Sèvres, 75015, Paris, EU, France.
| |
Collapse
|
15
|
Genetic Polymorphisms in Sepsis and Cardiovascular Disease: Do Similar Risk Genes Suggest Similar Drug Targets? Chest 2019; 155:1260-1271. [PMID: 30660782 DOI: 10.1016/j.chest.2019.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/18/2018] [Accepted: 01/02/2019] [Indexed: 01/10/2023] Open
Abstract
Genetic variants are associated with altered clinical outcome of patients with sepsis and cardiovascular diseases. Common gene signaling pathways may be involved in the pathophysiology of these diseases. A better understanding of genetic commonality among these diseases may enable the discovery of important genes, signaling pathways, and therapeutic targets for these diseases. We investigated the common genetic factors by a systematic search of the literature. Twenty-four genes (ADRB2, CD14, FGB, FV, HMOX1, IL1B, IL1RN, IL6, IL10, IL17A, IRAK1, MASP2, MBL, MIR608, MIF, NOD2, PCSK9, PPARG, PROC, SERPINE1, SOD2, SVEP1, TF, TIRAP, TLR1) were extracted as reported genetic variations associated with altered outcome of both sepsis and cardiovascular diseases. Of these genes, the adverse allele (or combinations) was same in nine (ADRB2, FV, HMOX1, IL6, MBL, MIF, NOD2, PCSK9, SERPINE1), and the effect appears to be in the same direction in both sepsis and cardiovascular disease. Shared gene signaling pathways suggest that these are true biological results and could point to overlapping drug targets in sepsis and cardiovascular disease.
Collapse
|
16
|
Singer JW, Fleischman A, Al-Fayoumi S, Mascarenhas JO, Yu Q, Agarwal A. Inhibition of interleukin-1 receptor-associated kinase 1 (IRAK1) as a therapeutic strategy. Oncotarget 2018; 9:33416-33439. [PMID: 30279971 PMCID: PMC6161786 DOI: 10.18632/oncotarget.26058] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 08/15/2018] [Indexed: 02/06/2023] Open
Abstract
Interleukin-1 receptor-associated kinases (IRAK1, IRAK2, IRAK3 [IRAK-M], and IRAK4) are serine-threonine kinases involved in toll-like receptor and interleukin-1 signaling pathways, through which they regulate innate immunity and inflammation. Evidence exists that IRAKs play key roles in the pathophysiologies of cancers, and metabolic and inflammatory diseases, and that IRAK inhibition has potential therapeutic benefits. Molecules capable of selectively interfering with IRAK function and expression have been reported, paving the way for the clinical evaluation of IRAK inhibition. Herein, we focus on IRAK1, review its structure and physiological roles, and summarize emerging data for IRAK1 inhibitors in preclinical and clinical studies.
Collapse
Affiliation(s)
| | - Angela Fleischman
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | | | - John O. Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qiang Yu
- Genome Institute of Singapore, Singapore, SG, Singapore
| | - Anupriya Agarwal
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
17
|
Chamekh M, Deny M, Romano M, Lefèvre N, Corazza F, Duchateau J, Casimir G. Differential Susceptibility to Infectious Respiratory Diseases between Males and Females Linked to Sex-Specific Innate Immune Inflammatory Response. Front Immunol 2017; 8:1806. [PMID: 29321783 PMCID: PMC5733536 DOI: 10.3389/fimmu.2017.01806] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 11/30/2017] [Indexed: 11/13/2022] Open
Abstract
It is widely acknowledged that males and females exhibit contrasting degrees of susceptibility to infectious and non-infectious inflammatory diseases. This is particularly observed in respiratory diseases where human males are more likely to be affected by infection-induced acute inflammations compared to females. The type and magnitude of the innate immune inflammatory response play a cardinal role in this sex bias. Animal models mimicking human respiratory diseases have been used to address the biological factors that could explain the distinct outcomes. In this review, we focus on our current knowledge about experimental studies investigating sex-specific differences in infection-induced respiratory diseases and we provide an update on the most important innate immune mechanisms that could explain sex bias of the inflammatory response. We also discuss whether conclusions drawn from animal studies could be relevant to human.
Collapse
Affiliation(s)
- Mustapha Chamekh
- Inflammation Unit, Laboratory of Pediatric Research, Faculty of Medicine, Queen Fabiola University Children's Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Maud Deny
- Inflammation Unit, Laboratory of Pediatric Research, Faculty of Medicine, Queen Fabiola University Children's Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Marta Romano
- Service of Immunology, Scientific Institute for Public Health (WIV-ISP), Brussels, Belgium
| | - Nicolas Lefèvre
- Laboratory of Translational Research, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of Pulmonology, Allergology and Cystic Fibrosis, Queen Fabiola University Children's Hospital, Brussels, Belgium
| | - Francis Corazza
- Laboratory of Translational Research, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean Duchateau
- Inflammation Unit, Laboratory of Pediatric Research, Faculty of Medicine, Queen Fabiola University Children's Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Georges Casimir
- Inflammation Unit, Laboratory of Pediatric Research, Faculty of Medicine, Queen Fabiola University Children's Hospital, Université Libre de Bruxelles, Brussels, Belgium.,Department of Pulmonology, Allergology and Cystic Fibrosis, Queen Fabiola University Children's Hospital, Brussels, Belgium
| |
Collapse
|
18
|
Spolarics Z, Peña G, Qin Y, Donnelly RJ, Livingston DH. Inherent X-Linked Genetic Variability and Cellular Mosaicism Unique to Females Contribute to Sex-Related Differences in the Innate Immune Response. Front Immunol 2017; 8:1455. [PMID: 29180997 PMCID: PMC5694032 DOI: 10.3389/fimmu.2017.01455] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 10/18/2017] [Indexed: 01/21/2023] Open
Abstract
Females have a longer lifespan and better general health than males. Considerable number of studies also demonstrated that, after trauma and sepsis, females present better outcomes as compared to males indicating sex-related differences in the innate immune response. The current notion is that differences in the immuno-modulatory effects of sex hormones are the underlying causative mechanism. However, the field remains controversial and the exclusive role of sex hormones has been challenged. Here, we propose that polymorphic X-linked immune competent genes, which are abundant in the population are important players in sex-based immuno-modulation and play a key role in causing sex-related outcome differences following trauma or sepsis. We describe the differences in X chromosome (ChrX) regulation between males and females and its consequences in the context of common X-linked polymorphisms at the individual as well as population level. We also discuss the potential pathophysiological and immune-modulatory aspects of ChrX cellular mosaicism, which is unique to females and how this may contribute to sex-biased immune-modulation. The potential confounding effects of ChrX skewing of cell progenitors at the bone marrow is also presented together with aspects of acute trauma-induced de novo ChrX skewing at the periphery. In support of the hypothesis, novel observations indicating ChrX skewing in a female trauma cohort as well as case studies depicting the temporal relationship between trauma-induced cellular skewing and the clinical course are also described. Finally, we list and discuss a selected set of polymorphic X-linked genes, which are frequent in the population and have key regulatory or metabolic functions in the innate immune response and, therefore, are primary candidates for mediating sex-biased immune responses. We conclude that sex-related differences in a variety of disease processes including the innate inflammatory response to injury and infection may be related to the abundance of X-linked polymorphic immune-competent genes, differences in ChrX regulation, and inheritance patterns between the sexes and the presence of X-linked cellular mosaicism, which is unique to females.
Collapse
Affiliation(s)
- Zoltan Spolarics
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Geber Peña
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Yong Qin
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Robert J Donnelly
- Department of Pathology and Laboratory Medicine, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - David H Livingston
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
19
|
Blumhagen RZ, Hedin BR, Malcolm KC, Burnham EL, Moss M, Abraham E, Huie TJ, Nick JA, Fingerlin TE, Alper S. Alternative pre-mRNA splicing of Toll-like receptor signaling components in peripheral blood mononuclear cells from patients with ARDS. Am J Physiol Lung Cell Mol Physiol 2017; 313:L930-L939. [PMID: 28775099 DOI: 10.1152/ajplung.00247.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/28/2017] [Accepted: 07/30/2017] [Indexed: 12/14/2022] Open
Abstract
A key physiological feature of acute respiratory distress syndrome (ARDS) is inflammation. Toll-like receptor (TLR) signaling is required to combat the infection that underlies many ARDS cases but also contributes to pathological inflammation. Several TLR signaling pathway genes encoding positive effectors of inflammation also produce alternatively spliced mRNAs encoding negative regulators of inflammation. An imbalance between these isoforms could contribute to pathological inflammation and disease severity. To determine whether splicing in TLR pathways is altered in patients with ARDS, we monitored alternative splicing of MyD88 and IRAK1, two genes that function in multiple TLR pathways. The MyD88 and IRAK1 genes produce long proinflammatory mRNAs (MyD88L and IRAK1) and shorter anti-inflammatory mRNAs (MyD88S and IRAK1c). We quantified mRNA encoding inflammatory cytokines and MyD88 and IRAK1 isoforms in peripheral blood mononuclear cells (PBMCs) from 104 patients with ARDS and 30 healthy control subjects. We found that MyD88 pre-mRNA splicing is altered in patients with ARDS in a proinflammatory direction. We also observed altered MyD88 isoform levels in a second critically ill patient cohort, suggesting that these changes may not be unique to ARDS. Early in ARDS, PBMC IRAK1c levels were associated with patient survival. Despite the similarities in MyD88 and IRAK1 alternative splicing observed in previous in vitro studies, there were differences in how MyD88 and IRAK1 alternative splicing was altered in patients with ARDS. We conclude that pre-mRNA splicing of TLR signaling genes is altered in patients with ARDS, and further investigation of altered splicing may lead to novel prognostic and therapeutic approaches.
Collapse
Affiliation(s)
- Rachel Z Blumhagen
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado.,Department of Biomedical Research, National Jewish Health, Denver, Colorado
| | - Brenna R Hedin
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado.,Department of Biomedical Research, National Jewish Health, Denver, Colorado
| | - Kenneth C Malcolm
- Department of Medicine, National Jewish Health, Denver, Colorado.,Division of Pulmonary Science and Critical Care Medicine, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado
| | - Ellen L Burnham
- Division of Pulmonary Science and Critical Care Medicine, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado
| | - Marc Moss
- Division of Pulmonary Science and Critical Care Medicine, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado
| | - Edward Abraham
- Office of the Dean, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tristan J Huie
- Department of Medicine, National Jewish Health, Denver, Colorado.,Division of Pulmonary Science and Critical Care Medicine, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado
| | - Jerry A Nick
- Department of Medicine, National Jewish Health, Denver, Colorado.,Division of Pulmonary Science and Critical Care Medicine, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado
| | - Tasha E Fingerlin
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado.,Department of Biomedical Research, National Jewish Health, Denver, Colorado.,Department of Biostatistics and Bioinformatics, Colorado School of Public Health, Aurora, Colorado
| | - Scott Alper
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado; .,Department of Biomedical Research, National Jewish Health, Denver, Colorado.,Program in Mucosal Inflammation and Immunity, National Jewish Health, Denver, Colorado; and.,Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
20
|
Syk Plays a Critical Role in the Expression and Activation of IRAK1 in LPS-Treated Macrophages. Mediators Inflamm 2017; 2017:1506248. [PMID: 28680194 PMCID: PMC5478860 DOI: 10.1155/2017/1506248] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 04/13/2017] [Indexed: 01/10/2023] Open
Abstract
To address how interleukin-1 receptor-associated kinase 1 (IRAK1) is controlled by other enzymes activated by toll-like receptor (TLR) 4, we investigated the possibility that spleen tyrosine kinase (Syk), a protein tyrosine kinase that is activated at an earlier stage during TLR4 activation, plays a central role in regulating the functional activation of IRAK1. Indeed, we found that overexpression of myeloid differentiation primary response gene 88 (MyD88), an adaptor molecule that drives TLR signaling, induced IRAK1 expression and that piceatannol, a Syk inhibitor, successfully suppressed the MyD88-dependent upregulation of IRAK1 under LPS treatment conditions. Interestingly, in Syk-knockout RAW264.7 cells, IRAK1 activity was almost completely blocked after LPS treatment, while providing a Syk-recovery gene to the knockout cells successfully restored IRAK1 expression. According to our measurements of IRAK1 mRNA levels, the transcriptional upregulation of IRAK1 was induced by LPS treatment between 4 and 60 min, and this can be suppressed in Syk knockout cells, providing an effect similar that that seen under piceatannol treatment. The overexpression of Syk reverses this effect and leads to a significantly higher IRAK1 mRNA level. Collectively, our results strongly suggest that Syk plays a critical role in regulating both the activity and transcriptional level of IRAK1.
Collapse
|
21
|
O'Driscoll DN, De Santi C, McKiernan PJ, McEneaney V, Molloy EJ, Greene CM. Expression of X-linked Toll-like receptor 4 signaling genes in female vs. male neonates. Pediatr Res 2017; 81:831-837. [PMID: 28060792 DOI: 10.1038/pr.2017.2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 12/03/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND Male neonates display poorer disease prognosis and outcomes compared with females. Immune genes which exhibit higher expression in umbilical cord blood (UCB) of females may contribute to the female immune advantage during infection and inflammation. The aim of this study was to quantify expression of Toll-like receptor (TLR) 4 signaling genes encoded on the X-chromosome in UCB from term female vs. male neonates. METHODS UCB samples were collected from term neonates (n = 26) born by elective Caesarean section and whole blood was collected from adults (n = 20). Leukocyte RNA was isolated and used in quantitative PCR reactions for IκB kinase γ (IKKγ), Bruton's tyrosine kinase (BTK), and IL-1 receptor associated kinase (IRAK)1. IRAK1 protein was analyzed by Western blot and confocal microscopy. RESULTS In neonates there was no significant difference in the relative expression of IKKγ or BTK mRNA between genders. IRAK1 gene and protein expression was significantly higher in female vs. male UCB, with increased cytosolic IRAK1 expression also evident in female UCB mononuclear cells. Adults had higher expression of all three genes compared with neonates. CONCLUSION Increased expression of IRAK1 could be responsible, in part, for sex-specific responses to infection and subsequent immune advantage in female neonates.
Collapse
Affiliation(s)
- David N O'Driscoll
- Neonatology, National Maternity Hospital, Dublin, Ireland.,Respiratory Research, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland.,Paediatrics, Academic Centre, Tallaght Hospital, Trinity College, The University of Dublin, Dublin, Ireland
| | - Chiara De Santi
- Clinical Microbiology, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Paul J McKiernan
- Respiratory Research, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Victoria McEneaney
- Paediatrics, Academic Centre, Tallaght Hospital, Trinity College, The University of Dublin, Dublin, Ireland
| | - Eleanor J Molloy
- Neonatology, National Maternity Hospital, Dublin, Ireland.,Respiratory Research, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland.,Paediatrics, Academic Centre, Tallaght Hospital, Trinity College, The University of Dublin, Dublin, Ireland.,Neonatology, Coombe Women and Infants' University Hospital, Dublin, Ireland.,Neonatology, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Catherine M Greene
- Clinical Microbiology, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
22
|
Fc Gamma Receptor IIA (CD32A) R131 Polymorphism as a Marker of Genetic Susceptibility to Sepsis. Inflammation 2017; 39:518-25. [PMID: 26490967 DOI: 10.1007/s10753-015-0275-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Sepsis is a devastating disease that can affect humans at any time between neonates and the elderly and is associated with mortality rates that range from 30 to 80%. Despite intensive efforts, its treatment has remained the same over the last few decades. Fc receptors regulate multiple immune responses and have been investigated in diverse complex diseases. FcγRIIA (CD32A) is an immunoreceptor, tyrosine-based activation motif-bearing receptor that binds immunoglobulin G and C-reactive protein, important opsonins in host defense. We conducted a study of 702 patients (184 healthy individuals, 171 non-infected critically ill patients, and 347 sepsis patients) to investigate if genetic polymorphisms in the CD32A coding region affect the risk of septic shock. All individuals were genotyped for a variant at position 131 of the FcγRIIA gene. We found that allele G, associated with the R131 genotype, was significantly more frequent in septic patients than in the other groups (p = 0.05). Our data indicate that FcγRIIA genotyping can be used as a marker of genetic susceptibility to sepsis.
Collapse
|
23
|
Cellular and viral microRNAs in sepsis: mechanisms of action and clinical applications. Cell Death Differ 2016; 23:1906-1918. [PMID: 27740627 DOI: 10.1038/cdd.2016.94] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 07/30/2016] [Accepted: 08/02/2016] [Indexed: 12/17/2022] Open
Abstract
Regardless of its etiology, once septic shock is established, survival rates drop by 7.6% for every hour antibiotic therapy is delayed. The early identification of the cause of infection and prognostic stratification of patients with sepsis are therefore important clinical priorities. Biomarkers are potentially valuable clinical tools in this context, but to date, no single biomarker has been shown to perform adequately. Hence, in an effort to discover novel diagnostic and prognostic markers in sepsis, new genomic approaches have been employed. As a result, a number of small regulatory molecules called microRNAs (miRNAs) have been identified as key regulators of the inflammatory response. Although deregulated miRNA expression is increasingly well described, the pathophysiological roles of these molecules in sepsis have yet to be fully defined. Moreover, non-human miRNAs, including two Kaposi Sarcoma herpesvirus-encoded miRNAs, are implicated in sepsis and may drive enhanced secretion of pro-inflammatory and anti-inflammatory cytokines exacerbating sepsis. A better understanding of the mechanism of action of both cellular and viral miRNAs, and their interactions with immune and inflammatory cascades, may therefore identify novel therapeutic targets in sepsis and make biomarker-guided therapy a realistic prospect.
Collapse
|
24
|
Toubiana J, Courtine E, Tores F, Asfar P, Daubin C, Rousseau C, Ouaaz F, Marin N, Cariou A, Chiche JD, Mira JP. Association of REL polymorphisms and outcome of patients with septic shock. Ann Intensive Care 2016; 6:28. [PMID: 27059500 PMCID: PMC4826362 DOI: 10.1186/s13613-016-0130-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 03/21/2016] [Indexed: 01/15/2023] Open
Abstract
Background cRel, a subunit of NF-κB, is implicated in the inflammatory response observed in autoimmune disease. Hence, knocked-out mice for cRel had a significantly higher mortality, providing new and important functions of cRel in the physiopathology of septic shock. Whether genetic variants in the human REL gene are associated with severity of septic shock is unknown. Methods We genotyped a population of 1040 ICU patients with septic shock and 855 ICU controls for two known polymorphisms of REL; REL rs842647 and REL rs13031237. Outcome of patients according to the presence of REL variant alleles was compared. Results The distribution of REL variant alleles was not significantly different between patients and controls. Among the septic shock group, REL rs13031237*T minor allele was not associated with worse outcome. In contrast, REL rs842647*G minor allele was significantly associated with more multi-organ failure and early death [OR 1.4; 95 % CI (1.02–1.8)]. Conclusion In a large ICU population, we report a significant clinical association between a variation in the human REL gene and severity and mortality of septic shock, suggesting for the first time a new insight into the role of cRel in response to infection in humans.
Collapse
Affiliation(s)
- Julie Toubiana
- Medical School, Paris Descartes University, Paris, France. .,INSERM U1016, CNRS UMR 8104, Cochin Institute, Paris, France. .,Department of Pediatric and Infectious Diseases, Necker University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.
| | - Emilie Courtine
- Medical School, Paris Descartes University, Paris, France.,INSERM U1016, CNRS UMR 8104, Cochin Institute, Paris, France
| | - Frederic Tores
- Bioinformatics Platform, Institut Imagine, Paris Descartes University- Sorbonne Paris Cité, 75015, Paris, France
| | - Pierre Asfar
- Medical Intensive Care Unit, Angers University Hospital, Angers, France
| | - Cédric Daubin
- Medical Intensive Care, Caen University Hospital, Caen, France
| | | | - Fatah Ouaaz
- INSERM U1016, CNRS UMR 8104, Cochin Institute, Paris, France
| | - Nathalie Marin
- Intensive Care Unit, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Alain Cariou
- Medical School, Paris Descartes University, Paris, France.,Intensive Care Unit, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Daniel Chiche
- Medical School, Paris Descartes University, Paris, France.,INSERM U1016, CNRS UMR 8104, Cochin Institute, Paris, France.,Intensive Care Unit, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Paul Mira
- Medical School, Paris Descartes University, Paris, France.,INSERM U1016, CNRS UMR 8104, Cochin Institute, Paris, France.,Intensive Care Unit, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
25
|
Mebazaa A, Laterre PF, Russell JA, Bergmann A, Gattinoni L, Gayat E, Harhay MO, Hartmann O, Hein F, Kjolbye AL, Legrand M, Lewis RJ, Marshall JC, Marx G, Radermacher P, Schroedter M, Scigalla P, Stough WG, Struck J, Van den Berghe G, Yilmaz MB, Angus DC. Designing phase 3 sepsis trials: application of learned experiences from critical care trials in acute heart failure. J Intensive Care 2016; 4:24. [PMID: 27034779 PMCID: PMC4815117 DOI: 10.1186/s40560-016-0151-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 03/17/2016] [Indexed: 12/18/2022] Open
Abstract
Substantial attention and resources have been directed to improving outcomes of patients with critical illnesses, in particular sepsis, but all recent clinical trials testing various interventions or strategies have failed to detect a robust benefit on mortality. Acute heart failure is also a critical illness, and although the underlying etiologies differ, acute heart failure and sepsis are critical care illnesses that have a high mortality in which clinical trials have been difficult to conduct and have not yielded effective treatments. Both conditions represent a syndrome that is often difficult to define with a wide variation in patient characteristics, presentation, and standard management across institutions. Referring to past experiences and lessons learned in acute heart failure may be informative and help frame research in the area of sepsis. Academic heart failure investigators and industry have worked closely with regulators for many years to transition acute heart failure trials away from relying on dyspnea assessments and all-cause mortality as the primary measures of efficacy, and recent trials have been designed to assess novel clinical composite endpoints assessing organ dysfunction and mortality while still assessing all-cause mortality as a separate measure of safety. Applying the lessons learned in acute heart failure trials to severe sepsis and septic shock trials might be useful to advance the field. Novel endpoints beyond all-cause mortality should be considered for future sepsis trials.
Collapse
Affiliation(s)
- Alexandre Mebazaa
- University Paris Diderot, Sorbonne Paris Cité, Paris, France ; U942 Inserm, APHP, Paris, France ; APHP, Department of Anesthesia and Critical Care, Hôpitaux Universitaires Saint Louis-Lariboisière, Paris, France
| | - Pierre François Laterre
- Department of Critical Care Medicine, St. Luc University Hospital, Université Catholique de Louvain (UCL), Brussels, Belgium
| | - James A Russell
- Center for Heart Lung Innovation and the Division of Critical Care Medicine, St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| | | | - Luciano Gattinoni
- Università di Milano, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Etienne Gayat
- Département d'Anesthésie - Réanimation - SMUR, Hôpitaux Universitaires Saint Louis - Lariboisière, INSERM - UMR 942, Assistance Publique - Hôpitaux de Paris, Université Paris Diderot, Paris, France
| | - Michael O Harhay
- Division of Epidemiology, Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | | | | | | | - Matthieu Legrand
- Department of Anesthesiology, Critical Care and Burn Unit, St. Louis Hospital, University Paris 7 Denis Diderot, UMR-S942, Inserm, Paris, France
| | - Roger J Lewis
- Department of Emergency Medicine, Harbor-UCLA Medical Center, Torrance, CA USA
| | - John C Marshall
- Department of Surgery, Interdepartmental Division of Critical Care Medicine, University of Toronto, St. Michael's Hospital, Toronto, Ontario Canada
| | - Gernot Marx
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Peter Radermacher
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum, Ulm, Germany
| | | | | | - Wendy Gattis Stough
- Campbell University College of Pharmacy and Health Sciences, Buies Creek, NC USA
| | | | - Greet Van den Berghe
- Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Mehmet Birhan Yilmaz
- Department of Cardiology, Cumhuriyet University Faculty of Medicine, Sivas, Turkey
| | - Derek C Angus
- CRISMA Center, Department of Critical Care Medicine, McGowan Institute for Regnerative Medicine, Clinical and Translational Science Institute, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, PA USA ; Department of Health Policy and Management, McGowan Institute for Regnerative Medicine, Clinical and Translational Science Institute, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, PA USA
| |
Collapse
|
26
|
Thair SA, Topchiy E, Boyd JH, Cirstea M, Wang C, Nakada TA, Fjell CD, Wurfel M, Russell JA, Walley KR. TNFAIP2 Inhibits Early TNFα-Induced NF-x03BA;B Signaling and Decreases Survival in Septic Shock Patients. J Innate Immun 2015; 8:57-66. [PMID: 26347487 DOI: 10.1159/000437330] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 07/01/2015] [Indexed: 12/24/2022] Open
Abstract
During septic shock, tumor necrosis factor alpha (TNFα) is an early response gene and induces a plethora of genes and signaling pathways. To identify robust signals in genes reliably upregulated by TNFα, we first measured microarray gene expression in vitro and searched methodologically comparable, publicly available data sets to identify concordant signals. Using tag single-nucleotide polymorphisms in the genes common to all data sets, we identified a genetic variant of the TNFAIP2 gene, rs8126, associated with decreased 28-day survival and increased organ dysfunction in an adult cohort in the Vasopressin and Septic Shock Trial. Similar to this cohort, we found that an association with rs8126 and increased organ dysfunction is replicated in a second cohort of septic shock patients in the St. Paul's Hospital Intensive Care Unit. We found that TNFAIP2 inhibits NF-x03BA;B activity, impacting the downstream cytokine interleukin (IL)-8. The minor G allele of TNFAIP2 rs8126 resulted in greater TNFAIP2 expression, decreased IL-8 production and was associated with decreased survival in patients experiencing septic shock. These data suggest that TNFAIP2 is a novel inhibitor of NF-x03BA;B that acts as an autoinhibitor of the TNFα response during septic shock.
Collapse
Affiliation(s)
- Simone A Thair
- Department of Emergency and Surgery, Stanford University School of Medicine, Stanford, Calif., USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
The intracerebroventricular injection of rimonabant inhibits systemic lipopolysaccharide-induced lung inflammation. J Neuroimmunol 2015; 286:16-24. [PMID: 26298320 DOI: 10.1016/j.jneuroim.2015.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 06/22/2015] [Accepted: 07/01/2015] [Indexed: 01/23/2023]
Abstract
We investigated the role of intracerebroventricular (ICV) injection of rimonabant (500ng), a CB1 antagonist, on lipopolysaccharide ((LPS) 5mg/kg)-induced pulmonary inflammation in rats in an isolated perfused lung model. There were decreases in pulmonary capillary pressure (Ppc) and increases in the ((Wet-Dry)/Dry lung weight)/(Ppc) ratio in the ICV-vehicle/LPS group at 4h. There were decreases in TLR4 pathway markers, such as interleukin receptor-associated kinase-1, IκBα, Raf1 and phospho-SFK (Tyr416) at 30min and at 4h increases in IL-6, vascular cell adhesion molecule-1 and myeloperoxidase in lung homogenate. Intracerebroventricular rimonabant attenuated these LPS-induced responses, indicating that ICV rimonabant modulates LPS-initiated pulmonary inflammation.
Collapse
|
28
|
Pène F, Ait-Oufella H, Taccone FS, Monneret G, Sharshar T, Tamion F, Mira JP. Insights and limits of translational research in critical care medicine. Ann Intensive Care 2015; 5:8. [PMID: 25977834 PMCID: PMC4420765 DOI: 10.1186/s13613-015-0050-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 04/20/2015] [Indexed: 12/23/2022] Open
Abstract
Experimental research has always been the cornerstone of pathophysiological and therapeutic advances in critical care medicine, where clinical observations and basic research mutually fed each other in a so-called translational approach. The objective of this review is to address the different aspects of translational research in the field of critical care medicine. We herein highlighted some demonstrative examples including the animal-to-human approach to study host-pathogen interactions, the human-to-animal approach for sepsis-induced immunosuppression, the still restrictive human approach to study critical illness-related neuromyopathy, and the technological developments to assess the microcirculatory changes in critically ill patients. These examples not only emphasize how translational research resulted in major improvements in the comprehension of the pathophysiology of severe clinical conditions and offered promising perspectives in critical care medicine but also point out the obstacles to translate such achievements into clinical practice.
Collapse
Affiliation(s)
- Frédéric Pène
- Service de Réanimation Médicale, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 27 Rue du Faubourg Saint-Jacques, 75014 Paris, France ; Faculté de Médecine, Université Paris Descartes, 12 Rue de l'Ecole de Médecine, 75006 Paris, France
| | - Hafid Ait-Oufella
- Service de Réanimation Médicale, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, 184 Rue du Faubourg Saint-Antoine, 75012 Paris, France ; Faculté de Médecine, Université Pierre-et-Marie-Curie, 27 Rue Chaligny, 75571 Paris, France
| | - Fabio Silvio Taccone
- Département de Soins Intensifs, Hôpital Erasme, Université Libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium
| | - Guillaume Monneret
- Laboratoire d'Immunologie Cellulaire, Hôpital Edouard Herriot, Hospices Civils de Lyon, 5 Place d'Arsonval, 69003 Lyon, France ; Institut des Sciences Pharmaceutiques et Biologiques, Université Lyon I, 8 Avenue Rockefeller, 69373 Lyon, France
| | - Tarek Sharshar
- Service de Réanimation, Hôpital Raymond Poincaré, Assistance Publique-Hôpitaux de Paris, 104 Boulevard Raymond Poincaré, 92380 Garches, France ; Université de Versailles-Saint Quentin en Yvelines, 55 Avenue de Paris, 78000 Versailles, France
| | - Fabienne Tamion
- Service de Réanimation Médicale, Hôpital Charles Nicolle, CHU Rouen, 1 Rue de Germont, 76000 Rouen, France ; Faculté de Médecine, Institut de Recherche et Innovation Biomédicale (IRIB), 22 Boulevard Gambetta, 76000 Rouen, France
| | - Jean-Paul Mira
- Service de Réanimation Médicale, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 27 Rue du Faubourg Saint-Jacques, 75014 Paris, France ; Faculté de Médecine, Université Paris Descartes, 12 Rue de l'Ecole de Médecine, 75006 Paris, France
| | | |
Collapse
|
29
|
Sethuraman KN, Marcolini EG, McCunn M, Hansoti B, Vaca FE, Napolitano LM. Gender-specific issues in traumatic injury and resuscitation: consensus-based recommendations for future research. Acad Emerg Med 2014; 21:1386-94. [PMID: 25420732 DOI: 10.1111/acem.12536] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 07/26/2014] [Accepted: 07/29/2014] [Indexed: 02/03/2023]
Abstract
Traumatic injury remains an unacceptably high contributor to morbidity and mortality rates across the United States. Gender-specific research in trauma and emergency resuscitation has become a rising priority. In concert with the 2014 Academic Emergency Medicine consensus conference "Gender-specific Research in Emergency Care: Investigate, Understand, and Translate How Gender Affects Patient Outcomes," a consensus-building group consisting of experts in emergency medicine, critical care, traumatology, anesthesiology, and public health convened to generate research recommendations and priority questions to be answered and thus move the field forward. Nominal group technique was used for the consensus-building process and a combination of face-to-face meetings, monthly conference calls, e-mail discussions, and preconference surveys were used to refine the research questions. The resulting research agenda focuses on opportunities to improve patient outcomes by expanding research in sex- and gender-specific emergency care in the field of traumatic injury and resuscitation.
Collapse
Affiliation(s)
- Kinjal N. Sethuraman
- Department of Emergency Medicine and the Division of Hyperbaric Medicine; R Adams Cowley Shock Trauma Center; University of Maryland School of Medicine; Baltimore MD
| | | | - Maureen McCunn
- Department of Anesthesiology; Divisions of Trauma Anesthesiology and Surgical Critical Care; Baltimore MD
| | - Bhakti Hansoti
- Department of Emergency Medicine; Johns Hopkins University; Baltimore MD
| | | | | |
Collapse
|
30
|
X chromosome-linked IRAK-1 polymorphism is a strong predictor of multiple organ failure and mortality postinjury. Ann Surg 2014; 260:698-703; discussion 703-5. [PMID: 25203887 DOI: 10.1097/sla.0000000000000918] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE(S) Clinical research characterizing the mechanisms responsible for sex-based outcome differences postinjury remain conflicting. We sought to characterize an X chromosome-linked IRAK-1 (IL-1 receptor-associated kinase) polymorphism as an alternative mechanism responsible for sex differences postinjury. IRAK-1 is key intermediate in the toll-like receptor (TLR) pathway thought to drive inflammation postinjury. METHODS A prospective cohort study was performed over a 24-month period. Bluntly injured patients requiring intensive care unit admission were enrolled, whereas patients with isolated brain and spinal cord injuries were excluded. Outcomes of interest included multiple organ failure (MOF, Marshall MOD score > 5) and mortality. Logistic regression was utilized to determine the independent risk of poor outcome associated with the IRAK-1 variant after controlling for important differences. RESULTS In an enrolled cohort of 321 patients, the IRAK-1 variant was common (12.5%). Patients with and without the variant were similar in age, injury severity, and 24hr blood transfusion. After controlling for important confounders, the IRAK1 variant was independently associated with more than eightfold (OR = 8.4, P = 0.005, 95% CI: 1.9-37.1) and 11-fold (OR = 11.8, P = 0.037, 95% CI: 1.1-121) greater risk of MOF and mortality, respectively. These differences were most prominent in men, whereas women heterozygous for the variant demonstrated worse outcome in a dose-dependent fashion. CONCLUSIONS The IRAK1 polymorphism is a strong independent predictor of MOF and mortality postinjury and represents a common variant with prognostic potential. These data demonstrate the importance of TLR signaling postinjury and supports that a genetic mechanism may drive sex outcome differences postinjury.
Collapse
|
31
|
Yin J, Yao CL, Liu CL, Song ZJ, Tong CY, Huang PZ. Association of genetic variants in the IRAK-4 gene with susceptibility to severe sepsis. World J Emerg Med 2014; 3:123-7. [PMID: 25215050 DOI: 10.5847/wjem.j.issn.1920-8642.2012.02.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Accepted: 04/19/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The association of genetic variation in the IRAK-1 gene with sepsis outcome has been proved. However, few studies have addressed the impact of the IRAK-4 gene variants on sepsis risk. This study aimed to determine whether the polymorphisms in the IRAK-4 gene are associated with susceptibility to and prognosis of severe sepsis in the Chinese Han ethnic population. METHODS In this case-control study, 192 patients with severe sepsis hospitalized in the emergency department of Zhongshan Hospital from February 2006 to December 2009 and 192 healthy volunteers were enrolled. Exclusion criteria included metastatic tumors, autoimmune diseases, AIDS or treatment with immunosuppressive drugs. This study was approved by the ethical committee of Zhongshan Hospital, Fudan University. Sepsis patients were divided into a survival group (n=124) and a non-survival group (n=68) according to the 30-day mortality. Primer 3 software was used to design PCR and sequencing primers. Genomic DNA was extracted from peripheral blood mononuclear cells. Seven tagSNPs in IRAK-4 were selected according to the data of the Chinese Han population in Beijing from the Hapmap project and genotyped by direct sequencing. The chi-square test was used to evaluate the differences in genotype and allele frequencies between the two groups. RESULTS The distributions of all tagSNPs were consistent with Hardy-Weinberg equilibrium. The allele and genotype frequencies of rs4251545 (G/A) were significantly different between the severe sepsis and healthy control groups (P=0.015, P=0.035, respectively). Carriers of the rs4251545A had a higher risk for severe sepsis compared with carriers of the rs4251545G (OR=1.69, 95% CI: 1.10-2.58). The allele and genotype frequencies of all SNPs were not significantly different between the survival group and non-survival group. CONCLUSION These findings indicate that the variants in IRAK-4 are significantly associated with susceptibility to severe sepsis in the Chinese Han ethnic population.
Collapse
Affiliation(s)
- Jun Yin
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032,China
| | - Chen-Ling Yao
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032,China
| | - Cheng-Long Liu
- Department of Emergency Medicine, Ri Zhao Hospital of Traditional Chinese Medicine, Ri Zhao, 276800,China
| | - Zhen-Ju Song
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032,China
| | - Chao-Yang Tong
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032,China
| | - Pei-Zhi Huang
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032,China
| |
Collapse
|
32
|
Lee I, Hüttemann M. Energy crisis: the role of oxidative phosphorylation in acute inflammation and sepsis. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:1579-86. [PMID: 24905734 PMCID: PMC4147665 DOI: 10.1016/j.bbadis.2014.05.031] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/17/2014] [Accepted: 05/27/2014] [Indexed: 12/13/2022]
Abstract
Mitochondrial dysfunction is increasingly recognized as an accomplice in most of the common human diseases including cancer, neurodegeneration, diabetes, ischemia/reperfusion injury as seen in myocardial infarction and stroke, and sepsis. Inflammatory conditions, both acute and chronic, have recently been shown to affect mitochondrial function. We here discuss the role of oxidative phosphorylation (OxPhos), focusing on acute inflammatory conditions, in particular sepsis and experimental sepsis models. We discuss mitochondrial alterations, specifically the suppression of oxidative metabolism and the role of mitochondrial reactive oxygen species in disease pathology. Several signaling pathways including metabolic, proliferative, and cytokine signaling affect mitochondrial function and appear to be important in inflammatory disease conditions. Cytochrome c oxidase (COX) and cytochrome c, the latter of which plays a central role in apoptosis in addition to mitochondrial respiration, serve as examples for the entire OxPhos system since they have been studied in more detail with respect to cell signaling. We propose a model in which inflammatory signaling leads to changes in the phosphorylation state of mitochondrial proteins, including Tyr304 phosphorylation of COX catalytic subunit I. This results in an inhibition of OxPhos, a reduction of the mitochondrial membrane potential, and consequently a lack of energy, which can cause organ failure and death as seen in septic patients.
Collapse
Affiliation(s)
- Icksoo Lee
- College of Medicine, Dankook University, Cheonan-si, Chungcheongnam-do 330-714, Republic of Korea
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA; Cardiovascular Research Institute, Wayne State University, Detroit, MI 48201, USA; Department of Biochemistry and Molecular Biology, Wayne State University, Detroit, MI 48201, USA; Karmanos Cancer Institute, Detroit, MI 48201, USA.
| |
Collapse
|
33
|
Abstract
Interleukin-1 receptor-associated kinase (IRAK1) is a key regulatory protein in TLR/IL1R-mediated cell activation during inflammatory response. Studies indicated that pending on the nature of the used inflammatory model, downregulation of IRAK1 may be beneficial or detrimental. However, the role of IRAK1 in affecting outcome in polymicrobial sepsis is unknown. We tested this question using an IRAK1-deficient mouse strain and cecal ligation and puncture (CLP) procedure, which is a clinically relevant rodent septic model. Sepsis-induced mortality was markedly lower in IRAK1-deficient mice (35 %) compared to WT (85 %). Sepsis-induced increases in blood IL-6 and IL-10 levels were blunted at 6 h post-CLP in IRAK1 deficiency compared to WT, but cytokine levels were similar at 20 h post-CLP. Sepsis-induced blood granulocytosis and depletion of splenic B cells were also blunted in IRAK1-deficient mice as compared to WT. Analysis of TLR-mediated cytokine responses by IRAK1-deficient and WT macrophages ex vivo indicated a TLR4-dependent downregulation of IL-6 and IL1β in IRAK1 deficiency, whereas TLR2-dependent responses were unaffected. TLR7/8-mediated IL-6, IL1β, and IL-10 production was also blunted in IRAK1 macrophages as compared to WT. The study shows that IRAK1 deficiency impacts multiple TLR-dependent pathways and decreases early cytokine responses following polymicrobial sepsis. The delayed inflammatory response caused by the lack of IRAK1 expression is beneficial, as it manifests a marked increased chance of survival after polymicrobial sepsis.
Collapse
|
34
|
|
35
|
Chandra R, Federici S, Németh ZH, Csóka B, Thomas JA, Donnelly R, Spolarics Z. Cellular mosaicism for X-linked polymorphisms and IRAK1 expression presents a distinct phenotype and improves survival following sepsis. J Leukoc Biol 2013; 95:497-507. [PMID: 24193737 DOI: 10.1189/jlb.0713397] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
ChrX cellular mosaicism for X-linked genetic polymorphisms in females versus the single ChrX representation in males denotes a genetic difference, which may contribute to gender bias in the inflammatory response. This hypothesis was tested in female F1 offspring of consomic mice (BL6J-ChrX(A/J)/NaJ) that were homokaryotic or mosaic for the active BL6 and AJ ChrXs or for IRAK1 deficiency linked to the BL6 ChrX. Sepsis was initiated by CLP. IRAK1-deficient and IRAK1-mosaic mice showed similar protection from sepsis-induced mortality and reduced IL-6 and IL-10 release compared with WT. BM cellularity and blood B cell counts were increased in naive IRAK1-mosaic mice compared with WT-mosaic or IRAK1-deficient animals. Sepsis-induced BM cell depletion was greater in IRAK1-mosaic mice compared with WT-mosaic or IRAK1-deficient subjects, whereas splenic B and T cell depletion was less in IRAK1-mosaic and IRAK1-deficient than WT-mosaic mice. Skewing toward AJ or BL6-ChrX-expressing cells was assessed by testing allele-specific expression of strain-variant Xkrx and BTK genes. In naive IRAK1-mosaic mice, BM and blood cells with the active BL6-ChrX, were greater than cells expressing the AJ-ChrX (cell ratio 2.5 in IRAK1-mosaic; 1.5 in WT-mosaic mice). Sepsis decreased cell ratios more in IRAK1-mosaic than in WT-mosaic mice. The study reveals functional variability in cellular mosaicism for IRAK1 expression and natural X-linked polymorphisms during sepsis. Mosaicism for IRAK1 expression is accompanied by skewing toward deficient immune cell populations, producing a phenotype that is preconditioned for improved sepsis outcome similar to that observed in IRAK1 deficiency.
Collapse
Affiliation(s)
- Rachna Chandra
- 1.Rutgers New Jersey Medical School, 185 South Orange Ave., MSB G-578, Newark, NJ 07103, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Cruzat VF, Bittencourt A, Scomazzon SP, Leite JSM, de Bittencourt PIH, Tirapegui J. Oral free and dipeptide forms of glutamine supplementation attenuate oxidative stress and inflammation induced by endotoxemia. Nutrition 2013; 30:602-11. [PMID: 24698353 DOI: 10.1016/j.nut.2013.10.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 09/12/2013] [Accepted: 10/24/2013] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The aim of the present study was to determine the effects of oral supplementation with L-glutamine plus L-alanine (GLN+ALA), both in the free form and L-alanyl-L-glutamine dipeptide (DIP) in endotoxemic mice. METHODS B6.129 F2/J mice were subjected to endotoxemia (Escherichia coli lipopolysaccharide [LPS], 5 mg/kg, LPS group) and orally supplemented for 48 h with either L-glutamine (1 g/kg) plus L-alanine (0.61 g/kg) (GLN+ALA-LPS group) or 1.49 g/kg DIP (DIP-LPS group). Plasma glutamine, cytokines, and lymphocyte proliferation were measured. Liver and skeletal muscle glutamine, glutathione (GSH), oxidized GSH (GSSG), tissue lipoperoxidation (TBARS), and nuclear factor (NF)-κB-interleukin-1 receptor-associated kinase 1 (IRAK1)-Myeloid differentiation primary response gene 88 pathway also were determined. RESULTS Endotoxemia depleted plasma (by 71%), muscle (by 44%), and liver (by 49%) glutamine concentrations (relative to the control group), which were restored in both GLN+ALA-LPS and DIP-LPS groups (P < 0.05). Supplemented groups reestablished GSH content, intracellular redox status (GSSG/GSH ratio), and TBARS concentration in muscle and liver (P < 0.05). T- and B-lymphocyte proliferation increased in supplemented groups compared with controls and LPS group (P < 0.05). Tumor necrosis factor-α, interleukin (IL)-6, IL-1 β, and IL-10 increased in LPS group but were attenuated by the supplements (P < 0.05). Endotoxemic mice exhibited higher muscle gene expression of components of the NF-κB pathway, with the phosphorylation of IκB kinase-α/β. These returned to basal levels (relative to the control group) in both GLN+ALA-LPS and DIP-LPS groups (P < 0.05). Higher mRNA of IRAK1 and MyD88 were observed in muscle of LPS group compared with the control and supplemented groups (P < 0.05). CONCLUSION Oral supplementations with GLN+ALA or DIP are effective in attenuating oxidative stress and the proinflammatory responses induced by endotoxemia in mice.
Collapse
Affiliation(s)
- Vinicius Fernandes Cruzat
- Department of Food Science and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Faculty of Health Sciences, Curtin University, Perth, Western Australia.
| | - Aline Bittencourt
- Department of Physiology, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Rua Sarmento Leite, Porto Alegre, Brazil
| | - Sofia Pizzato Scomazzon
- Department of Physiology, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Rua Sarmento Leite, Porto Alegre, Brazil
| | - Jaqueline Santos Moreira Leite
- Department of Food Science and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Paulo Ivo Homem de Bittencourt
- Department of Physiology, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Rua Sarmento Leite, Porto Alegre, Brazil
| | - Julio Tirapegui
- Department of Food Science and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
37
|
Abstract
At mucosal surfaces, phagocytes such as macrophages coexist with microbial communities; highly controlled regulation of these interactions is essential for immune homeostasis. Pattern-recognition receptors (PRRs) are critical in recognizing and responding to microbial products, and they are subject to negative regulation through various mechanisms, including downregulation of PRR-activating components or induction of inhibitors. Insights into these regulatory mechanisms have been gained through human genetic disease-association studies, in vivo mouse studies utilizing disease models or targeted gene perturbations, and in vitro and ex vivo human cellular studies examining phagocytic cell functions. Although mouse models provide an important approach to study macrophage regulation, human and mouse macrophages exhibit differences, which must be considered when extrapolating mouse findings to human physiology. This review discusses inhibitory regulation of PRR-induced macrophage functions and the consequences of dysregulation of these functions and highlights mechanisms that have a role in intestinal macrophages and in human macrophage studies.
Collapse
Affiliation(s)
- M Hedl
- Department of Internal Medicine, Yale University, New Haven, Connecticut, USA
| | | |
Collapse
|
38
|
Song Z, Song Y, Yin J, Shen Y, Yao C, Sun Z, Jiang J, Zhu D, Zhang Y, Shen Q, Gao L, Tong C, Bai C. Genetic variation in the TNF gene is associated with susceptibility to severe sepsis, but not with mortality. PLoS One 2012; 7:e46113. [PMID: 23029405 PMCID: PMC3459853 DOI: 10.1371/journal.pone.0046113] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 08/28/2012] [Indexed: 01/18/2023] Open
Abstract
Background Tumor necrosis factor (TNF) and TNF receptor superfamily (TNFR)-mediated immune response play an essential role in the pathogenesis of severe sepsis. Studies examining associations of TNF and lymphotoxin-α (LTA) single nucleotide polymorphisms (SNPs) with severe sepsis have produced conflicting results. The objective of this study was to investigate whether genetic variation in TNF, LTA, TNFRSF1A and TNFRSF1B was associated with susceptibility to or death from severe sepsis in Chinese Han population. Methodology/Principal Findings Ten SNPs in TNF, LTA, TNFRSF1A and TNFRSF1B were genotyped in samples of patients with severe sepsis (n = 432), sepsis (n = 384) and healthy controls (n = 624). Our results showed that rs1800629, a SNP in the promoter region of TNF, was significantly associated with risk for severe sepsis. The minor allele frequency of rs1800629 was significantly higher in severe sepsis patients than that in both healthy controls (Padj = 0.00046, odds ratio (OR)adj = 1.92) and sepsis patients (Padj = 0.002, ORadj = 1.56). Further, we investigated the correlation between rs1800629 genotypes and TNF-α concentrations in peripheral blood mononuclear cells (PBMCs) of healthy volunteers exposed to lipopolysaccharides (LPS) ex vivo, and the association between rs1800629 and TNF-α serum levels in severe sepsis patients. After exposure to LPS, the TNF-α concentration in culture supernatants of PBMCs was significantly higher in the subjects with AA+AG genotypes than that with GG genotype (P = 0.007). Moreover, in patients with severe sepsis, individuals with AA+AG genotypes had significantly higher TNF-α serum concentrations than those with GG genotype (Padj = 0.02). However, there were no significant associations between SNPs in the four candidate genes and 30 day mortality for patients with severe sepsis. Conclusions/Significance Our findings suggested that the functional TNF gene SNP rs1800629 was strongly associated with susceptibility to severe sepsis, but not with lethality in Chinese Han population.
Collapse
Affiliation(s)
- Zhenju Song
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yuanlin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jun Yin
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yao Shen
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Chenling Yao
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Zhan Sun
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jinjun Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Duming Zhu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yong Zhang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Qinjun Shen
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Lei Gao
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Chaoyang Tong
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- * E-mail: (CT); (CB)
| | - Chunxue Bai
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- * E-mail: (CT); (CB)
| |
Collapse
|
39
|
Song Z, Yao C, Yin J, Tong C, Zhu D, Sun Z, Jiang J, Shao M, Zhang Y, Deng Z, Tao Z, Sun S, Bai C. Genetic variation in the TNF receptor-associated factor 6 gene is associated with susceptibility to sepsis-induced acute lung injury. J Transl Med 2012; 10:166. [PMID: 22901274 PMCID: PMC3478205 DOI: 10.1186/1479-5876-10-166] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 07/09/2012] [Indexed: 12/31/2022] Open
Abstract
Background Recent studies showed that overwhelming inflammatory response mediated by the toll-like receptor (TLR)-related pathway was important in the development of acute lung injury (ALI). The aim of this study was to determine whether common genetic variation in four genes of the TLR signaling pathway were associated with sepsis-induced ALI susceptibility and risk of death in Chinese Han population. Methods Fourteen tag single nucleotide polymorphisms (tagSNPs) in MyD88, IRAK1, IRAK4 and TRAF6 were genotyped in samples of sepsis-induced ALI (n = 272) and sepsis alone patients (n = 276), and tested for association in this case-control collection. Then, we investigated correlation between the associated SNP and the mRNA expression level of the corresponding gene. And we also investigated correlation between the associated SNP and tumor necrosis factor alpha (TNF-α) as well as interleukin-6 (IL-6) concentrations in peripheral blood mononuclear cells (PBMCs) exposed to lipopolysaccharides (LPS) ex vivo. The mRNA expression level was determined using real-time quantitative Polymerase Chain Reaction (PCR) assays, and concentrations of TNF-α and IL-6 were measured by enzyme-linked immunosorbent assay (ELISA). Results The association analysis revealed that rs4755453, an intronic SNP of TRAF6, was significantly associated with susceptibility to sepsis-induced ALI. The C allele frequency of rs4755453 in the sepsis alone group was significantly higher than that in the sepsis-induced ALI group (P = 0.00026, odds ratio (OR) = 0.52, 95% confidence interval (CI) 0.37–0.74). These associations remained significant after adjustment for covariates in multiple logistic regression analysis and for multiple comparisons. TRAF6 mRNA expression levels in PBMCs from homozygotes of the rs4755453G allele were significantly higher than that in heterozygotes and homozygotes of the rs4755453C allele at baseline (P = 0.012 and P = 0.003, respectively) as well as after LPS stimulation (P = 0.009 and P = 0.005). Moreover, the concentrations of TNF-α and IL-6 in cell culture supernatants were also significantly higher in the subjects with rs4755453GG genotype than in subjects with CG and CC genotype. None of the 14 tagSNPs showed associations with risk of death and severity among ALI cases. Conclusions Our findings indicated that common genetic variants in TRAF6 were significantly associated with susceptibility to sepsis-induced ALI in Chinese Han population. This was the first genetic evidence supporting a role for TRAF6 in ALI.
Collapse
Affiliation(s)
- Zhenju Song
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Sampath V, Garland JS, Le M, Patel AL, Konduri GG, Cohen JD, Simpson PM, Hines RN. A TLR5 (g.1174C > T) variant that encodes a stop codon (R392X) is associated with bronchopulmonary dysplasia. Pediatr Pulmonol 2012; 47:460-8. [PMID: 22058078 DOI: 10.1002/ppul.21568] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 08/19/2011] [Indexed: 11/11/2022]
Abstract
Current evidence supports a major role for inherited factors in determining bronchopulmonary dysplasia (BPD) susceptibility. The Toll-like receptor (TLR) family of proteins maintain pulmonary homeostasis in the developing lung by aiding pathogen recognition and clearance, regulating inflammation, and facilitating reparative tissue growth. We hypothesized that sequence variation in the TLR pathway genes would alter the susceptibility/severity of BPD in preterm infants. Very low birth-weight infants were recruited prospectively in a multi-center study involving collection of blood samples and clinical information. Nine TLR pathway single-nucleotide polymorphisms were genotyped using a multiplexed single-base extension assay. BPD outcomes were compared among infants with and without the variant allele using Chi-square or Fisher's exact tests. In our cohort (n = 289), 66 (23.6%) infants developed BPD, out of which 32 (11.2%) developed severe BPD. The TLR5 (g.1174C > T) variant was associated with BPD (P = 0.03) and severe BPD (P = 0.004). The TIRAP (g.2054C > T) variant was associated with BPD (P = 0.04). Infants heterozygous for the X-linked IRAK1 (g.6435T > C) variant had a lower incidence of BPD compared to infants homozygous for either the reference or variant allele (P = 0.03). In regression models that controlled for potential epidemiological confounders, the TIRAP variant was associated with BPD, and the TLR5 variant was associated with severe BPD. Our data support the hypothesis that aberrant pathogen recognition in premature infants arising from TLR pathway genetic variation can contribute to BPD pathogenesis.
Collapse
Affiliation(s)
- Venkatesh Sampath
- Department of Pediatrics, Medical College of Wisconsin, and Children's Research Institute, Children's Hospital and Health Systems, Milwaukee, Wisconsin, USA.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Sperry JL, Vodovotz Y, Ferrell RE, Namas R, Chai YM, Feng QM, Jia WP, Forsythe RM, Peitzman AB, Billiar TR. Racial disparities and sex-based outcomes differences after severe injury. J Am Coll Surg 2012; 214:973-80. [PMID: 22521668 DOI: 10.1016/j.jamcollsurg.2012.02.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 02/23/2012] [Accepted: 02/23/2012] [Indexed: 12/28/2022]
Abstract
BACKGROUND Controversy exists about the mechanisms responsible for sex-based outcomes differences post-injury. X-chromosome-linked immune response pathway polymorphisms represent a potential mechanism resulting in sex-based outcomes differences post-injury. The prevalence of these variants is known to differ across race. We sought to characterize racial differences and the strength of any sex-based dimorphism post-injury. STUDY DESIGN A retrospective analysis was performed using data derived from the National Trauma Data Bank 7.1 (2002-2006). Blunt-injured adult (older than 15 years) patients, surviving >24 hours and with an Injury Severity Score >16 were analyzed (n = 244,371). Patients were stratified by race (Caucasian, black, Hispanic, Asian) and multivariable regression analysis was used to characterize the risk of mortality and the strength of protection associated with sex (female vs male). RESULTS When stratified by race, multivariable models demonstrated Caucasian females had an 8.5% lower adjusted risk of mortality (odds ratio [OR] = 0.91; 95% CI, 0.88-0.95; p < 0.001) relative to Caucasian males, with no significant association found for Hispanics or blacks. An exaggerated survival benefit was afforded to Asian females relative to Asian males, having a >40% lower adjusted risk of mortality (OR = 0.59; 95% CI, 0.44-78; p < 0.001). Asian males had a >75% higher adjusted risk of mortality relative to non-Asian males (OR = 1.77; 95% CI, 1.5-2.0; p < 0.001), and no significant difference in the mortality risk was found for Asian females relative to non-Asian females. CONCLUSIONS These results suggest that Asian race is associated with sex-based outcomes differences that are exaggerated, resulting from worse outcomes for Asian males. These racial disparities suggest a negative male X-chromosome-linked effect as the mechanism responsible for these sex-based outcomes differences.
Collapse
Affiliation(s)
- Jason L Sperry
- Division of General Surgery andTrauma, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lucking SE, Maffei FA, Tamburro RF, Thomas NJ. Genetic Predisposition to Critical Illness in the Pediatric Intensive Care Unit. PEDIATRIC CRITICAL CARE STUDY GUIDE 2012. [PMCID: PMC7178837 DOI: 10.1007/978-0-85729-923-9_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Much progress has been made in the past decade in the understanding of the genetic contribution to the development of human disease in general, and critical care illness specifically. With the mapping of the human genome and on-going mapping of genetic polymorphisms and haplotypes in humans, the field of critical care is now in prime position to study the impact of genetics on common illnesses that affect children who require critical care, to examine how differences of the host defense response lead to variable outcomes in outwardly appearing similar disease states, and to study how genetic differences in response to therapy will help practitioners tailor therapeutic interventions to an individual child’s genetic composition. While we are still years away from true individualized medicine, we are now closer than ever to understanding why two might children respond to the same environmental insult in vastly different ways.
Collapse
Affiliation(s)
- Steven E. Lucking
- Children's Heart Group, Div. Pediatric Critical Care, Penn State Children's Hospital, University Drive 500, Hershey, 17078 Pennsylvania USA
| | - Frank A. Maffei
- Janet Weis Children's Hospital @ Geising, Pediatric Critical Care Medicine, Temple University School of Medicine, N. Academy Ave 100, Danville, 17822 Pennsylvania USA
| | - Robert F. Tamburro
- Milton S. Hershey Medical Center, Penn State College of Medicine, University Drive 500, Hershey, 17033-2390 Pennsylvania USA
| | - Neal J. Thomas
- College of Medicine, Penn State Children's Hospital, Pennsylvania State University, University Drive 500, Hershey, 17078 Pennsylvania USA
| |
Collapse
|
43
|
Reade MC, Yende S, Angus DC. Revisiting Mars and Venus: understanding gender differences in critical illness. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2011; 15:180. [PMID: 21888682 PMCID: PMC3387615 DOI: 10.1186/cc10319] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Understanding the nature and biological basis of gender-determined differences in risk of and outcome from infection might identify new therapeutic targets, allow more individualised treatment, and facilitate better risk prediction and application of healthcare resources. Gender differences in behaviours, comorbidities, access to healthcare and biology may result in differences in acquiring infection, or in response to infection once acquired. Some studies have reported higher male susceptibility to infection, and higher risk of death with sepsis, but others have found the opposite effect. The explanation for this disagreement is probably that different studies have included patients at different stages on the continuum from infectious agent exposure to death or recovery. Studying sufficient patient numbers to explore this entire continuum while accounting for heterogeneity in type of infection and comorbidity is difficult because of the number of patients required. However, if true gender effects can be identified, examination of their biological or psychosocial causes will be warranted.
Collapse
Affiliation(s)
- Michael C Reade
- Department of Intensive Care Medicine, Austin Hospital and University of Melbourne, Melbourne, VIC 3084, Australia
| | | | | |
Collapse
|
44
|
Thair SA, Walley KR, Nakada TA, McConechy MK, Boyd JH, Wellman H, Russell JA. A single nucleotide polymorphism in NF-κB inducing kinase is associated with mortality in septic shock. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:2321-8. [PMID: 21257964 DOI: 10.4049/jimmunol.1002864] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
We tested the hypothesis that single nucleotide polymorphisms (SNPs) within genes of the NF-κB pathway are associated with altered clinical outcome of septic shock patients. We genotyped 59 SNPs in the NF-κB pathway in a discovery cohort of septic shock patients (St. Paul's Hospital [SPH], N = 589), which identified the C allele of rs7222094 T/C within MAP3K14 (NF-κB inducing kinase; NIK) associated with increased 28-d mortality (uncorrected p = 0.00024, Bonferroni corrected p = 0.014). This result was replicated in a second cohort of septic shock patients (Vasopressin and Septic Shock Trial [VASST; N = 616]) in which the CC genotype of rs7222094 was associated with increased 28-d mortality (Cox regression: SPH cohort hazard ratio [HR], 1.35; 95% confidence interval [CI], 1.12-1.64; p = 0.002 Caucasian only; and VASST cohort HR, 1.24; 95% CI, 1.00-1.52; p = 0.048 Caucasian only). Patients having the CC genotype of rs7222094 in SPH experienced more renal and hematological dysfunction (p = 0.003 and p = 0.011), while patients of the VASST cohort with the rs7222094 CC genotype showed the same trend toward more renal dysfunction. In lymphoblastoid cell lines, we found the rs7222094 genotype most strongly associated with mRNA expression of CXCL10, a chemokine regulated by NF-κB. Accordingly, we measured CXCL10 protein levels and found that the CC genotype of rs7222094 was associated with significantly lower levels than those of the TT genotype in lymphoblastoid cell lines (p < 0.05) and in septic shock patients (p = 0.017). This suggests that the CC genotype of NIK rs7222094 is associated with increased mortality and organ dysfunction in septic shock patients, perhaps due to altered regulation of NF-κB pathway genes, including CXCL10.
Collapse
Affiliation(s)
- Simone A Thair
- University of British Columbia Critical Care Research Laboratories, Heart and Lung Institute, St. Paul's Hospital, Vancouver, British Columbia V6Z 1Y6, Canada
| | | | | | | | | | | | | |
Collapse
|
45
|
Interleukin-1 receptor-associated kinase-1: more evidence. Crit Care Med 2010; 38:2410-1. [PMID: 21088507 DOI: 10.1097/ccm.0b013e3181feb5fc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|