Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 May 1.
Published in final edited form as: Crit Care Med. 2012 May;40(5):1618–1626. doi: 10.1097/CCM.0b013e318246b546

GENETICS AND GENOMICS IN PEDIATRIC SEPTIC SHOCK

Hector R Wong 1
PMCID: PMC3329642  NIHMSID: NIHMS350315  PMID: 22511139

Abstract

Objectives

Pediatric septic shock continues to be an important public health problem. Several investigative groups have applied genetic and genomic approaches as a means of identifying novel pathways and therapeutic targets, discovery of sepsis-related biomarkers, and identification of septic shock subclasses. This review will highlight studies in pediatric sepsis with a focus on gene association studies and genome-wide expression profiling.

Data Sources

A summary of published literature involving gene association and expression profiling studies specifically involving pediatric sepsis and septic shock.

Summary

Several polymorphisms of genes broadly involved in inflammation, immunity, and coagulation have been linked with susceptibility to sepsis, or outcome of sepsis in children. Many of these studies involve meningococcemia, and the strongest association involves a functional polymorphism of the plasminogen activator inhibitor-1 promoter region and meningococcal sepsis. Expression profiling studies in pediatric septic shock have identified zinc supplementation and inhibition of matrix metalloproteinase-8 activity as potential, novel therapeutic approaches in sepsis. Studies focused on discovery of sepsis-related biomarkers have identified interleukin-8 as a robust outcome biomarker in pediatric septic shock. Additional studies have demonstrated the feasibility and clinical relevance of gene expression-based subclassification of pediatric septic shock.

Conclusions

Pediatric sepsis and septic shock are increasingly being studied by genetic and genomic approaches and the accumulating data hold the promise of enhancing our future approach to this ongoing clinical problem.

INTRODUCTION

Pediatric septic shock remains a major public health problem despite the development of effective antibiotics, vaccines, intensive care unit-based support modalities, and standardized treatment guidelines [15]. The recognition of septic shock as a persistent challenge in the pediatric intensive care unit (PICU), has led several investigators to study this syndrome using genetic and genomic approaches [68]. This review will focus on the two areas of genomics most widely applied thus far to the field of pediatric septic shock: gene association studies and genome-wide expression profiling. The concluding section will briefly speculate on the potential link between epigenetics and long term outcomes.

GENE ASSOCIATION STUDIES

Death from infection is reported to have a stronger heritable component than death from cancer or cardiovascular disease [9]. While this observation did not identify the causative genetic alterations and involves a relatively non-contemporary patient cohort, the study nonetheless provides compelling evidence that genetics play an important role in both susceptibility and response to infection. The existence of a singular “sepsis gene” is not biologically plausible. More plausible is the existence of genetic variations within multiple candidate genes that affect how the host responds to an infectious challenge.

The majority of gene association studies involving pediatric sepsis have focused on polymorphisms: the regular occurrence (>1%) of two or more alleles at a particular chromosome location. The most frequent type of polymorphism is called a single nucleotide polymorphism (SNP): a substitution, deletion, or insertion of a single nucleotide occurring in approximately 1 per 1,000 base pairs of human DNA. SNPs can result in an altered protein, a change in the amount of normal protein expression, or no discernable change in protein function.

Many SNPs fall into the latter category because they occur in non-coding regions, or they are synonymous SNPs that do not lead to an amino acid change. These SNPs may be nonetheless worthy of study because they may be co-inherited along with causal variants through linkage disequilibrium (LD), which refers to the non-random association of alleles at two or more chromosome locations. Related to the concept of LD is that of haplotype, which refers to a set of multiple SNPs on a single chromosome that are typically co-inherited. These haplotype “blocks” can be identified by haplotype tag SNPs, and the International HapMap project is developing a haplotype map of the entire human genome as means to more effectively conduct genetic association studies [10].

A selected group of gene association studies in pediatrics sepsis will be discussed below. Several reviews exist on the topic involving both adults and children, which also discuss methodological issues and limitations [6, 1115]. Notably, there are rigorous criteria dictating the quality of an ideal gene association study [1620]. Unfortunately, many gene association studies in critical care medicine do not meet this level of rigor [19].

Plasminogen activator inhibitor-1 (PAI-1)

PAI-1 is the principal inhibitor of tissue plasminogen activator and urokinase [21, 22]. As such, PAI-1 can be viewed as a pro-coagulant factor in that it inhibits fibrinolysis. The PAI-1 promoter contains an insertion/deletion polymorphism at -675 bp: 5G/4G [23]. In vitro, the 4G allele produces six times the amount of PAI-1 mRNA compared to the 5G allele, and individuals homozygous for the 4G allele produce greater amounts of PAI-1 compared to individuals homozygous for the 5G allele [23].

Microvascular thrombosis is a common pathologic component of sepsis, particularly in meningococcemia [2427]. Accordingly, the PAI-1 4G/5G polymorphism has been a focus of investigation in meningococcal sepsis. Hermans et al. first demonstrated that children with meningococcemia and the 4G/4G genotype, produce higher concentrations of systemic PAI-1 and have worse outcomes, compared to children with the 4G/5G or 5G/5G genotype [28]. This link between the 4G/4G genotype and severity of meningococcal disease has been independently corroborated by several investigators [2931], and a recent meta-analysis provides further confirmation that the 4G/4G allele is associated with mortality [32].

The studies linking the PAI-1 4G/5G insertion/deletion polymorphism and meningococcal sepsis provide relevant examples of well-conducted gene association studies [1620]. The association between the gene and the disease has a high level of biological plausibility. The allele (4G) affects the gene product in a physiologically meaningful manner. Cases are clearly defined and represent a spectrum of disease severity. The initial study has been independently replicated. Thus, the impact of the 4G allele on outcome in meningococcal disease is perhaps the most well-founded association between genetic variation and outcome in pediatric septic shock. However, this “genomic knowledge” has yet to be unambiguously translated to the bedside in the form of a novel therapy. This unfortunate circumstance reflects the complexities of coagulation balance in septic shock, as demonstrated by the inability of activated protein-C therapy to improve outcome in a heterogeneous cohort of children with septic shock [33].

Toll like receptors (TLRs) and related signaling molecules

TLRs are a family of pathogen recognition receptors that provide a major mechanism for cells of the innate immune system to recognize and respond to pathogens [34]. TLR4 is responsible for recognizing lipopolysaccharide from gram negative bacteria, while TLR2 is responsible for recognizing cell wall components of gram positive bacteria (lipoteichoic acid and peptidoglycan).

The coding region of the human TLR2 gene contains a non-synonymous SNP leading to a substitution of arginine, for glutamine, at amino acid 753 (Arg753Gln). Lorenz et al. first reported this polymorphism and that the Arg753Gln polymorphism renders TLR2 less responsive to components of gram positive bacteria [35]. Lorenz et al. also detected this polymorphism in 2 of 91 patients with septic shock, both of whom had Staphylococcal infections [35]. Subsequent studies, however, have not been able to confirm a strong association between the Arg753Gln allele and severity of gram positive infection in adults [36, 37]. Studies in pediatric populations indicate an association between the Arg753Gln polymorphism and risk of recurrent infection [38], urinary tract infection [39], premature birth [40], and acute rheumatic fever [41]. Other TLR2 SNPs have been described that may warrant further investigation [42, 43].

TLR4 mutations exist in mice that lead to abnormal responses to endotoxin and increase susceptibility to gram negative infections [4447]. The human TLR4 gene contains two mutations (Asp299Gly and Thr399Ile) that lead to hypo-responsiveness when human volunteers are challenged with inhaled endotoxin [48, 49]. Conversely, a study involving peripheral blood mononuclear cells from children, showed no differential response to endotoxin or respiratory syncytial (RSV) in association with these two mutations [50]. Nonetheless, studies comparing adults with septic shock and healthy blood donor controls revealed the TLR4 Asp299Gly allele exclusively in the patients with septic shock, and also found that patients with the Asp299Gly/Thr399Ile alleles had a higher prevalence of gram-negative infections [51, 52]. In one report involving children with meningococcal disease, a heterozygous Asp299Gly genotype was associated with increased mortality [53], while two other reports have not been able corroborate this association [54, 55]. Smirnova et al have reported no link between “common” TLR4 variants and meningococcal disease, but have provided evidence that “rare” TLR4 coding variants are substantially over-represented in patients with meningococcal disease [56]. Finally, TLR4 polymorphisms have been linked with susceptibility to malaria in children [57].

Despite the biological importance of TLRs (a focus of the most recent Nobel Prize in Medicine) an absolute and unambiguous link between TLR genetic variants and human septic shock remains relatively elusive. Accordingly, investigators have recently focused on adapter proteins constituting the downstream signaling apparatus of TLRs. Polymorphisms of one such adapter protein, Mal (a.k.a. TIRAP), have been linked to invasive pneumococcal disease [58], an evolution-related increased resistance to infection [59], increased risk of infection in critically ill adults [60], and susceptibility to invasive Haemophilus influenzae infection in immunized children [61]. Given the existence of several other adapter proteins that contribute to TLR signaling [34], it would be expected that several other gene association studies, focused on these adapter protein genes, are forthcoming.

Tumor necrosis factor-α (TNFα)

TNFα is recognized as a primary mediator in the pathophysiology of sepsis and septic shock [6264], and has well described polymorphisms [65]. A substitution polymorphism of the TNFα promoter region involves a guanine (TNF1 allele) or an adenine (TNF2 allele) a −308 base pairs [66], and the TNF2 allele correlates with increased production of TNFα [6769]. The TNF2 allele has been associated with increased susceptibility to septic shock and mortality from septic shock in adults [70]. However, this association has not been consistently observed [71, 72], and a recent meta-analysis involving 25 selected articles concluded that the TNF2 allele is associated with the development of sepsis, but not with sepsis mortality [73]. In children with meningococcemia, Nadel et al. reported an association between the TNF2 allele and illness severity [74], whereas Read et al. reported an association between the TNF2 allele and susceptibility to meningococcemia in a mixed population of adults and children. A small study involving children with heterogeneous sepsis etiologies suggested that the TNF2 allele is more common in patients with septic shock compared to normal controls, but could not detect an association between the TNF2 allele and mortality [75].

A related polymorphism involves lymphotoxin-α, a member of the TNF superfamily (TNF-β) [74]. The first intron of the lymphotoxin-α gene contains a restriction length polymorphism: the TNFB1 and TNFB2 alleles. Adults with septic shock, and homozygous for the TNFB2 allele, are characterized by higher systemic levels of TNFα and a higher mortality rate [76]. In bacteremic children, the TNFB2 allele was also demonstrated to be associated with higher systemic levels of TNFα and higher mortality [77].

Summary and perspective

Several other polymorphisms have been studied in pediatric sepsis and a selected group is summarized in a Table. Presently, no gene association study has directly impacted care in the PICU. Nonetheless, the concept of genetics influencing pediatric sepsis remains valid. In order to translate this concept to the bedside, large scale collaborations will need to be developed, positive association studies will need to be validated, and the field should consider focusing on functional polymorphisms for which there potentially exist reasonable therapeutic options.

Table 1.

Selected gene association studies in pediatric sepsis and septic shock

Reference Gene/polymorphism Main findings
Read, et al. [143] Brouwer et al. [32] Polymorphisms of interleukin-1 (IL1B (−511)) and IL-1 receptor antagonist (IL1RN (+2018)). IL1B (−511) allele associated with increased survival in meningococcemia. Combination of the IL1B (−511) and IL1RN (+2018) alleles associated with decreased survival.
Endler, et al. [144] Brouwer et al. [32] Multiple polymorphisms for the IL-1 locus. The IL1RA (+2018) polymorphism was associated with risk of meningococcal disease and with its outcome.
Michalek, et al. [145] IL-6 polymorphisms (G-174C and G-572C). Both polymorphisms could be predictors of risk of development and/or predictors of sepsis severity.
Lehrnbecher, et al. [146] IL-6 G-174C polymorphism Population of children with acute myeloid leukemia. G allele associated with risk of infection with gram negative bacteria.
Artifoni, et al. [147] IL-8 −251 A>T polymorphism A allele associated with pyelonephritis.
Binder, et al. [148] Polymorphisms of the protein C promoter: C-1654T and A-1641G Carriers of the CG allele had an increased risk of developing meningococcal sepsis.
Multiple [149153] Fc gamma receptor polymorphisms Increased risk of meningococcal disease and increased illness severity.
Hibberd et al. [154] Mannose binding lectin (MBL) polymorphisms Increased susceptibility to meningococcal disease.
Summerfield et al. [155] MBL polymorphisms Increased susceptibility to severe infections.
Koch et al. [156] MBL polymorphisms Increased risk of acute respiratory infections in children 6 to 17 months of age.
Michalek et al. [157] Bactericidal permeability increasing (BPI) protein polymorphisms Increased risk of gram negative sepsis and increased risk of death.
El Saleeby et al. [158] Surfactant protein A2 polymorphisms Increased illness severity in infants with RSV infection.
Dahmer et al. [159] Surfactant protein B polymorphisms Increased severity of acute lung injury after community acquired pneumonia in African-American children.
Agbeko et al. [160] Functional polymorphisms of the complement activation cascade. Homozygosity for the complement factor H Y402H polymorphism carries a decreased risk of sepsis.
Haralambous et al. [161] Davila et al. [162] Complement factor H polymorphisms Increased risk of invasive meningococcal disease, in association with increased serum factor H levels and reduced bactericidal activity against menigococcus.
Harding et al. [163] Insertion (I)/deletion (D) polymorphism of angiotensin converting enzyme (ACE). DD genotype associated with increased illness severity in meningococcal disease.
Cogulu et al. [164] ACE I/D polymorphism. DD genotype associated with decreased risk of sepsis.
Tekin et al. [165] NOD2 receptor (pathogen recognition receptor) polymorphism. Gene variants of the NOD2 receptor associated with increased risk of sepsis and increased illness severity.
Khor et al. [166] Cytokine-inducible SRC homology 2 domain protein (CISH) polymorphisms: a suppressor of cytokine signaling. CISH variants are associated with increased risk of various types of infections in a mixed population of adults and children. Over 8,000 individuals sampled.

EXPRESSION PROFILING STUDIES

Expression profiling involves the use of microarray technology to simultaneously measure mRNA abundance of thousands of transcripts from biological specimens [78, 79]. The approach is said to be “discovery oriented” in that no a priori assumptions are made regarding the relevance of any particular genes to the biological process of interest. This relatively unbiased, whole-genome approach is also referred to as “transcriptomics”, and is generally hypothesis-generating, rather than hypothesis-driven (Figure). Several whole-genome expression profiling studies have been conducted in human volunteers challenged with endotoxin and adults with sepsis [8091] and excellent reviews have detailed the technical aspects, caveats, and limitations of expression profiling [78, 79]. This section will review the analogous studies involving children, with a focus on leveraging expression data for the discovery of novel pathways and therapeutic targets, biomarker discovery, and gene expression-based subclassification.

Figure 1.

Figure 1

Schematic illustrating the discovery-oriented and hypothesis-generating approach of whole-genome expression profiling. The potential deliverables of expression profiling data include the discovery of novel pathways and therapeutic targets, biomarker discovery, and expression-based subclassification of patients with septic shock.

Discovery of novel pathways and therapeutic targets

The ability to interrogate the entire genome provides an opportunity to discover previously unrecognized targets and pathways relevant to sepsis biology. For example, Pathan et al have taken this approach to address the phenomenon of myocardial dysfunction in meningococcal sepsis [92]. Using a combination of expression profiling and in vitro approaches, these investigators identified interleukin-6 as a major contributor to myocardial depression in meningococcal sepsis.

Multiple expression profiling studies in children with septic shock have documented early and persistent repression of gene programs directly related to, or dependent on, zinc homeostasis, as well as low serum zinc concentrations [9397]. Since normal zinc homeostasis is critical for normal immune function [98], these observations raise the possibility of zinc supplementation as a potential therapeutic strategy for sepsis [99101]. Independent of the pediatric studies, Knoell et al. demonstrated that zinc supplementation is a beneficial strategy in experimental sepsis [102, 103]. Additional studies by Knoell et al. corroborated decreased plasma zinc concentrations in patients with sepsis, and a correlation between low plasma zinc concentrations and higher illness severity [104]. These same investigators have also reported that plasma zinc concentrations correlate inversely with monocyte expression of the zinc transporter gene SLC39A8 [104, 105], and expression profiling studies in children with septic shock have corroborated high level SLC39A8 expression in non-survivors, relative to survivors [97].

Despite this interesting convergence of independent data sources, the safety and efficacy of zinc supplementation in clinical sepsis remains to be directly demonstrated. The pediatric critical illness stress-induced immune suppression (CRISIS) trial tested the efficacy of enteral zinc supplementation, along with selenium, glutamine, and metoclopramide, as a means of preventing nosocomial infection or sepsis in critically ill children [106]. This trial was terminated early for futility (http://clinicaltrials.gov; NCT00395161). Potential confounders in this trial included the testing of multiple agents, thus making it difficult to assess the effect of any single agent [107, 108], and decreased bioavailability of enteral zinc [100]. Consequently, there is an active Phase 1 trial involving intravenous zinc supplementation in critically ill children (NCT01062009).

In multiple studies involving children with septic shock, metalloproteinase-8 (MMP-8) has consistently been the highest expressed gene in patients with septic shock, relative to normal controls [9397, 109, 110]. MMP-8 is also more highly expressed in patients with septic shock, compared to patients with sepsis, and in septic shock non-survivors, compared to septic shock survivors [111]. While MMP-8 is best known as a neutrophil-derived protease that cleaves extracellular matrix (ECM) collagen, MMP-8 has other cellular sources and non-ECM substrates [112]. The discovery of high level MMP-8 expression in clinical septic shock has led to the formal study of MMP-8 in experimental sepsis. These studies demonstrated that pharmacologic inhibition of MMP-8, or genetic ablation of MMP-8, confers a significant survival advantage in a murine model of sepsis [111]. Collectively, these studies identify MMP-8 as a novel, candidate therapeutic target in sepsis, and this assertion is particularly intriguing given the existence of drugs to inhibit MMP-8 activity in the clinical setting [113].

Triggering receptor expressed on myeloid cells 1 (TREM-1) is critical for amplification of the inflammatory response to pathogen challenge and there is interest in blockade of the TREM-1 pathway in sepsis [114]. A recent gene expression profiling study in pediatric septic shock compared 4 distinct developmental age groups [110]. A primary finding of this study was that children in the “neonate” group (0 to 28 days of age) had widespread repression of genes corresponding to the TREM-1 signaling pathway, compared to older children. The observation that the TREM-1 pathway may not be particularly active in neonates with sepsis illustrates how some candidate therapeutic strategies may not have a biological basis across all developmental age groups.

Biomarker discovery

The diagnostic approach to the febrile child without an obvious source of infection, and distinguishing viral from bacterial infection, remain important challenges in clinical pediatrics [115]. Ramilo et al. have applied gene expression profiling to differentiate bacterial versus viral infection in hospitalized febrile children [116]. Specifically, they have reported expression signatures that can distinguish Influenza A infection from bacterial infection, and E. coli infection from S. aureus infection. In a conceptually analogous study, Allantaz et al. reported a gene expression signature that differentiates children with systemic onset juvenile idiopathic arthritis (e.g. “sterile inflammation) from children with acute bacterial or viral systemic infections [117]. These data provide a foundation to better address an important problem in clinical pediatrics.

Another area of interest for sepsis-related biomarker discovery involves outcome biomarkers [118120]. Expression profiling experiments in children with septic shock identified interlerukin-8 (IL-8) as a differentially regulated gene between survivors and non-survivors, and this observation was validated by serum IL-8 protein measurements [97]. A subsequent study tested the ability of serum IL-8 levels, measured within 24 hours of admission to the PICU, to predict survival/non-survival in pediatric septic shock [121]. Using separate derivation and validation cohorts, this study demonstrated that serum IL-8 measurements could predict a 95% probability of survival with standard care. Interestingly, IL-8 was not able to predict survival with this degree of robustness in a cohort of adults with septic shock [122]. It has been proposed that serum IL-8 levels can be used to exclude children from future interventional clinical trials as a means of improving the risk to benefit ratio of a given therapy [121]. Using a similar approach, Nowak et al. identified chemokine ligand (C-C motif) ligand 4 as an outcome biomarker in pediatric septic shock, but this observation remains to be validated [123].

Currently, there is an ongoing effort to derive and validate a multi-biomarker sepsis outcome risk model in pediatric septic shock. The foundation of this effort is the unbiased selection of a panel of candidate outcome biomarkers using microarray data from a large cohort of children with septic shock [118, 124].

Gene expression-based septic shock subclasses

Viewing septic shock as a heterogeneous syndrome implies the existence of “disease subclasses”, analogous to the oncology field [119, 120]. Recent studies reported pediatric septic shock subclasses based exclusively on genome-wide expression profiles. In the initial study, 3 subclasses of children with septic shock (subclasses “A”, “B”, and “C”) were identified using a computer algorithm (unsupervised hierarchical clustering) that groups patients based on statistically similar patterns of gene expression, with no a priori knowledge of the clinical phenotype [96]. Post hoc analysis of the clinical subclass phenotypes revealed that subclass A patients had a significantly higher level of illness severity, including mortality.

Recognizing that standard genomic data outputs are not clinically intuitive, a subsequent study explored the feasibility of bringing expression-based subclassification closer to the bedside. The subclass-defining gene expression patterns were distilled to a 100 gene expression signature and depicted using visually intuitive gene expression mosaics [125127]. Clinicians, without any bioinformatics training, were able to reliably allocate patients to the correct subclasses with a high degree of sensitivity and specificity. In a follow-up study, the 100 gene expression signature and the expression mosaics were used to classify a separate validation cohort, and again, the subclass A patients were characterized by higher illness severity [128]. Thus, gene expression-based subclassification of pediatric septic shock is feasible and clinically relevant. The assertion of clinical relevance is further substantiated given that the 100 class defining genes correspond to adaptive immunity, glucocorticoid receptor signaling, and peroxisome proliferator activated receptor-α signaling [128].

EPIGENETICS

Epigenetics refers to heritable changes in gene expression that are not related to direct DNA sequence changes [129]. The epigenetic mechanisms dictating increased or decreased gene expression include chemical modifications of DNA and post-translational modifications of histones. A key concept of epigenetics is that the epigenetic modifications can be “inherited” (i.e. passed on to daughter cells) and can therefore lead to long lasting effects on gene expression.

Immunity- and inflammation-related genes are subject to epigenetic regulation [130137], and experimental data indicate that sepsis induces epigenetic changes in dendritic cells and lymphocytes rendering the host immune-deficient for a long period after the initial sepsis challenge [138140]. In children with septic shock, there is evidence of differential expression of genes involved in epigenetic regulation, in parallel with suppression of adaptive immunity genes [109].

Patients that recover from critical illness, sepsis in particular, are at increased risk of death for several years after discharge [5, 141, 142]. Czaja et al. recently studied over 7,000 pediatric severe sepsis cases [5]. Almost one-half of the patients that were discharged after the initial admission were re-admitted at least once, at a median of 3 months after discharge. Respiratory infection was the most common indication for readmission and >30% of these readmissions were in children without co-morbidities. An additional 6.5% of patients died during these readmissions. While the cause of these late deaths and the high rate of readmission are likely to be multi-factorial, it is tempting to speculate on a potential role for epigenetic mechanisms involving the immune system.

CONCLUSIONS

Genetic/genomic approaches to pediatric septic shock have proliferated over the last decade. While novel information has been derived from these studies, it must be kept in mind that none of these data have been directly translated to the bedside of the critically ill child, yet. Meeting the lofty goal of clinical translation will require multi-investigator collaborations and further rigorous studies with an emphasis on independent validation. The potential deliverables of clinical translation include robust and clinically effective patient stratification strategies, and novel therapies, which will enhance, rather than replace, our current clinical protocols and guidelines.

Acknowledgments

Supported by NIH grants R01-GM064619, R01GM096994, and RC1HL100474

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCES

  • 1.Odetola FO, Gebremariam A, Freed GL. Patient and hospital correlates of clinical outcomes and resource utilization in severe pediatric sepsis. Pediatrics. 2007;119(3):487–494. doi: 10.1542/peds.2006-2353. [DOI] [PubMed] [Google Scholar]
  • 2.Markovitz BP, Goodman DM, Watson RS, Bertoch D, Zimmerman J. A retrospective cohort study of prognostic factors associated with outcome in pediatric severe sepsis: what is the role of steroids? Pediatr Crit Care Med. 2005;6(3):270–274. doi: 10.1097/01.PCC.0000160596.31238.72. [DOI] [PubMed] [Google Scholar]
  • 3.Watson RS, Carcillo JA, Linde-Zwirble WT, Clermont G, Lidicker J, Angus DC. The epidemiology of severe sepsis in children in the United States. Am J Respir Crit Care Med. 2003;167(5):695–701. doi: 10.1164/rccm.200207-682OC. [DOI] [PubMed] [Google Scholar]
  • 4.Watson RS, Carcillo JA. Scope and epidemiology of pediatric sepsis. Pediatr Crit Care Med. 2005;6(3 Suppl):S3–S5. doi: 10.1097/01.PCC.0000161289.22464.C3. [DOI] [PubMed] [Google Scholar]
  • 5.Czaja AS, Zimmerman JJ, Nathens AB. Readmission and late mortality after pediatric severe sepsis. Pediatrics. 2009;123(3):849–857. doi: 10.1542/peds.2008-0856. [DOI] [PubMed] [Google Scholar]
  • 6.Cornell TT, Wynn J, Shanley TP, Wheeler DS, Wong HR. Mechanisms and regulation of the gene-expression response to sepsis. Pediatrics. 2010;125(6):1248–1258. doi: 10.1542/peds.2009-3274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Wong HR. Pediatric septic shock treatment: new clues from genomic profiling. Pharmacogenomics. 2007;8(10):1287–1290. doi: 10.2217/14622416.8.10.1287. [DOI] [PubMed] [Google Scholar]
  • 8.Shanley TP, Wong HR. Molecular genetics in the pediatric intensive care unit. Crit Care Clin. 2003;19(3):577–594. doi: 10.1016/s0749-0704(03)00004-6. [DOI] [PubMed] [Google Scholar]
  • 9.Sorensen TI, Nielsen GG, Andersen PK, Teasdale TW. Genetic and environmental influences on premature death in adult adoptees. N Engl J Med. 1988;318(12):727–732. doi: 10.1056/NEJM198803243181202. [DOI] [PubMed] [Google Scholar]
  • 10.Manolio TA, Collins FS. The HapMap and genome-wide association studies in diagnosis and therapy. Annu Rev Med. 2009;60:443–456. doi: 10.1146/annurev.med.60.061907.093117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Dahmer MK, Randolph A, Vitali S, Quasney MW. Genetic polymorphisms in sepsis. Pediatr Crit Care Med. 2005;6(3 Suppl):S61–S73. doi: 10.1097/01.PCC.0000161970.44470.C7. [DOI] [PubMed] [Google Scholar]
  • 12.Liangos O, Jaber BL. Multiple organ dysfunction syndrome in children with sepsis: role of genetic factors. Semin Nephrol. 2008;28(5):499–509. doi: 10.1016/j.semnephrol.2008.05.011. [DOI] [PubMed] [Google Scholar]
  • 13.Namath A, Patterson AJ. Genetic polymorphisms in sepsis. Crit Care Clin. 2009;25(4):835–856. doi: 10.1016/j.ccc.2009.06.004. x. [DOI] [PubMed] [Google Scholar]
  • 14.Sutherland AM, Walley KR. Bench-to-bedside review: Association of genetic variation with sepsis. Crit Care. 2009;13(2):210. doi: 10.1186/cc7702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Yende S, Kammerer CM, Angus DC. Genetics and proteomics: deciphering gene association studies in critical illness. Crit Care. 2006;10(4):227. doi: 10.1186/cc5015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Freely associating. Nat Genet. 1999;22:1–2. doi: 10.1038/8702. [DOI] [PubMed] [Google Scholar]
  • 17.Hobson MJ, Wong HR. Genetic association research: understanding its challenges and limitations. Pediatr Crit Care Med. 2010;11(6):762–763. doi: 10.1097/PCC.0b013e3181d503e7. [DOI] [PubMed] [Google Scholar]
  • 18.Stalets E, Wong HR. Critically associating. Crit Care Med. 2009;37(4):1492–1493. doi: 10.1097/CCM.0b013e31819d2be0. [DOI] [PubMed] [Google Scholar]
  • 19.Clark MF, Baudouin SV. A systematic review of the quality of genetic association studies in human sepsis. Intensive Care Med. 2006;32(11):1706–1712. doi: 10.1007/s00134-006-0327-y. [DOI] [PubMed] [Google Scholar]
  • 20.Little J, Higgins JP, Ioannidis JP, Moher D, Gagnon F, von Elm E, Khoury MJ, Cohen B, Davey-Smith G, Grimshaw J, et al. STrengthening the REporting of Genetic Association Studies (STREGA) an extension of the STROBE statement. PLoS Med. 2009;6(2):e22. doi: 10.1371/journal.pmed.1000022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Aso Y. Plasminogen activator inhibitor (PAI)-1 in vascular inflammation and thrombosis. Front Biosci. 2007;12:2957–2966. doi: 10.2741/2285. [DOI] [PubMed] [Google Scholar]
  • 22.Pannekoek H, Veerman H, Lambers H, Diergaarde P, Verweij CL, van Zonneveld AJ, van Mourik JA. Endothelial plasminogen activator inhibitor (PAI) a new member of the Serpin gene family. EMBO J. 1986;5(10):2539–2544. doi: 10.1002/j.1460-2075.1986.tb04532.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Dawson SJ, Wiman B, Hamsten A, Green F, Humphries S, Henney AM. The two allele sequences of a common polymorphism in the promoter of the plasminogen activator inhibitor-1 (PAI-1) gene respond differently to interleukin-1 in HepG2 cells. J Biol Chem. 1993;268(15):10739–10745. [PubMed] [Google Scholar]
  • 24.Petaja J. Inflammation and coagulation. An overview. Thromb Res. 2011;127(Suppl 2):S34–S37. doi: 10.1016/S0049-3848(10)70153-5. [DOI] [PubMed] [Google Scholar]
  • 25.Powars DR, Rogers ZR, Patch MJ, McGehee WG, Francis RB., Jr Purpura fulminans in meningococcemia: association with acquired deficiencies of proteins C and S. N Engl J Med. 1987;317(9):571–572. doi: 10.1056/NEJM198708273170912. [DOI] [PubMed] [Google Scholar]
  • 26.Hazelzet JA, Risseeuw-Appel IM, Kornelisse RF, Hop WC, Dekker I, Joosten KF, de Groot R, Hack CE. Age-related differences in outcome and severity of DIC in children with septic shock and purpura. Thromb Haemost. 1996;76(6):932–938. [PubMed] [Google Scholar]
  • 27.Machado FR, Cesar MS. Sepsis, coagulation and anticoagulants. Endocr Metab Immune Disord Drug Targets. 2010;10(3):204–213. doi: 10.2174/187153010791936892. [DOI] [PubMed] [Google Scholar]
  • 28.Hermans PW, Hibberd ML, Booy R, Daramola O, Hazelzet JA, de Groot R, Levin M. 4G/5G promoter polymorphism in the plasminogen-activator-inhibitor-1 gene and outcome of meningococcal disease. Meningococcal Research Group. Lancet. 1999;354(9178):556–560. doi: 10.1016/s0140-6736(99)02220-5. [DOI] [PubMed] [Google Scholar]
  • 29.Geishofer G, Binder A, Muller M, Zohrer B, Resch B, Muller W, Faber J, Finn A, Endler G, Mannhalter C, et al. 4G/5G promoter polymorphism in the plasminogen-activator-inhibitor-1 gene in children with systemic meningococcaemia. Eur J Pediatr. 2005;164(8):486–490. doi: 10.1007/s00431-005-1673-4. [DOI] [PubMed] [Google Scholar]
  • 30.Haralambous E, Hibberd ML, Hermans PW, Ninis N, Nadel S, Levin M. Role of functional plasminogen-activator-inhibitor-1 4G/5G promoter polymorphism in susceptibility, severity, and outcome of meningococcal disease in Caucasian children. Crit Care Med. 2003;31(12):2788–2793. doi: 10.1097/01.CCM.0000100122.57249.5D. [DOI] [PubMed] [Google Scholar]
  • 31.Westendorp RG, Hottenga JJ, Slagboom PE. Variation in plasminogen-activator-inhibitor-1 gene and risk of meningococcal septic shock. Lancet. 1999;354(9178):561–563. doi: 10.1016/S0140-6736(98)09376-3. [DOI] [PubMed] [Google Scholar]
  • 32.Brouwer MC, Read RC, van de Beek D. Host genetics and outcome in meningococcal disease: a systematic review and meta-analysis. Lancet Infect Dis. 2010;10(4):262–274. doi: 10.1016/S1473-3099(10)70045-1. [DOI] [PubMed] [Google Scholar]
  • 33.Nadel S, Goldstein B, Williams MD, Dalton H, Peters M, Macias WL, Abd-Allah SA, Levy H, Angle R, Wang D, et al. Drotrecogin alfa (activated) in children with severe sepsis: a multicentre phase III randomised controlled trial. Lancet. 2007;369(9564):836–843. doi: 10.1016/S0140-6736(07)60411-5. [DOI] [PubMed] [Google Scholar]
  • 34.Casanova JL, Abel L, Quintana-Murci L. Human TLRs and IL-1Rs in host defense: natural insights from evolutionary, epidemiological, and clinical genetics. Annu Rev Immunol. 2011;29:447–491. doi: 10.1146/annurev-immunol-030409-101335. [DOI] [PubMed] [Google Scholar]
  • 35.Lorenz E, Mira JP, Cornish KL, Arbour NC, Schwartz DA. A novel polymorphism in the toll-like receptor 2 gene and its potential association with staphylococcal infection. Infect Immun. 2000;68(11):6398–6401. doi: 10.1128/iai.68.11.6398-6401.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Moore CE, Segal S, Berendt AR, Hill AV, Day NP. Lack of association between Toll-like receptor 2 polymorphisms and susceptibility to severe disease caused by Staphylococcus aureus. Clin Diagn Lab Immunol. 2004;11(6):1194–1197. doi: 10.1128/CDLI.11.6.1194-1197.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.von Aulock S, Schroder NW, Traub S, Gueinzius K, Lorenz E, Hartung T, Schumann RR, Hermann C. Heterozygous toll-like receptor 2 polymorphism does not affect lipoteichoic acid-induced chemokine and inflammatory responses. Infect Immun. 2004;72(3):1828–1831. doi: 10.1128/IAI.72.3.1828-1831.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kutukculer N, Yeniay BS, Aksu G, Berdeli A. Arg753Gln polymorphism of the human toll-like receptor-2 gene in children with recurrent febrile infections. Biochem Genet. 2007;45(7–8):507–514. doi: 10.1007/s10528-007-9091-0. [DOI] [PubMed] [Google Scholar]
  • 39.Tabel Y, Berdeli A, Mir S. Association of TLR2 gene Arg753Gln polymorphism with urinary tract infection in children. Int J Immunogenet. 2007;34(6):399–405. doi: 10.1111/j.1744-313X.2007.00709.x. [DOI] [PubMed] [Google Scholar]
  • 40.Krediet TG, Wiertsema SP, Vossers MJ, Hoeks SB, Fleer A, Ruven HJ, Rijkers GT. Toll-like receptor 2 polymorphism is associated with preterm birth. Pediatr Res. 2007;62(4):474–476. doi: 10.1203/PDR.0b013e31813c9401. [DOI] [PubMed] [Google Scholar]
  • 41.Berdeli A, Celik HA, Ozyurek R, Dogrusoz B, Aydin HH. TLR-2 gene Arg753Gln polymorphism is strongly associated with acute rheumatic fever in children. J Mol Med (Berl) 2005;83(7):535–541. doi: 10.1007/s00109-005-0677-x. [DOI] [PubMed] [Google Scholar]
  • 42.Abu-Maziad A, Schaa K, Bell EF, Dagle JM, Cooper M, Marazita ML, Murray JC. Role of polymorphic variants as genetic modulators of infection in neonatal sepsis. Pediatr Res. 2010;68(4):323–329. doi: 10.1203/PDR.0b013e3181e6a068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Sutherland AM, Walley KR, Russell JA. Polymorphisms in CD14, mannose-binding lectin, and Toll-like receptor-2 are associated with increased prevalence of infection in critically ill adults. Crit Care Med. 2005;33(3):638–644. doi: 10.1097/01.ccm.0000156242.44356.c5. [DOI] [PubMed] [Google Scholar]
  • 44.Beutler B, Poltorak A. The sole gateway to endotoxin response: how LPS was identified as Tlr4, and its role in innate immunity. Drug Metab Dispos. 2001;29(4 Pt 2):474–478. [PubMed] [Google Scholar]
  • 45.Qureshi ST, Lariviere L, Leveque G, Clermont S, Moore KJ, Gros P, Malo D. Endotoxin-tolerant mice have mutations in Toll-like receptor 4 (Tlr4) J Exp Med. 1999;189(4):615–625. doi: 10.1084/jem.189.4.615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Poltorak A, He X, Smirnova I, Liu MY, Van Huffel C, Du X, Birdwell D, Alejos E, Silva M, Galanos C, et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science. 1998;282(5396):2085–2088. doi: 10.1126/science.282.5396.2085. [DOI] [PubMed] [Google Scholar]
  • 47.Hoshino K, Takeuchi O, Kawai T, Sanjo H, Ogawa T, Takeda Y, Takeda K, Akira S. Cutting edge: Toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide: evidence for TLR4 as the Lps gene product. J Immunol. 1999;162(7):3749–3752. [PubMed] [Google Scholar]
  • 48.Arbour NC, Lorenz E, Schutte BC, Zabner J, Kline JN, Jones M, Frees K, Watt JL, Schwartz DA. TLR4 mutations are associated with endotoxin hyporesponsiveness in humans. Nat Genet. 2000;25(2):187–191. doi: 10.1038/76048. [DOI] [PubMed] [Google Scholar]
  • 49.Michel O, LeVan TD, Stern D, Dentener M, Thorn J, Gnat D, Beijer ML, Cochaux P, Holt PG, Martinez FD, et al. Systemic responsiveness to lipopolysaccharide and polymorphisms in the toll-like receptor 4 gene in human beings. J Allergy Clin Immunol. 2003;112(5):923–929. doi: 10.1016/j.jaci.2003.05.001. [DOI] [PubMed] [Google Scholar]
  • 50.Douville RN, Lissitsyn Y, Hirschfeld AF, Becker AB, Kozyrskyj AL, Liem J, Bastien N, Li Y, Victor RE, Sekhon M, et al. TLR4 Asp299Gly and Thr399Ile polymorphisms: no impact on human immune responsiveness to LPS or respiratory syncytial virus. PLoS One. 2010;5(8):e12087. doi: 10.1371/journal.pone.0012087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lorenz E, Mira JP, Frees KL, Schwartz DA. Relevance of mutations in the TLR4 receptor in patients with gram-negative septic shock. Arch Intern Med. 2002;162(9):1028–1032. doi: 10.1001/archinte.162.9.1028. [DOI] [PubMed] [Google Scholar]
  • 52.Agnese DM, Calvano JE, Hahm SJ, Coyle SM, Corbett SA, Calvano SE, Lowry SF. Human toll-like receptor 4 mutations but not CD14 polymorphisms are associated with an increased risk of gram-negative infections. J Infect Dis. 2002;186(10):1522–1525. doi: 10.1086/344893. [DOI] [PubMed] [Google Scholar]
  • 53.Faber J, Henninger N, Finn A, Zenz W, Zepp F, Knuf M. A toll-like receptor 4 variant is associated with fatal outcome in children with invasive meningococcal disease. Acta Paediatr. 2009;98(3):548–552. doi: 10.1111/j.1651-2227.2008.01163.x. [DOI] [PubMed] [Google Scholar]
  • 54.Read RC, Pullin J, Gregory S, Borrow R, Kaczmarski EB, di Giovine FS, Dower SK, Cannings C, Wilson AG. A functional polymorphism of toll-like receptor 4 is not associated with likelihood or severity of meningococcal disease. J Infect Dis. 2001;184(5):640–642. doi: 10.1086/322798. [DOI] [PubMed] [Google Scholar]
  • 55.Allen A, Obaro S, Bojang K, Awomoyi AA, Greenwood BM, Whittle H, Sirugo G, Newport MJ. Variation in Toll-like receptor 4 and susceptibility to group A meningococcal meningitis in Gambian children. Pediatr Infect Dis J. 2003;22(11):1018–1019. doi: 10.1097/01.inf.0000095431.15606.68. [DOI] [PubMed] [Google Scholar]
  • 56.Smirnova I, Mann N, Dols A, Derkx HH, Hibberd ML, Levin M, Beutler B. Assay of locus-specific genetic load implicates rare Toll-like receptor 4 mutations in meningococcal susceptibility. Proc Natl Acad Sci U S A. 2003;100(10):6075–6080. doi: 10.1073/pnas.1031605100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Mockenhaupt FP, Cramer JP, Hamann L, Stegemann MS, Eckert J, Oh NR, Otchwemah RN, Dietz E, Ehrhardt S, Schroder NW, et al. Toll-like receptor (TLR) polymorphisms in African children: common TLR-4 variants predispose to severe malaria. J Commun Dis. 2006;38(3):230–245. [PubMed] [Google Scholar]
  • 58.Khor CC, Chapman SJ, Vannberg FO, Dunne A, Murphy C, Ling EY, Frodsham AJ, Walley AJ, Kyrieleis O, Khan AA, et al. Mal functional variant is associated with protection against invasive pneumococcal disease, bacteremia, malaria and tuberculosis. Nat Genet. 2007;39(4):523–528. doi: 10.1038/ng1976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ferwerda B, Alonso S, Banahan K, McCall MB, Giamarellos-Bourboulis EJ, Ramakers BP, Mouktaroudi M, Fain PR, Izagirre N, et al. Syafruddin DFunctional and genetic evidence that the Mal/TIRAP allele variant 180L has been selected by providing protection against septic shock. Proc Natl Acad Sci U S A. 2009;106(25):10272–10277. doi: 10.1073/pnas.0811273106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kumpf O, Giamarellos-Bourboulis EJ, Koch A, Hamann L, Mouktaroudi M, Oh DY, Latz E, Lorenz E, Schwartz DA, Ferwerda B, et al. Influence of genetic variations in TLR4 and TIRAP/Mal on the course of sepsis and pneumonia and cytokine release: an observational study in three cohorts. Crit Care. 2010;14(3):R103. doi: 10.1186/cc9047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ladhani SN, Davila S, Hibberd ML, Heath PT, Ramsay ME, Slack MP, Pollard AJ, Booy R. Association between single-nucleotide polymorphisms in Mal/TIRAP and interleukin-10 genes and susceptibility to invasive haemophilus influenzae serotype b infection in immunized children. Clin Infect Dis. 2010;51(7):761–767. doi: 10.1086/656236. [DOI] [PubMed] [Google Scholar]
  • 62.Beutler B. TNF, immunity and inflammatory disease: lessons of the past decade. J Investig Med. 1995;43(3):227–235. [PubMed] [Google Scholar]
  • 63.Beutler B, Cerami A. Tumor necrosis, cachexia, shock, and inflammation: a common mediator. Annu Rev Biochem. 1988;57:505–518. doi: 10.1146/annurev.bi.57.070188.002445. [DOI] [PubMed] [Google Scholar]
  • 64.Lorente JA, Marshall JC. Neutralization of tumor necrosis factor in preclinical models of sepsis. Shock. 2005;24(Suppl 1):107–119. doi: 10.1097/01.shk.0000191343.21228.78. [DOI] [PubMed] [Google Scholar]
  • 65.Smith AJ, Humphries SE. Cytokine and cytokine receptor gene polymorphisms and their functionality. Cytokine Growth Factor Rev. 2009;20(1):43–59. doi: 10.1016/j.cytogfr.2008.11.006. [DOI] [PubMed] [Google Scholar]
  • 66.Wilson AG, de Vries N, Pociot F, di Giovine FS, van der Putte LB, Duff GW. An allelic polymorphism within the human tumor necrosis factor alpha promoter region is strongly associated with HLA A1, B8, and DR3 alleles. J Exp Med. 1993;177(2):557–560. doi: 10.1084/jem.177.2.557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Wilson AG, Symons JA, McDowell TL, McDevitt HO, Duff GW. Effects of a polymorphism in the human tumor necrosis factor alpha promoter on transcriptional activation. Proc Natl Acad Sci U S A. 1997;94(7):3195–3199. doi: 10.1073/pnas.94.7.3195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kroeger KM, Carville KS, Abraham LJ. The-308 tumor necrosis factor-alpha promoter polymorphism effects transcription. Mol Immunol. 1997;34(5):391–399. doi: 10.1016/s0161-5890(97)00052-7. [DOI] [PubMed] [Google Scholar]
  • 69.Louis E, Franchimont D, Piron A, Gevaert Y, Schaaf-Lafontaine N, Roland S, Mahieu P, Malaise M, De Groote D, Louis R, et al. Tumour necrosis factor (TNF) gene polymorphism influences TNF-alpha production in lipopolysaccharide (LPS)-stimulated whole blood cell culture in healthy humans. Clin Exp Immunol. 1998;113(3):401–406. doi: 10.1046/j.1365-2249.1998.00662.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Mira JP, Cariou A, Grall F, Delclaux C, Losser MR, Heshmati F, Cheval C, Monchi M, Teboul JL, Riche F, et al. Association of TNF2, a TNF-alpha promoter polymorphism, with septic shock susceptibility and mortality: a multicenter study. Jama. 1999;282(6):561–568. doi: 10.1001/jama.282.6.561. [DOI] [PubMed] [Google Scholar]
  • 71.Stuber F, Udalova IA, Book M, Drutskaya LN, Kuprash DV, Turetskaya RL, Schade FU, Nedospasov SA. −308 tumor necrosis factor (TNF) polymorphism is not associated with survival in severe sepsis and is unrelated to lipopolysaccharide inducibility of the human TNF promoter. J Inflamm. 1995;46(1):42–50. [PubMed] [Google Scholar]
  • 72.Read RC, Teare DM, Pridmore AC, Naylor SC, Timms JM, Kaczmarski EB, Borrow R, Wilson AG. The tumor necrosis factor polymorphism TNF (-308) is associated with susceptibility to meningococcal sepsis, but not with lethality. Crit Care Med. 2009;37(4):1237–1243. doi: 10.1097/CCM.0b013e31819c39bc. [DOI] [PubMed] [Google Scholar]
  • 73.Teuffel O, Ethier MC, Beyene J, Sung L. Association between tumor necrosis factor-alpha promoter −308 A/G polymorphism and susceptibility to sepsis and sepsis mortality: a systematic review and meta-analysis. Crit Care Med. 2010;38(1):276–282. doi: 10.1097/CCM.0b013e3181b42af0. [DOI] [PubMed] [Google Scholar]
  • 74.Nadel S, Newport MJ, Booy R, Levin M. Variation in the tumor necrosis factor-alpha gene promoter region may be associated with death from meningococcal disease. J Infect Dis. 1996;174(4):878–880. doi: 10.1093/infdis/174.4.878. [DOI] [PubMed] [Google Scholar]
  • 75.Sipahi T, Pocan H, Akar N. Effect of various genetic polymorphisms on the incidence and outcome of severe sepsis. Clin Appl Thromb Hemost. 2006;12(1):47–54. doi: 10.1177/107602960601200108. [DOI] [PubMed] [Google Scholar]
  • 76.Stuber F, Petersen M, Bokelmann F, Schade U. A genomic polymorphism within the tumor necrosis factor locus influences plasma tumor necrosis factor-alpha concentrations and outcome of patients with severe sepsis. Crit Care Med. 1996;24(3):381–384. doi: 10.1097/00003246-199603000-00004. [DOI] [PubMed] [Google Scholar]
  • 77.McArthur JA, Zhang Q, Quasney MW. Association between the A/A genotype at the lymphotoxin-alpha+250 site and increased mortality in children with positive blood cultures. Pediatr Crit Care Med. 2002;3(4):341–344. doi: 10.1097/00130478-200210000-00002. [DOI] [PubMed] [Google Scholar]
  • 78.Christie JD. Microarrays. Crit Care Med. 2005;33(12 Suppl):S449–S452. doi: 10.1097/01.ccm.0000186078.26361.96. [DOI] [PubMed] [Google Scholar]
  • 79.Gershon D. Microarray technology: an array of opportunities. Nature. 2002;416(6883):885–891. doi: 10.1038/416885a. [DOI] [PubMed] [Google Scholar]
  • 80.Calvano SE, Xiao W, Richards DR, Felciano RM, Baker HV, Cho RJ, Chen RO, Brownstein BH, Cobb JP, Tschoeke SK, et al. A network-based analysis of systemic inflammation in humans. Nature. 2005;437(7061):1032–1037. doi: 10.1038/nature03985. [DOI] [PubMed] [Google Scholar]
  • 81.Johnson SB, Lissauer M, Bochicchio GV, Moore R, Cross AS, Scalea TM. Gene expression profiles differentiate between sterile SIRS and early sepsis. Ann Surg. 2007;245(4):611–621. doi: 10.1097/01.sla.0000251619.10648.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.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(1):63–71. doi: 10.1016/j.imlet.2006.04.010. [DOI] [PubMed] [Google Scholar]
  • 83.Payen D, Lukaszewicz AC, Belikova I, Faivre V, Gelin C, Russwurm S, Launay JM, Sevenet N. Gene profiling in human blood leucocytes during recovery from septic shock. Intensive Care Med. 2008;34(8):1371–1376. doi: 10.1007/s00134-008-1048-1. [DOI] [PubMed] [Google Scholar]
  • 84.Prabhakar U, Conway TM, Murdock P, Mooney JL, Clark S, Hedge P, Bond BC, Jazwinska EC, Barnes MR, Tobin F, et al. Correlation of protein and gene expression profiles of inflammatory proteins after endotoxin challenge in human subjects. DNA Cell Biol. 2005;24(7):410–431. doi: 10.1089/dna.2005.24.410. [DOI] [PubMed] [Google Scholar]
  • 85.Prucha M, Ruryk A, Boriss H, Moller E, Zazula R, Herold I, Claus RA, Reinhart KA, Deigner P, Russwurm S. Expression profiling: toward an application in sepsis diagnostics. Shock. 2004;22(1):29–33. doi: 10.1097/01.shk.0000129199.30965.02. [DOI] [PubMed] [Google Scholar]
  • 86.Suffredini AF, Fromm RE, Parker MM, Brenner M, Kovacs JA, Wesley RA, Parrillo JE. The cardiovascular response of normal humans to the administration of endotoxin. N Engl J Med. 1989;321(5):280–287. doi: 10.1056/NEJM198908033210503. [DOI] [PubMed] [Google Scholar]
  • 87.Talwar S, Munson PJ, Barb J, Fiuza C, Cintron AP, Logun C, Tropea M, Khan S, Reda D, Shelhamer JH, et al. Gene expression profiles of peripheral blood leukocytes after endotoxin challenge in humans. Physiol Genomics. 2006;25(2):203–215. doi: 10.1152/physiolgenomics.00192.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Tang BM, Huang SJ, McLean AS. Genome-wide transcription profiling of human sepsis: a systematic review. Crit Care. 2010;14(6):R237. doi: 10.1186/cc9392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Tang BM, McLean AS, Dawes IW, Huang SJ, Cowley MJ, Lin RC. Gene-expression profiling of gram-positive and gram-negative sepsis in critically ill patients. Crit Care Med. 2008;36(4):1125–1128. doi: 10.1097/CCM.0b013e3181692c0b. [DOI] [PubMed] [Google Scholar]
  • 90.Tang BM, McLean AS, Dawes IW, Huang SJ, Lin RC. The use of gene-expression profiling to identify candidate genes in human sepsis. Am J Respir Crit Care Med. 2007;176(7):676–684. doi: 10.1164/rccm.200612-1819OC. [DOI] [PubMed] [Google Scholar]
  • 91.Tang BM, McLean AS, Dawes IW, Huang SJ, Lin RC. Gene-expression profiling of peripheral blood mononuclear cells in sepsis. Crit Care Med. 2009;37(3):882–888. doi: 10.1097/CCM.0b013e31819b52fd. [DOI] [PubMed] [Google Scholar]
  • 92.Pathan N, Hemingway CA, Alizadeh AA, Stephens AC, Boldrick JC, Oragui EE, McCabe C, Welch SB, Whitney A, O'Gara P, et al. Role of interleukin 6 in myocardial dysfunction of meningococcal septic shock. Lancet. 2004;363(9404):203–209. doi: 10.1016/S0140-6736(03)15326-3. [DOI] [PubMed] [Google Scholar]
  • 93.Cvijanovich N, Shanley TP, Lin R, Allen GL, Thomas NJ, Checchia P, Anas N, Freishtat RJ, Monaco M, Odoms K, et al. Validating the genomic signature of pediatric septic shock. Physiol Genomics. 2008;34(1):127–134. doi: 10.1152/physiolgenomics.00025.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Shanley TP, Cvijanovich N, Lin R, Allen GL, Thomas NJ, Doctor A, Kalyanaraman M, Tofil NM, Penfil S, Monaco M, et al. Genome-level longitudinal expression of signaling pathways and gene networks in pediatric septic shock. Mol Med. 2007;13(9–10):495–508. doi: 10.2119/2007-00065.Shanley. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Wong HR, Cvijanovich N, Allen GL, Lin R, Anas N, Meyer K, Freishtat RJ, Monaco M, Odoms K, Sakthivel B, et al. Genomic expression profiling across the pediatric systemic inflammatory response syndrome, sepsis, and septic shock spectrum. Crit Care Med. 2009;37(5):1558–1566. doi: 10.1097/CCM.0b013e31819fcc08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Wong HR, Cvijanovich N, Lin R, Allen GL, Thomas NJ, Willson DF, Freishtat RJ, Anas N, Meyer K, Checchia PA, et al. Identification of pediatric septic shock subclasses based on genome-wide expression profiling. BMC Med. 2009;7:34. doi: 10.1186/1741-7015-7-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Wong HR, Shanley TP, Sakthivel B, Cvijanovich N, Lin R, Allen GL, Thomas NJ, Doctor A, Kalyanaraman M, Tofil NM, et al. Genome-level expression profiles in pediatric septic shock indicate a role for altered zinc homeostasis in poor outcome. Physiol Genomics. 2007;30(2):146–155. doi: 10.1152/physiolgenomics.00024.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Rink L, Haase H. Zinc homeostasis and immunity. Trends Immunol. 2007;28(1):1–4. doi: 10.1016/j.it.2006.11.005. [DOI] [PubMed] [Google Scholar]
  • 99.Cvijanovich NZ, King JC, Flori HR, Gildengorin G, Wong HR. Zinc homeostasis in pediatric critical illness. Pediatr Crit Care Med. 2009;10(1):29–34. doi: 10.1097/PCC.0b013e31819371ce. [DOI] [PubMed] [Google Scholar]
  • 100.Heyland DK, Jones N, Cvijanovich NZ, Wong H. Zinc supplementation in critically ill patients: a key pharmaconutrient? JPEN J Parenter Enteral Nutr. 2008;32(5):509–519. doi: 10.1177/0148607108322402. [DOI] [PubMed] [Google Scholar]
  • 101.Weitzel LR, Mayles WJ, Sandoval PA, Wischmeyer PE. Effects of pharmaconutrients on cellular dysfunction and the microcirculation in critical illness. Curr Opin Anaesthesiol. 2009;22(2):177–183. doi: 10.1097/ACO.0b013e328328d32f. [DOI] [PubMed] [Google Scholar]
  • 102.Bao S, Liu MJ, Lee B, Besecker B, Lai JP, Guttridge DC, Knoell DL. Zinc modulates the innate immune response in vivo to polymicrobial sepsis through regulation of NF-kappaB. Am J Physiol Lung Cell Mol Physiol. 2010;298(6):L744–L754. doi: 10.1152/ajplung.00368.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Knoell DL, Julian MW, Bao S, Besecker B, Macre JE, Leikauf GD, DiSilvestro RA, Crouser ED. Zinc deficiency increases organ damage and mortality in a murine model of polymicrobial sepsis. Crit Care Med. 2009;37(4):1380–1388. doi: 10.1097/CCM.0b013e31819cefe4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Besecker BY, Exline MC, Hollyfield J, Phillips G, Disilvestro RA, Wewers MD, Knoell DL. A comparison of zinc metabolism, inflammation, and disease severity in critically ill infected and noninfected adults early after intensive care unit admission. Am J Clin Nutr. 2011;93(6):1356–1364. doi: 10.3945/ajcn.110.008417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Knoell DL, Liu MJ. Impact of zinc metabolism on innate immune function in the setting of sepsis. Int J Vitam Nutr Res. 2010;80(4–5):271–277. doi: 10.1024/0300-9831/a000034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Carcillo J, Holubkov R, Dean JM, Berger J, Meert KL, Anand KJ, Zimmerman J, Newth CJ, Harrison R, Willson DF, et al. Rationale and design of the pediatric critical illness stress-induced immune suppression (CRISIS) prevention trial. JPEN J Parenter Enteral Nutr. 2009;33(4):368–374. doi: 10.1177/0148607108327392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Huang DT, Ochoa JB. Nutrition trials in critical illness: bigger, faster, stronger. JPEN J Parenter Enteral Nutr. 2010;34(6):608–609. doi: 10.1177/0148607110375700. [DOI] [PubMed] [Google Scholar]
  • 108.Neu J. Use of nutrition to prevent stress-induced immunosuppression in the pediatric intensive care unit: a clinical trials minefield. JPEN J Parenter Enteral Nutr. 2009;33(4):440–441. doi: 10.1177/0148607108328521. [DOI] [PubMed] [Google Scholar]
  • 109.Wong HR, Freishtat RJ, Monaco M, Odoms K, Shanley TP. Leukocyte subset-derived genomewide expression profiles in pediatric septic shock. Pediatr Crit Care Med. 2010;11(3):349–355. doi: 10.1097/PCC.0b013e3181c519b4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Wynn JL, Cvijanovich NZ, Allen GL, Thomas NJ, Freishtat RJ, Anas N, Meyer K, Checchia PA, Lin R, Shanley TP, et al. The Influence of Developmental Age on the Early Transcriptomic Response of Children with Septic Shock. Mol Med. 2011 doi: 10.2119/molmed.2011.00169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Solan PD, Dunsmore KE, Denenberg AG, Odom K, Zingarelli B, Wong HR. A novel role for matrix metalloproteinase-8 in sepsis. Crit Care Med. 2011 doi: 10.1097/CCM.0b013e318232e404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Van Lint P, Libert C. Matrix metalloproteinase-8: cleavage can be decisive. Cytokine Growth Factor Rev. 2006;17(4):217–223. doi: 10.1016/j.cytogfr.2006.04.001. [DOI] [PubMed] [Google Scholar]
  • 113.Vanlaere I, Libert C. Matrix metalloproteinases as drug targets in infections caused by gram-negative bacteria and in septic shock. Clin Microbiol Rev. 2009;22(2):224–239. doi: 10.1128/CMR.00047-08. Table of Contents. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Bouchon A, Facchetti F, Weigand MA, Colonna M. TREM-1 amplifies inflammation and is a crucial mediator of septic shock. Nature. 2001;410(6832):1103–1107. doi: 10.1038/35074114. [DOI] [PubMed] [Google Scholar]
  • 115.Galetto-Lacour A, Gervaix A. Identifying severe bacterial infection in children with fever without source. Expert Rev Anti Infect Ther. 2010;8(11):1231–1237. doi: 10.1586/eri.10.118. [DOI] [PubMed] [Google Scholar]
  • 116.Ramilo O, Allman W, Chung W, Mejias A, Ardura M, Glaser C, Wittkowski KM, Piqueras B, Banchereau J, Palucka AK, et al. Gene expression patterns in blood leukocytes discriminate patients with acute infections. Blood. 2007;109(5):2066–2077. doi: 10.1182/blood-2006-02-002477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Allantaz F, Chaussabel D, Stichweh D, Bennett L, Allman W, Mejias A, Ardura M, Chung W, Smith E, Wise C, et al. Blood leukocyte microarrays to diagnose systemic onset juvenile idiopathic arthritis and follow the response to IL-1 blockade. J Exp Med. 2007;204(9):2131–2144. doi: 10.1084/jem.20070070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kaplan JM, Wong HR. Biomarker discovery and development in pediatric critical care medicine. Pediatr Crit Care Med. 2011;12(2):165–173. doi: 10.1097/PCC.0b013e3181e28876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Marshall JC. Sepsis: rethinking the approach to clinical research. J Leukoc Biol. 2008;83(3):471–482. doi: 10.1189/jlb.0607380. [DOI] [PubMed] [Google Scholar]
  • 120.Marshall JC, Vincent JL, Fink MP, Cook DJ, Rubenfeld G, Foster D, Fisher CJ, Jr, Faist E, Reinhart K. Measures, markers, and mediators: toward a staging system for clinical sepsis. A report of the Fifth Toronto Sepsis Roundtable, Toronto, Ontario, Canada, October 25–26, 2000. Crit Care Med. 2003;31(5):1560–1567. doi: 10.1097/01.CCM.0000065186.67848.3A. [DOI] [PubMed] [Google Scholar]
  • 121.Wong HR, Cvijanovich N, Wheeler DS, Bigham MT, Monaco M, Odoms K, Macias WL, Williams MD. Interleukin-8 as a stratification tool for interventional trials involving pediatric septic shock. Am J Respir Crit Care Med. 2008;178(3):276–282. doi: 10.1164/rccm.200801-131OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Calfee CS, Thompson BT, Parsons PE, Ware LB, Matthay MA, Wong HR. Plasma interleukin-8 is not an effective risk stratification tool for adults with vasopressor-dependent septic shock. Crit Care Med. 2010;38(6):1436–1441. doi: 10.1097/CCM.0b013e3181de42ad. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Nowak JE, Wheeler DS, Harmon KK, Wong HR. Admission chemokine (C-C motif) ligand 4 levels predict survival in pediatric septic shock. Pediatr Crit Care Med. 2010;11(2):213–216. doi: 10.1097/PCC.0b013e3181b8076c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Standage SW, Wong HR. Biomarkers for pediatric sepsis and septic shock. Expert Rev Anti Infect Ther. 2011;9(1):71–79. doi: 10.1586/eri.10.154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Wong HR, Wheeler DS, Tegtmeyer K, Poynter SE, Kaplan JM, Chima RS, Stalets E, Basu RK, Doughty LA. Toward a clinically feasible gene expression-based subclassification strategy for septic shock: proof of concept. Crit Care Med. 2010;38(10):1955–1961. doi: 10.1097/CCM.0b013e3181eb924f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Eichler GS, Huang S, Ingber DE. Gene Expression Dynamics Inspector (GEDI) for integrative analysis of expression profiles. Bioinformatics. 2003;19(17):2321–2322. doi: 10.1093/bioinformatics/btg307. [DOI] [PubMed] [Google Scholar]
  • 127.Guo Y, Eichler GS, Feng Y, Ingber DE, Huang S. Towards a holistic, yet gene-centered analysis of gene expression profiles: a case study of human lung cancers. J Biomed Biotechnol. 2006;2006(5):69141. doi: 10.1155/JBB/2006/69141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Wong HR, Cvijanovich NZ, Allen GL, Thomas NJ, Freishtat RJ, Anas N, Meyer K, Checchia PA, Lin R, Shanley TP, et al. Validation of a gene expression-based subclassification strategy for pediatric septic shock. Crit Care Med. 2011 doi: 10.1097/CCM.0b013e3182257675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Delcuve GP, Rastegar M, Davie JR. Epigenetic control. J Cell Physiol. 2009;219(2):243–250. doi: 10.1002/jcp.21678. [DOI] [PubMed] [Google Scholar]
  • 130.Carson WF, Cavassani KA, Dou Y, Kunkel SL. Epigenetic regulation of immune cell functions during post-septic immunosuppression. Epigenetics. 2011;6(3):273–283. doi: 10.4161/epi.6.3.14017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Chan C, Li L, McCall CE, Yoza BK. Endotoxin tolerance disrupts chromatin remodeling and NF-kappaB transactivation at the IL-1beta promoter. J Immunol. 2005;175(1):461–468. doi: 10.4049/jimmunol.175.1.461. [DOI] [PubMed] [Google Scholar]
  • 132.El Gazzar M, Yoza BK, Chen X, Garcia BA, Young NL, McCall CE. Chromatin-specific remodeling by HMGB1 and linker histone H1 silences proinflammatory genes during endotoxin tolerance. Mol Cell Biol. 2009;29(7):1959–1971. doi: 10.1128/MCB.01862-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.El Gazzar M, Yoza BK, Chen X, Hu J, Hawkins GA, McCall CE. G9a and HP1 couple histone and DNA methylation to TNFalpha transcription silencing during endotoxin tolerance. J Biol Chem. 2008;283(47):32198–32208. doi: 10.1074/jbc.M803446200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Foster SL, Hargreaves DC, Medzhitov R. Gene-specific control of inflammation by TLR-induced chromatin modifications. Nature. 2007;447(7147):972–978. doi: 10.1038/nature05836. [DOI] [PubMed] [Google Scholar]
  • 135.Brogdon JL, Xu Y, Szabo SJ, An S, Buxton F, Cohen D, Huang Q. Histone deacetylase activities are required for innate immune cell control of Th1 but not Th2 effector cell function. Blood. 2007;109(3):1123–1130. doi: 10.1182/blood-2006-04-019711. [DOI] [PubMed] [Google Scholar]
  • 136.Tsaprouni LG, Ito K, Adcock IM, Punchard N. Suppression of lipopolysaccharide- and tumour necrosis factor-alpha-induced interleukin (IL)-8 expression by glucocorticoids involves changes in IL-8 promoter acetylation. Clin Exp Immunol. 2007;150(1):151–157. doi: 10.1111/j.1365-2249.2007.03484.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.McCall CE, Yoza B, Liu T, El Gazzar M. Gene-specific epigenetic regulation in serious infections with systemic inflammation. J Innate Immun. 2010;2(5):395–405. doi: 10.1159/000314077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Ishii M, Wen H, Corsa CA, Liu T, Coelho AL, Allen RM, Carson WFt, Cavassani KA, Li X, Lukacs NW, et al. Epigenetic regulation of the alternatively activated macrophage phenotype. Blood. 2009;114(15):3244–3254. doi: 10.1182/blood-2009-04-217620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Wen H, Dou Y, Hogaboam CM, Kunkel SL. Epigenetic regulation of dendritic cell-derived interleukin-12 facilitates immunosuppression after a severe innate immune response. Blood. 2008;111(4):1797–1804. doi: 10.1182/blood-2007-08-106443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Wen H, Schaller MA, Dou Y, Hogaboam CM, Kunkel SL. Dendritic cells at the interface of innate and acquired immunity: the role for epigenetic changes. J Leukoc Biol. 2008;83(3):439–446. doi: 10.1189/jlb.0607357. [DOI] [PubMed] [Google Scholar]
  • 141.Quartin AA, Schein RM, Kett DH, Peduzzi PN. Magnitude and duration of the effect of sepsis on survival. Department of Veterans Affairs Systemic Sepsis Cooperative Studies Group. Jama. 1997;277(13):1058–1063. [PubMed] [Google Scholar]
  • 142.Winters BD, Eberlein M, Leung J, Needham DM, Pronovost PJ, Sevransky JE. Long-term mortality and quality of life in sepsis: a systematic review. Crit Care Med. 2010;38(5):1276–1283. doi: 10.1097/CCM.0b013e3181d8cc1d. [DOI] [PubMed] [Google Scholar]
  • 143.Read RC, Cannings C, Naylor SC, Timms JM, Maheswaran R, Borrow R, Kaczmarski EB, Duff GW. Variation within genes encoding interleukin-1 and the interleukin-1 receptor antagonist influence the severity of meningococcal disease. Ann Intern Med. 2003;138(7):534–541. doi: 10.7326/0003-4819-138-7-200304010-00009. [DOI] [PubMed] [Google Scholar]
  • 144.Endler G, Marculescu R, Starkl P, Binder A, Geishofer G, Muller M, Zohrer B, Resch B, Zenz W, Mannhalter C. Polymorphisms in the interleukin-1 gene cluster in children and young adults with systemic meningococcemia. Clin Chem. 2006;52(3):511–514. doi: 10.1373/clinchem.2005.058537. [DOI] [PubMed] [Google Scholar]
  • 145.Michalek J, Svetlikova P, Fedora M, Klimovic M, Klapacova L, Bartosova D, Hrstkova H, Hubacek JA. Interleukin-6 gene variants and the risk of sepsis development in children. Hum Immunol. 2007;68(9):756–760. doi: 10.1016/j.humimm.2007.06.003. [DOI] [PubMed] [Google Scholar]
  • 146.Lehrnbecher T, Bernig T, Hanisch M, Koehl U, Behl M, Reinhardt D, Creutzig U, Klingebiel T, Chanock SJ, Schwabe D. Common genetic variants in the interleukin-6 and chitotriosidase genes are associated with the risk for serious infection in children undergoing therapy for acute myeloid leukemia. Leukemia. 2005;19(10):1745–1750. doi: 10.1038/sj.leu.2403922. [DOI] [PubMed] [Google Scholar]
  • 147.Artifoni L, Negrisolo S, Montini G, Zucchetta P, Molinari PP, Cassar W, Destro R, Anglani F, Rigamonti W, Zacchello G, et al. Interleukin-8 and CXCR1 receptor functional polymorphisms and susceptibility to acute pyelonephritis. J Urol. 2007;177(3):1102–1106. doi: 10.1016/j.juro.2006.10.037. [DOI] [PubMed] [Google Scholar]
  • 148.Binder A, Endler G, Rieger S, Geishofer G, Resch B, Mannhalter C, Zenz W. Protein C promoter polymorphisms associate with sepsis in children with systemic meningococcemia. Hum Genet. 2007;122(2):183–190. doi: 10.1007/s00439-007-0392-5. [DOI] [PubMed] [Google Scholar]
  • 149.Bredius RG, Derkx BH, Fijen CA, de Wit TP, de Haas M, Weening RS, van de Winkel JG, Out TA. Fc gamma receptor IIa (CD32) polymorphism in fulminant meningococcal septic shock in children. J Infect Dis. 1994;170(4):848–853. doi: 10.1093/infdis/170.4.848. [DOI] [PubMed] [Google Scholar]
  • 150.van Sorge NM, van der Pol WL, van de Winkel JG. FcgammaR polymorphisms: Implications for function, disease susceptibility and immunotherapy. Tissue Antigens. 2003;61(3):189–202. doi: 10.1034/j.1399-0039.2003.00037.x. [DOI] [PubMed] [Google Scholar]
  • 151.Domingo P, Muniz-Diaz E, Baraldes MA, Arilla M, Barquet N, Pericas R, Juarez C, Madoz P, Vazquez G. Associations between Fc gamma receptor IIA polymorphisms and the risk and prognosis of meningococcal disease. Am J Med. 2002;112(1):19–25. doi: 10.1016/s0002-9343(01)01047-6. [DOI] [PubMed] [Google Scholar]
  • 152.Platonov AE, Shipulin GA, Vershinina IV, Dankert J, van de Winkel JG, Kuijper EJ. Association of human Fc gamma RIIa (CD32) polymorphism with susceptibility to and severity of meningococcal disease. Clin Infect Dis. 1998;27(4):746–750. doi: 10.1086/514935. [DOI] [PubMed] [Google Scholar]
  • 153.Platonov AE, Kuijper EJ, Vershinina IV, Shipulin GA, Westerdaal N, Fijen CA, van de Winkel JG. Meningococcal disease and polymorphism of FcgammaRIIa (CD32) in late complement component-deficient individuals. Clin Exp Immunol. 1998;111(1):97–101. doi: 10.1046/j.1365-2249.1998.00484.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Hibberd ML, Sumiya M, Summerfield JA, Booy R, Levin M. Association of variants of the gene for mannose-binding lectin with susceptibility to meningococcal disease. Meningococcal Research Group. Lancet. 1999;353(9158):1049–1053. doi: 10.1016/s0140-6736(98)08350-0. [DOI] [PubMed] [Google Scholar]
  • 155.Summerfield JA, Sumiya M, Levin M, Turner MW. Association of mutations in mannose binding protein gene with childhood infection in consecutive hospital series. BMJ. 1997;314(7089):1229–1232. doi: 10.1136/bmj.314.7089.1229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Koch A, Melbye M, Sorensen P, Homoe P, Madsen HO, Molbak K, Hansen CH, Andersen LH, Hahn GW, Garred P. Acute respiratory tract infections and mannose-binding lectin insufficiency during early childhood. JAMA. 2001;285(10):1316–1321. doi: 10.1001/jama.285.10.1316. [DOI] [PubMed] [Google Scholar]
  • 157.Michalek J, Svetlikova P, Fedora M, Klimovic M, Klapacova L, Bartosova D, Elbl L, Hrstkova H, Hubacek JA. Bactericidal permeability increasing protein gene variants in children with sepsis. Intensive Care Med. 2007;33(12):2158–2164. doi: 10.1007/s00134-007-0860-3. [DOI] [PubMed] [Google Scholar]
  • 158.El Saleeby CM, Li R, Somes GW, Dahmer MK, Quasney MW, DeVincenzo JP. Surfactant protein A2 polymorphisms and disease severity in a respiratory syncytial virus-infected population. J Pediatr. 2010;156(3):409–414. doi: 10.1016/j.jpeds.2009.09.043. [DOI] [PubMed] [Google Scholar]
  • 159.Dahmer MK, O'Cain P, Patwari PP, Simpson P, Li SH, Halligan N, Quasney MW. The influence of genetic variation in surfactant protein B on severe lung injury in African American children. Crit Care Med. 2011;39(5):1138–1144. doi: 10.1097/CCM.0b013e31820a9416. [DOI] [PubMed] [Google Scholar]
  • 160.Agbeko RS, Fidler KJ, Allen ML, Wilson P, Klein NJ, Peters MJ. Genetic variability in complement activation modulates the systemic inflammatory response syndrome in children. Pediatr Crit Care Med. 2010;11(5):561–567. doi: 10.1097/PCC.0b013e3181d900ba. [DOI] [PubMed] [Google Scholar]
  • 161.Haralambous E, Dolly SO, Hibberd ML, Litt DJ, Udalova IA, O'Dwyer C, Langford PR, Simon Kroll J, Levin M. Factor H, a regulator of complement activity, is a major determinant of meningococcal disease susceptibility in UK Caucasian patients. Scand J Infect Dis. 2006;38(9):764–771. doi: 10.1080/00365540600643203. [DOI] [PubMed] [Google Scholar]
  • 162.Davila S, Wright VJ, Khor CC, Sim KS, Binder A, Breunis WB, Inwald D, Nadel S, Betts H, Carrol ED, et al. Genome-wide association study identifies variants in the CFH region associated with host susceptibility to meningococcal disease. Nat Genet. 2010;42(9):772–776. doi: 10.1038/ng.640. [DOI] [PubMed] [Google Scholar]
  • 163.Harding D, Baines PB, Brull D, Vassiliou V, Ellis I, Hart A, Thomson AP, Humphries SE, Montgomery HE. Severity of meningococcal disease in children and the angiotensin-converting enzyme insertion/deletion polymorphism. Am J Respir Crit Care Med. 2002;165(8):1103–1106. doi: 10.1164/ajrccm.165.8.2108089. [DOI] [PubMed] [Google Scholar]
  • 164.Cogulu O, Onay H, Uzunkaya D, Gunduz C, Pehlivan S, Vardar F, Atlihan F, Ozkinay C, Ozkinay F. Role of angiotensin-converting enzyme gene polymorphisms in children with sepsis and septic shock. Pediatr Int. 2008;50(4):477–480. doi: 10.1111/j.1442-200X.2008.02583.x. [DOI] [PubMed] [Google Scholar]
  • 165.Tekin D, Dalgic N, Kayaalti Z, Soylemezoglu T, Diler B, Isin Kutlubay B. Importance of NOD2/CARD15 gene variants for susceptibility to and outcome of sepsis in Turkish children. Pediatr Crit Care Med. 2011 doi: 10.1097/PCC.0b013e3182191c2e. [DOI] [PubMed] [Google Scholar]
  • 166.Khor CC, Vannberg FO, Chapman SJ, Guo H, Wong SH, Walley AJ, Vukcevic D, Rautanen A, Mills TC, Chang KC, et al. CISH and susceptibility to infectious diseases. N Engl J Med. 2010;362(22):2092–2101. doi: 10.1056/NEJMoa0905606. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES