Genomic landscape of the individual host response and outcomes in sepsis: a prospective cohort study

Emma E Davenport, Katie L Burnham, Jayachandran Radhakrishnan, Peter Humburg, Paula Hutton, Tara C Mills, Anna Rautanen, Anthony C Gordon, Christopher Garrard, Adrian V S Hill, Charles J Hinds, Julian C Knight, Emma E Davenport, Katie L Burnham, Jayachandran Radhakrishnan, Peter Humburg, Paula Hutton, Tara C Mills, Anna Rautanen, Anthony C Gordon, Christopher Garrard, Adrian V S Hill, Charles J Hinds, Julian C Knight

Abstract

Background: Effective targeted therapy for sepsis requires an understanding of the heterogeneity in the individual host response to infection. We investigated this heterogeneity by defining interindividual variation in the transcriptome of patients with sepsis and related this to outcome and genetic diversity.

Methods: We assayed peripheral blood leucocyte global gene expression for a prospective discovery cohort of 265 adult patients admitted to UK intensive care units with sepsis due to community-acquired pneumonia and evidence of organ dysfunction. We then validated our findings in a replication cohort consisting of a further 106 patients. We mapped genomic determinants of variation in gene transcription between patients as expression quantitative trait loci (eQTL).

Findings: We discovered that following admission to intensive care, transcriptomic analysis of peripheral blood leucocytes defines two distinct sepsis response signatures (SRS1 and SRS2). The presence of SRS1 (detected in 108 [41%] patients in discovery cohort) identifies individuals with an immunosuppressed phenotype that included features of endotoxin tolerance, T-cell exhaustion, and downregulation of human leucocyte antigen (HLA) class II. SRS1 was associated with higher 14 day mortality than was SRS2 (discovery cohort hazard ratio (HR) 2·4, 95% CI 1·3-4·5, p=0·005; validation cohort HR 2·8, 95% CI 1·5-5·1, p=0·0007). We found that a predictive set of seven genes enabled the classification of patients as SRS1 or SRS2. We identified cis-acting and trans-acting eQTL for key immune and metabolic response genes and sepsis response networks. Sepsis eQTL were enriched in endotoxin-induced epigenetic marks and modulated the individual host response to sepsis, including effects specific to SRS group. We identified regulatory genetic variants involving key mediators of gene networks implicated in the hypoxic response and the switch to glycolysis that occurs in sepsis, including HIF1α and mTOR, and mediators of endotoxin tolerance, T-cell activation, and viral defence.

Interpretation: Our integrated genomics approach advances understanding of heterogeneity in sepsis by defining subgroups of patients with different immune response states and prognoses, as well as revealing the role of underlying genetic variation. Our findings provide new insights into the pathogenesis of sepsis and create opportunities for a precision medicine approach to enable targeted therapeutic intervention to improve sepsis outcomes.

Funding: European Commission, Medical Research Council (UK), and the Wellcome Trust.

Copyright © 2016 Davenport et al. Open Access article distributed under the terms of CC BY. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Transcriptomic sepsis response signatures Unsupervised hierarchical cluster analysis for the top 10% most variable probes (n=2619) for the 265 patients in the discovery cohort (A). First three PCs plotted with the proportion of variance explained by each component (B); individuals are coloured by group membership based on two groups assigned with k-means. Volcano plot (C) of differentially expressed probes for SRS1 versus SRS2 (red colouring shows fold change >1·5, false discovery rate <0·05). Most enriched functions, disease phenotypes, and predicted upstream regulators were derived from differentially expressed genes (D). Kaplan-Meier survival plot by SRS group (95% CIs shaded) for (E) discovery cohort and (F) validation cohort. SRS=sepsis response group. PC=principal component.
Figure 2
Figure 2
Overview of differentially expressed genes between SRS groups involving HLA and T-cell activation Red shading shows upregulation and green shading shows downregulation of genes (FC>1·5, FDR

Figure 3

Differentially expressed networks between SRS…

Figure 3

Differentially expressed networks between SRS The network with the lowest p value (p=1…

Figure 3
Differentially expressed networks between SRS The network with the lowest p value (p=1 × 10−35) identified from differential expression analysis of SRS1 versus SRS2 in the discovery cohort included 35 genes, with HIF1A and EPAS1 as nodes (A). Log FC shown with FDR. Sepsis cis-eQTL affecting genes in hypoxia network (B). Presence of cis-eQTL shown by red molecules with p values. eQTL=expression quantitative trait loci. SRS=sepsis response signature. FC=fold change. FDR=false discovery rate.

Figure 4

Sepsis eQTL Circos plot (A)…

Figure 4

Sepsis eQTL Circos plot (A) of, from outer rim inwards, Manhattan plot for…

Figure 4
Sepsis eQTL Circos plot (A) of, from outer rim inwards, Manhattan plot for cis-eQTL in patients with sepsis (FDR r2>70%) or examples of pathophysiological relevance shown; chromosome number; trans-eQTL (FDR<0·05) involving expression-associated SNPs also showing cis-eQTL (associated trans genes [orange] and SNPs [blue]). Box plots and local association plots for TLR4 and TNF (B). eQTL=expression quantitative trait loci. SNP=single-nucleotide polymorphism. FDR=false discovery rate.

Figure 5

Sepsis eQTL and epigenetic marks…

Figure 5

Sepsis eQTL and epigenetic marks Enrichment of expression-associated SNPs with the lowest p…

Figure 5
Sepsis eQTL and epigenetic marks Enrichment of expression-associated SNPs with the lowest p values for each gene (FDR−16.

Figure 6

Sepsis eQTL and response state…

Figure 6

Sepsis eQTL and response state SRS1-specific eQTL for MTOR (A,B). There is reduced…

Figure 6
Sepsis eQTL and response state SRS1-specific eQTL for MTOR (A,B). There is reduced expression in SRS1 individuals (FC 0·85, FDR 4·9 × 10−8). SRS2-specific eQTL for LAX1 (C,D). LAX1 is differentially expressed between SRS groups (0·44 FC, FDR 7·8 × 10−25). Box plots by allele for SRS1 and for SRS2 groups (A,C). Regional association plots (B,D). eQTL=expression quantitative trait loci. SRS=sepsis response signature. FC=fold change. FDR=false discovery rate.
Figure 3
Figure 3
Differentially expressed networks between SRS The network with the lowest p value (p=1 × 10−35) identified from differential expression analysis of SRS1 versus SRS2 in the discovery cohort included 35 genes, with HIF1A and EPAS1 as nodes (A). Log FC shown with FDR. Sepsis cis-eQTL affecting genes in hypoxia network (B). Presence of cis-eQTL shown by red molecules with p values. eQTL=expression quantitative trait loci. SRS=sepsis response signature. FC=fold change. FDR=false discovery rate.
Figure 4
Figure 4
Sepsis eQTL Circos plot (A) of, from outer rim inwards, Manhattan plot for cis-eQTL in patients with sepsis (FDR r2>70%) or examples of pathophysiological relevance shown; chromosome number; trans-eQTL (FDR<0·05) involving expression-associated SNPs also showing cis-eQTL (associated trans genes [orange] and SNPs [blue]). Box plots and local association plots for TLR4 and TNF (B). eQTL=expression quantitative trait loci. SNP=single-nucleotide polymorphism. FDR=false discovery rate.
Figure 5
Figure 5
Sepsis eQTL and epigenetic marks Enrichment of expression-associated SNPs with the lowest p values for each gene (FDR−16.
Figure 6
Figure 6
Sepsis eQTL and response state SRS1-specific eQTL for MTOR (A,B). There is reduced expression in SRS1 individuals (FC 0·85, FDR 4·9 × 10−8). SRS2-specific eQTL for LAX1 (C,D). LAX1 is differentially expressed between SRS groups (0·44 FC, FDR 7·8 × 10−25). Box plots by allele for SRS1 and for SRS2 groups (A,C). Regional association plots (B,D). eQTL=expression quantitative trait loci. SRS=sepsis response signature. FC=fold change. FDR=false discovery rate.

References

    1. Vincent JL, Opal SM, Marshall JC, Tracey KJ. Sepsis definitions: time for change. Lancet. 2013;381:774–775.
    1. Marshall JC. Why have clinical trials in sepsis failed? Trends Mol Med. 2014;20:195–203.
    1. Cohen J, Vincent JL, Adhikari NK. Sepsis: a roadmap for future research. Lancet Infect Dis. 2015;15:581–614.
    1. Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol. 2013;13:862–874.
    1. Chapman SJ, Hill AV. Human genetic susceptibility to infectious disease. Nat Rev Genet. 2012;13:175–188.
    1. Rautanen A, Mills TC, Gordon AC. Genome-wide association study of survival from sepsis due to pneumonia: an observational cohort study. Lancet Respir Med. 2015;3:53–60.
    1. GTEx Consortium The Genotype-Tissue Expression (GTEx) pilot analysis: Multitissue gene regulation in humans. Science. 2015;348:648–660.
    1. Fairfax BP, Humburg P, Makino S. Innate immune activity conditions the effect of regulatory variants upon monocyte gene expression. Science. 2014;343:1246949.
    1. Knaus WA, Draper EA, Wagner DP, Zimmerman JE. APACHE II: a severity of disease classification system. Crit Care Med. 1985;13:818–829.
    1. Vincent JL, Moreno R, Takala J. The SOFA (Sepsis-related Organ Failure Assessment) score to describe organ dysfunction/failure. On behalf of the Working Group on Sepsis-Related Problems of the European Society of Intensive Care Medicine. Intensive Care Med. 1996;22:707–710.
    1. Kiiveri HT. A general approach to simultaneous model fitting and variable elimination in response models for biological data with many more variables than observations. BMC Bioinformatics. 2008;9:195.
    1. Arlot S, Celisse A. A survey of cross-validation procedures for model selection. Stat Surv. 2010;4:40–79.
    1. del Fresno C, Garcia-Rio F, Gomez-Pina V. Potent phagocytic activity with impaired antigen presentation identifying lipopolysaccharide-tolerant human monocytes: demonstration in isolated monocytes from cystic fibrosis patients. J Immunol. 2009;182:6494–6507.
    1. Pena OM, Pistolic J, Raj D, Fjell CD, Hancock RE. Endotoxin tolerance represents a distinctive state of alternative polarization (M2) in human mononuclear cells. J Immunol. 2011;186:7243–7254.
    1. Wu D, Lim E, Vaillant F, Asselin-Labat ML, Visvader JE, Smyth GK. ROAST: rotation gene set tests for complex microarray experiments. Bioinformatics. 2010;26:2176–2182.
    1. Biswas S, Storey JD, Akey JM. Mapping gene expression quantitative trait loci by singular value decomposition and independent component analysis. BMC Bioinformatics. 2008;9:244.
    1. Westra HJ, Peters MJ, Esko T. Systematic identification of trans eQTLs as putative drivers of known disease associations. Nat Genet. 2013;45:1238–1243.
    1. Saeed S, Quintin J, Kerstens HH. Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity. Science. 2014;345:1251086.
    1. Fairfax BP, Makino S, Radhakrishnan J. Genetics of gene expression in primary immune cells identifies cell type-specific master regulators and roles of HLA alleles. Nat Genet. 2012;44:502–510.
    1. Bryois J, Buil A, Evans DM. Cis and trans effects of human genomic variants on gene expression. PLoS Genet. 2014;10:e1004461.
    1. Dimas AS, Deutsch S, Stranger BE. Common regulatory variation impacts gene expression in a cell type-dependent manner. Science. 2009;325:1246–1250.
    1. Hutchins NA, Unsinger J, Hotchkiss RS, Ayala A. The new normal: immunomodulatory agents against sepsis immune suppression. Trends Mol Med. 2014;20:224–233.
    1. Pachot A, Lepape A, Vey S, Bienvenu J, Mougin B, Monneret G. Systemic transcriptional analysis in survivor and non-survivor septic shock patients: a preliminary study. Immunol Lett. 2006;106:63–71.
    1. Tang BM, Huang SJ, McLean AS. Genome-wide transcription profiling of human sepsis: a systematic review. Crit Care. 2010;14:R237.
    1. Biswas SK, Lopez-Collazo E. Endotoxin tolerance: new mechanisms, molecules and clinical significance. Trends Immunol. 2009;30:475–487.
    1. Pena OM, Hancock DG, Lyle NH. An Endotoxin Tolerance Signature Predicts Sepsis and Organ Dysfunction at Initial Clinical Presentation. EBioMedicine. 2014;1:64–71.
    1. Pachot A, Monneret G, Brion A. Messenger RNA expression of major histocompatibility complex class II genes in whole blood from septic shock patients. Crit Care Med. 2005;33:31–38.
    1. Boomer JS, To K, Chang KC. Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA. 2011;306:2594–2605.
    1. Falasca L, Farrace MG, Rinaldi A, Tuosto L, Melino G, Piacentini M. Transglutaminase type II is involved in the pathogenesis of endotoxic shock. J Immunol. 2008;180:2616–2624.
    1. Cai S, Lee CC, Kohwi-Shigematsu T. SATB1 packages densely looped, transcriptionally active chromatin for coordinated expression of cytokine genes. Nat Genet. 2006;38:1278–1288.
    1. Caliskan M, Baker SW, Gilad Y, Ober C. Host genetic variation influences gene expression response to rhinovirus infection. PLoS Genet. 2015;11:e1005111.
    1. Lill M, Koks S, Soomets U. Peripheral blood RNA gene expression profiling in patients with bacterial meningitis. Front Neurosci. 2013;7:33.
    1. Vanden Berghe T, Demon D, Bogaert P. Simultaneous targeting of IL-1 and IL-18 is required for protection against inflammatory and septic shock. Am J Respir Crit Care Med. 2014;189:282–291.
    1. Kirino Y, Bertsias G, Ishigatsubo Y. Genome-wide association analysis identifies new susceptibility loci for Behcet's disease and epistasis between HLA-B*51 and ERAP1. Nat Genet. 2013;45:202–207.
    1. He M, Horuk R, Moochhala SM, Bhatia M. Treatment with BX471, a CC chemokine receptor 1 antagonist, attenuates systemic inflammatory response during sepsis. Am J Physiol Gastrointest Liver Physiol. 2007;292:G1173–G1180.
    1. Venet F, Lepape A, Debard AL, Bienvenu J, Bohe J, Monneret G. The Th2 response as monitored by CRTH2 or CCR3 expression is severely decreased during septic shock. Clin Immunol. 2004;113:278–284.
    1. Arlt A, Schafer H. Role of the immediate early response 3 (IER3) gene in cellular stress response, inflammation and tumorigenesis. Eur J Cell Biol. 2011;90:545–552.
    1. Pham CT, Ley TJ. Dipeptidyl peptidase I is required for the processing and activation of granzymes A and B in vivo. Proc Natl Acad Sci USA. 1999;96:8627–8632.
    1. Brinkmann V, Reichard U, Goosmann C. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–1535.
    1. Shalova IN, Lim JY, Chittezhath M. Human monocytes undergo functional re-programming during sepsis mediated by hypoxia-inducible Factor-1α. Immunity. 2015;42:484–498.
    1. Cheng SC, Quintin J, Cramer RA. mTOR- and HIF-1alpha-mediated aerobic glycolysis as metabolic basis for trained immunity. Science. 2014;345:1250684.
    1. Bakker J, Gris P, Coffernils M, Kahn RJ, Vincent JL. Serial blood lactate levels can predict the development of multiple organ failure following septic shock. Am J Surg. 1996;171:221–226.
    1. Takaoka Y, Goto S, Nakano T. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) prevents lipopolysaccharide (LPS)-induced, sepsis-related severe acute lung injury in mice. Sci Rep. 2014;4:5204.
    1. Liu TF, Vachharajani V, Millet P, Bharadwaj MS, Molina AJ, McCall CE. Sequential actions of SIRT1-RELB-SIRT3 coordinate nuclear-mitochondrial communication during immunometabolic adaptation to acute inflammation and sepsis. J Biol Chem. 2015;290:396–408.
    1. Villar J, Ribeiro SP, Mullen JB, Kuliszewski M, Post M, Slutsky AS. Induction of the heat shock response reduces mortality rate and organ damage in a sepsis-induced acute lung injury model. Crit Care Med. 1994;22:914–921.
    1. Banton MC, Inder KL, Valk E, Rudd CE, Schneider H. Rab8 binding to immune cell-specific adaptor LAX facilitates formation of trans-Golgi network-proximal CTLA-4 vesicles for surface expression. Mol Cell Biol. 2014;34:1486–1499.
    1. Yang B, Wang J, Wang Y. Novel function of Trim44 promotes an antiviral response by stabilizing VISA. J Immunol. 2013;190:3613–3619.
    1. Ma Z, Moore R, Xu X, Barber GN. DDX24 negatively regulates cytosolic RNA-mediated innate immune signaling. PLoS Pathog. 2013;9:e1003721.
    1. Scicluna BP, Klein Klouwenberg PM, van Vught LA. A molecular biomarker to diagnose community-acquired pneumonia on intensive care unit admission. Am J Respir Crit Care Med. 2015;192:826–835.
    1. Wong HR, Cvijanovich NZ, Anas N. Developing a clinically feasible personalized medicine approach to pediatric septic shock. Am J Respir Crit Care Med. 2015;191:309–315.

Source: PubMed

3
Suscribir