Open Access

Identification of potential biomarkers of sepsis using bioinformatics analysis

  • Authors:
    • Yu-Xia Yang
    • Li Li
  • View Affiliations

  • Published online on: March 2, 2017     https://doi.org/10.3892/etm.2017.4178
  • Pages: 1689-1696
  • Copyright: © Yang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Sepsis is defined as the systemic inflammatory response to infection and is one of the leading causes of mortality in critically ill patients. The goal of the present study is to elucidate the molecular mechanism of sepsis. Transcription profile data (GSE12624) were downloaded that had a total of 70 samples (36 sepsis samples and 34 non‑sepsis samples) from the Gene Expression Omnibus database. Protein‑protein interaction network analysis was conducted in order to comprehensively understand the interactions of genes in all samples. Hierarchical clustering and analysis of covariance (ANCOVA) global test were performed to identify the differentially expressed clusters in the networks, followed by function and pathway enrichment analyses. Finally, a support vector machine (SVM) was performed to classify the clusters, and 10‑fold cross‑validation method was performed to evaluate the classification results. A total of 7,672 genes were obtained after preprocessing of the mRNA expression profile data. The PPI network of genes under sepsis and non-sepsis status collected 1,996/2,147 genes and 2,645/2,783 interactions. Moreover, following the ANCOVA global test (P<0.05), 24 differentially expressed clusters with 12 clusters in septic and 12 clusters in non‑septic samples were identified. Finally, 207 biomarker genes, including CDC42, CSF3R, GCA, HMGB2, RHOG, SERPINB1, TYROBP SERPINA1, FCER1 G and S100P in the top six clusters, were collected using the SVM method. The SERPINA1, FCER1 G and S100P genes are thought to be potential biomarkers. Furthermore, Gene oncology terms, including the intracellular signaling cascade, regulation of programmed cell death, regulation of cell death, regulation of apoptosis and leukocyte activation may participate in sepsis.

Introduction

Sepsis is defined as the systemic inflammatory response to infection and is one of the leading causes of mortality in critically ill patients despite the application of numerous antibiotics and resuscitation therapies (1). Sepsis syndromes could be classified as systemic inflammatory response syndromes (SIRSs), sepsis, severe sepsis and septic shock according to the American College of Chest Physicians and the Society of Critical Care first published Care (2). In addition, the incidence of sepsis is increasing worldwide. In the USA the current incidence of sepsis is ~3 in 1,000 people, whereas severe sepsis leads to at least 200,000 deaths per year (3). Furthermore, severe sepsis and septic shock account for 30–50% of hospital-reported mortality (4). Recently, consensus on the treatment of sepsis includes advanced supportive care in the intensive care unit and use of bundle therapies (5). However, due to the non-specific nature of the signs and symptoms of sepsis, the diagnosis and treatment of sepsis are complicated.

Recently, numerous studies have been performed to identify the pathogenesis of sepsis (68). A number of biomarkers can be used in the diagnosis of sepsis, however, none of them has sufficient specificity or sensitivity in the clinical setting (911). C-reactive protein (CRP) and procalcitonin (PCT) have been widely used because of their relatively better specificity and prognostic capability (1214). The concentration of CRP is <0.8 mg/l and can increase 1,000-fold in response to an acute-phase stimulus (15). CRP is a protein that is synthesized in the liver and rises in response to inflammation (16). Moreover, it may help macrophages remove microorganisms by binding the phospholipid components (17). Nowadays, CRP is also treated as a biomarker for evaluating sepsis severity and prognosis or to monitor treatment response (12). PCT, which is produced by parafollicular cells of the thyroid and neuroendocrine cells of the lungs and the intestines is a 116 amino acid polypeptide precursor of the hormone calcitonin. PCT was first linked to infectious disease by Assicot et al (18) and was formally proposed as an adjunctive diagnostic biomarker in 2008 (19). It is maintained at a low level in healthy people and increases 1,000-fold during active infection (20). Furthermore, there are also several meta-analyses demonstrating that PCT could be used as a diagnostic marker in sepsis (21,22).

Nevertheless, there is currently no gold biomarker that exists as a marker of sepsis. Thus, identification of a new biomarker is urgently required. In order to further identify the molecular pathogenesis of sepsis, microarray data were firstly downloaded, then the raw data were analyzed to construct a protein-protein interaction (PPI) network. Subsequently, differentially expressed clusters in the PPI network were identified and significantly enriched pathways and functions of the genes in the clusters were also screened. Finally, potential molecular markers were identified using the support vector machine (SVM) method.

Materials and methods

Obtaining and preprocessing of mRNA expression profile data

The mRNA expression profiles of sepsis and non-sepsis samples were obtained from the National Center of Biotechnology Information Gene Expression Omnibus database. The access number was GSE12624 (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE12624) and the datasets of 36 samples with septic shock following trauma (sepsis samples) and 34 samples without septic shock following trauma (non-sepsis samples) were used for further analysis. The platform used here was called GPL4204 GE Healthcare/Amersham Biosciences CodeLink UniSet Human I Bioarray. The original data at probe symbol level were first converted into expression values at gene symbol level. Next, missing data was imputed and median data normalization was performed using robust multichip averaging (23). Besides, principal component analysis (PCA) (24), which was used as a computational procedure for biomarker identification and for the classification of multiclass gene expression was performed to identify the difference between sepsis and non-sepsis samples.

PPI network construction

PPIs illustrate valuable information for the elucidation of cellular function, and protein interaction studies have been developed to be a focal point of recent biomolecular research. The Human Protein Reference Database (HPRD) (25) is a novel protein information resource illustrating various features of proteins, including the domain architecture, molecular function, tissue expression, subcellular localization, enzyme-substrate correlation and PPIs. In the present study, all the human PPI pairs in HPRD were initially collected. Next, the Pearson correlation coefficients for all the interacting genes were calculated based on their expression values under the sepsis and non-sepsis status with a coefficient <0.5 used as the cut-off criterion. This was done to obtain the PPI networks under these two statuses. Furthermore, Cytoscape (26) was used to visualize the PPI networks in order to further observe the correlation between genes.

Hierarchical clustering and analysis of covariance (ANCOVA) global test for differentially expressed clusters

Hierarchical clustering is a method of cluster analysis that seeks to build a hierarchy of clusters (27). Euclidean distance was selected as a measure of distance between pairs of genes in the PPI network. The present study used the package hclust (http://CRAN.R-project.org/package=gplots) in R language to perform the hierarchical clustering of two PPI networks, with the requirement that each cluster should have had >5 genes. Finally, Package GlobalAncova in R language was employed to identify the differentially expressed cluster using the ANCOVA global test (28), which focuses on phenotype effects and gene-phenotype interactions. P<0.05 was defined as a threshold.

Enrichment analysis of differentially expressed clusters

In order to study differentially expressed clusters at the functional level, Gene Ontology (GO) functional enrichment (29) and Kyoto Encyclopedia of Gene and Genomes (30) pathway enrichment analyses were performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID) software (31). DAVID software has been widely used to identify biological processes involving a given list of genes. In the present study, a fold change discovery of ≤0.05 was set as the cut-off criterion for enrichment analysis.

Identification of molecular markers by the SVM method

The SVM method has been demonstrated to be an useful classification and regression method that uses machine learning theory to maximize the predictive accuracy while avoiding overfitting of data (32). Differential clusters were initially ranked according to their P- and F-values (F test), and the cluster with the highest P-value was then defined as the class feature. Secondly, the SVM method was employed to classify the samples using the ksvm function in the kernlab package in R language, and the 10-fold cross-validation method was performed to evaluate the classification results. A feature selection error rate of <0.1 was selected as the criteria.

Results

Preprocessing of mRNA expression profile data

A total of 7,672 genes were obtained after preprocessing of the mRNA expression profile data. PCA analysis based on gene expression values revealed that sepsis samples had a great similarity with non-sepsis samples (Fig. 1). Therefore, further bioinformatics analyses were conducted to identify the molecular markers for distinguishing sepsis samples from non-sepsis samples.

PPI network construction

The HPRD database was used to construct the PPI network and Cytoscape software was employed to visualize the network. The PPI network of genes under sepsis collected 1,996 genes and 2,645 interactions between them; the PPI network of genes under non-sepsis status collected 2,147 genes and 2,783 interactions (Fig. 2). Further analysis revealed that there were 1,438 overlapping genes and only 992 overlapping interactions between sepsis and non-sepsis samples (Fig. 3).

Screening of differentially expressed clusters

In total, 40 clusters with 20 clusters in both PPI networks were identified by hierarchical clustering (Fig. 4). Furthermore, 24 differentially expressed clusters, each with 12 clusters in both networks, were identified by the ANCOVA global test with a threshold of P<0.05.

Enrichment analysis of differentially expressed clusters

Function enrichment analysis revealed that the genes in the 24 differentially expressed clusters were mainly enriched in the following GO terms: Positive regulation of macromolecule metabolic processes, transcription factor binding, regulation of cell death and the intracellular signaling cascade (Table I). However, no pathways were enriched.

Table I.

GO analysis of differentially expressed clusters (top 15).

Table I.

GO analysis of differentially expressed clusters (top 15).

CategoryTermFDR
GOTERM_BP_FATGO:0010604~positive regulation of macromolecule metabolic process 6.58×10−29
GOTERM_MF_FAT GO:0008134~transcription factor binding 1.09×10−27
GOTERM_BP_FATGO:0010605~negative regulation of macromolecule metabolic process 2.52×10−23
GOTERM_BP_FAT GO:0010941~regulation of cell death 1.83×10−22
GOTERM_BP_FAT GO:0007242~intracellular signaling cascade 2.59×10−22
GOTERM_BP_FAT GO:0043067~regulation of programmed cell death 8.30×10−22
GOTERM_MF_FAT GO:0030528~transcription regulator activity 7.43×10−21
GOTERM_BP_FAT GO:0042981~regulation of apoptosis 1.21×10−20
GOTERM_MF_FATGO:0004672~protein kinase activity 1.08×10−19
GOTERM_BP_FAT GO:0016310~phosphorylation 7.73×10−19
GOTERM_BP_FATGO:0006468~protein amino acid phosphorylation 8.91×10−19
GOTERM_BP_FATGO:0044093~positive regulation of molecular function 4.92×10−17
GOTERM_BP_FATGO:0010628~positive regulation of gene expression 5.97×10−17
GOTERM_BP_FAT GO:0006796~phosphate metabolic process 6.14×10−17
GOTERM_BP_FAT GO:0006793~phosphorus metabolic process 6.14×10−17

[i] GO, gene ontology; BP, biological process; FAT, functional annotation tool; MF, molecular function; FDR, false discovery rate.

Identification of molecular markers by the SVM method

The 24 clusters that were sorted by P-value are listed in Table II. The SVM method was employed in order to classify the samples according to the cluster sequences listed in Table II. Notably, when the seventh cluster was added, the feature selection error rate of 10-fold cross-validation was >0.1. Therefore, 207 genes in the top six clusters were selected as potential biomarkers, including CDC42, CSF3R, GCA, HMGB2, RHOG, SERPINB1 and TYROBP in cluster 15, as well as SERPINA1, FCER1G and S100P. Fig. 5 illustrates the SVM calculating process. In order to validate the non-randomness of these 207 genes, 207 genes were randomly selected from the mRNA expression profile data 1,000 times and the results of the 10-fold cross-validation were always >0.1. This proved the effectiveness of the selected molecular markers. Further enrichment analysis demonstrated that these 207 genes were mainly enriched in functions of the intracellular signaling cascade, regulation of programmed cell death, regulation of cell death, regulation of apoptosis and leukocyte activation (Table III); whereas no pathways were enriched.

Table II.

Sorted differentially expressed clusters of sepsis and non-sepsis samples.

Table II.

Sorted differentially expressed clusters of sepsis and non-sepsis samples.

ClustersF-valuep.permp.approxState
159.12<0.01   2.74×10−6s
186.66<0.01   4.87×10−3ns
146.03<0.01   1.06×10−4s
105.26<0.01   7.52×10−4ns
134.83<0.01   1.25×10−5ns
  84.61<0.01   1.40×10−3s
133.94<0.01   4.86×10−6s
  33.77<0.01   2.92×10−6ns
  23.58<0.01   6.30×10−4s
113.25<0.01   2.37×10−3ns
  33.23<0.01   2.26×10−3s
113.03<0.01   3.31×10−3s
  22.94<0.01   6.03×10−3ns
  52.82<0.01   2.06×10−2s
  62.82<0.01   6.06×10−3s
  42.64<0.01   8.88×10−3ns
164.88   0.01   1.11×10−3ns
  63.76   0.01   6.36×10−4ns
  43.42   0.01   2.21×10−3s
  82.94   0.01   5.65×10−3ns
  92.43   0.02   4.01×10−2s
122.26   0.02   3.19×10−2s
  12.65   0.03   1.53×10−2ns
  52.36   0.04   1.87×10−2ns

[i] s, septic samples; ns, non-septic samples; p-perm, p-values from the permutation test; p.approx, P-values by means of an approximation for a mixture of χ2 distribution.

Table III.

GO analysis of molecular markers (top 15).

Table III.

GO analysis of molecular markers (top 15).

CategoryTermFDR
GOTERM_CC_FAT GO:0005829~cytosol 1.33×10−8
GOTERM_BP_FAT GO:0007242~intracellular signaling cascade 3.76×10−5
GOTERM_CC_FAT GO:0031982~vesicle 9.54×10−3
GOTERM_CC_FATGO:0044459~plasma membrane part 1.12×10−2
GOTERM_BP_FATGO:0010033~response to organic substance 1.39×10−2
GOTERM_BP_FAT GO:0043067~regulation of programmed cell death 1.40×10−2
GOTERM_CC_FATGO:0005886~plasma membrane 1.46×10−2
GOTERM_BP_FAT GO:0010941~regulation of cell death 1.50×10−2
GOTERM_CC_FATGO:0015629~actin cytoskeleton 1.67×10−2
GOTERM_MF_FATGO:0032403~protein complex binding 2.73×10−2
GOTERM_BP_FAT GO:0042981~regulation of apoptosis 3.31×10−2
GOTERM_BP_FAT GO:0045321~leukocyte activation 6.04×10−2
GOTERM_BP_FATGO:0031400~negative regulation of protein modification process 7.36×10−2
GOTERM_CC_FATGO:0009986~cell surface 7.38×10−2
GOTERM_CC_FAT GO:0031988~membrane-bounded vesicle 7.53×10−2
GOTERM_BP_FAT GO:0016192~vesicle-mediated transport 9.46×10−2

[i] GO, gene ontology; CC, cellular components; FAT, functional annotation tool; BP, biological process; MF, molecular function; FDR, false discovery rate.

Discussion

Sepsis and its complications are a common cause of infectious disease and hospital-reported mortality worldwide (33). The present study aimed to investigate the potential mechanism of sepsis, and to identify genes that can be used for diagnosing and developing candidate molecularly targeted therapy. In total, 7,672 genes were obtained after preprocessing of the mRNA expression profile data. Following hierarchical clustering analysis and the ANCOVA global test, 24 differentially expressed clusters with 12 clusters in each PPI network were identified. Moreover, 207 genes in the top six clusters were selected using SVM, and the functional enrichment analysis revealed that they were mainly enriched in the intracellular signaling cascade, regulation of programmed cell death, regulation of apoptosis and leukocyte activation.

The ANCOVA global test identified 24 differentially expressed clusters, and cluster 15 had the highest P-value. Genes in this cluster, including CDC42 (34), CSF3R (35), GCA (36), HMGB2 (37), RHOG (38), SERPINB1 (39) and TYROBP (40) had already been linked with sepsis. SERPINA1, FCER1G and S100P were also genes in this cluster. Gene SERPINA1 encodes alpha-1-antitrypsin, which is a serine protease inhibitor. Moreover, the targets of SERPINA1 include elastase, plasmin, thrombin, trypsin, chymotrypsin and plasminogen activator, which participates in inflammatory processes (41). Buttenschoen et al (42) demonstrated that the diagnostic value of SERPINA1 levels could be applied in order to distinguish sepsis from SIRS and to assess prognosis. Recently, Su et al (43) revealed that SERPINA1 was downregulated in patients with sepsis compared with SIRS patients, and further analysis demonstrated that SERPINA1 was involved in sepsis differentiation. The FCER1G gene encodes the γ-subunit of Fc epsilon RI (FcRγ), which is an immunoreceptor tyrosine-based activation motif-bearing signal transduction subunit of the Fc receptor family (44). The FCER1G gene was upregulated in sepsis according to Hu et al (45). Furthermore, it has a deleterious effect on sepsis, and FcRγ-/− mice demonstrated an increased survival during sepsis due to increased Escherichia coli phagocytosis (46). The S100P gene encoding the S100 calcium binding protein, which is a member of the S100 family proteins, contains 2 EF-hand calcium-binding motifs. Sepsis-associated encephalopathy (SAE) is the organ dysfunction accompanied with sepsis (47). S100P is produced mainly by the central nervous system, and the elevated serum level of S100P is a biomarker of neuronal damage occurring in SAE (48). Therefore, elevated serum levels of S100P may be associated with critical illness and may be treated as the biomarkers of brain damage during sepsis.

GO functional enrichment analysis revealed that 207 genes identified by SVM, including TLR2 and RAB27A, were mainly enriched in the intracellular signaling cascade, regulation of programmed cell death and cell death, regulation of apoptosis and leukocyte activation. Furthermore, programmed cell death is an important mechanism during the immunopathogenesis of sepsis. Notably, apoptosis is one form of programmed cell death. In addition, early programmed cell death of lymphocytes destroys innate and adaptive immunity, which would reduce the ability of protecting against pathogens (49). Also, programmed cell death of parenchymal cells in the lung, liver and gut would facilitate organ failure and death (50). The TLR2 gene encodes Toll-Like Receptor 2, which is a member of the Toll-like receptor family expressed on the macrophage recognizing pathogen-associated molecular patterns (51). Several reports have suggested the role of TLR2 in the induction of pathogen-induced programmed cell death (5254). The present study identified that TLR2 demonstrated differential expression in sepsis samples, which was consistent with the observations of Armstrong et al (55). Therefore, we inferred that TLR2 may be involved in sepsis by interrupting programmed cell death. The RAB27A gene encoding guanosine triphosphate (GTP)-binding protein Ram belongs to the GTPase superfamily, Rab family (56). Ménasché et al (57) have reported that genetic defects in Rab27a may lead to immunodeficiency in humans caused by programmed cell death. Furthermore, Johnson et al (58) illustrated that Rab27a deficiency is associated with increased survival and reduced neutrophil infiltration of the liver in a model of lipopolysaccharide-induced systemic inflammation. Therefore, Rab27a may participate in organ failure which accompanies sepsis.

Overall, in order to illustrate the pathological mechanisms underlying sepsis, gene expression profiles containing 70 samples were downloaded and analyzed. SERPINA1, FCER1G and S100P in the selected differential clusters may be potential biomarkers. Moreover, TLR2 and Rab27a may exert certain roles in sepsis by interrupting programmed cell death. However, more experiments are required in order to confirm these results.

References

1 

Pierrakos C and Vincent JL: Sepsis biomarkers: A review. Crit Care. 14:R152010. View Article : Google Scholar : PubMed/NCBI

2 

American College of Chest Physicians/Society of Critical Care Medicine Consensus Conference: Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Crit Care Med. 20:864–874. 1992. View Article : Google Scholar : PubMed/NCBI

3 

Soong J and Soni N: Sepsis: Recognition and treatment. Clin Med. 12:276–280. 2012. View Article : Google Scholar

4 

Levy MM, Dellinger RP, Townsend SR, Linde-Zwirble WT, Marshall JC, Bion J, Schorr C, Artigas A, Ramsay G and Beale R: Surviving Sepsis Campaign: The surviving sepsis campaign: Results of an international guideline-based performance improvement program targeting severe sepsis. Crit Care Med. 38:367–374. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Mayr FB, Yende S and Angus DC: Epidemiology of severe sepsis. Virulence. 5:4–11. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Venet F and Chung CS: Increased circulating regulatory T cells (CD4(+)CD25 (+)CD127 (−)) contribute to lymphocyte anergy in septic shock patients. Intensive Care Med. 35:678–686. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Tidswell M, Tillis W, Larosa SP, Lynn M, Wittek AE, Kao R, Wheeler J, Gogate J and Opal SM: Phase 2 trial of eritoran tetrasodium (E5564), a Toll-like receptor 4 antagonist, in patients with severe sepsis. Crit Care Med. 38:72–83. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Machado JR, Soave DF, Da SM, de Menezes LB, Etchebehere RM, Monteiro ML, dos Reis MA, Corrêa RR and Celes MR: Neonatal sepsis and inflammatory mediators. Mediators Inflamm. 2014:269681. 2014.PubMed/NCBI

9 

Nancy B, Diana S, Kerstin H, Oliver K, Katrin L, Michael B, Martin BF, Diana I and Michael K: C-Terminal Alpha-1 Antitrypsin Peptide: A New Sepsis Biomarker with Immunomodulatory Function. Mediators Inflamm. 2016:1–13. 2016.

10 

Chi YF, Chai JK, Yu YM, Luo HM, Zhang QX and Feng R: Association between PAI-1 polymorphisms and plasma PAI-1 level with sepsis in severely burned patients. Genet Mol Res. 14:10081–10086. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Wang JF, Yu MG, Bian JJ, Deng XM, Wan XJ and Zhu KM: Serum miR-146a and miR-223 as potential new biomarkers for sepsis. Biochem Biophys Res Commun. 394:184–188. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Póvoa P, Coelho L, Almeida E, Fernandes A, Mealha R, Moreira P and Sabino H: C-reactive protein as a marker of infection in critically ill patients. Clin Microbiol Infect. 11:101–108. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Schmit X and Vincent JL: The time course of blood C-reactive protein concentrations in relation to the response to initial antimicrobial therapy in patients with sepsis. Infection. 36:213–219. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Luzzani A, Polati E, Dorizzi R, Rungatscher A, Pavan R and Merlini A: Comparison of procalcitonin and C-reactive protein as markers of sepsis. Crit Care Med. 31:1737–1741. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Gabay C and Kushner I: Acute-phase proteins and other systemic responses to inflammation. N Engl J Med. 340:448–454. 1999. View Article : Google Scholar : PubMed/NCBI

16 

Hanriot D, Bello G, Ropars ADC, Poitevin G, Grosjean S, Latger-Cannard V, Devaux Y, Zannad F, Regnault V and Lacolley P: C-reactive protein induces pro- and anti-inflammatory effects, including activation of the liver X receptor alpha, on human monocytes. Thromb Haemost. 99:558–569. 2008.PubMed/NCBI

17 

Culley FJ, Bodmansmith KB, Ferguson MA, Nikolaev AV, Shantilal N and Raynes JG: C-reactive protein binds to phosphorylated carbohydrates. Glycobiology. 10:59–65. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Assicot M, Gendrel D, Carsin H, Raymond J, Guilbaud J and Bohuon C: High serum procalcitonin concentrations in patients with sepsis and infection. Lancet. 341:515–518. 1993. View Article : Google Scholar : PubMed/NCBI

19 

O'Grady NP, Barie PS, Bartlett JG, Bleck T, Carroll K, Kalil AC, Linden P, Maki DG, Nierman D, Pasculle W and Masur H: American College of Critical Care Medicine; Infectious Diseases Society of America: Guidelines for evaluation of new fever in critically ill adult patients: 2008 update from the American college of critical care medicine and the infectious diseases society of America. Crit Care Med. 36:1330–1349. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Snider RH Jr, Nylen ES and Becker KL: Procalcitonin and its component peptides in systemic inflammation: Immunochemical characterization. J Investig Med. 45:552–560. 1997.PubMed/NCBI

21 

Tang BM, Eslick GD, Craig JC and McLean AS: Accuracy of procalcitonin for sepsis diagnosis in critically ill patients: Systematic review and meta-analysis. Lancet Infect Dis. 7:210–217. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Wacker C, Prkno A, Brunkhorst FM and Schlattmann P: Procalcitonin as a diagnostic marker for sepsis: A systematic review and meta-analysis. Lancet Infect Dis. 13:426–435. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U and Speed TP: Exploration, normalization and summaries of high density oligonucleotide array probe level data. Biostatistics. 4:249–264. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Bicciato S, Luchini A and Di Bello C: PCA disjoint models for multiclass cancer analysis using gene expression data. Bioinformatics. 19:571–578. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Peri S, Navarro JD, Kristiansen TZ, Amanchy R, Surendranath V, Muthusamy B, Gandhi TK, Chandrika KN, Deshpande N, Suresh S, et al: Human protein reference database as a discovery resource for proteomics. Nucleic Acids Res. 32:(Database issue). D497–D501. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Cameron DA, Middleton FA, Chenn A and Olson EC: Hierarchical clustering of gene expression patterns in the Eomes+lineage of excitatory neurons during early neocortical development. BMC Neurosci. 13:902012. View Article : Google Scholar : PubMed/NCBI

28 

Mansmann U and Meister R: Testing differential gene expression in functional groups. Goeman's global test versus an ANCOVA approach. Methods Inf Med. 44:449–453. 2005.PubMed/NCBI

29 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene ontology: Tool for the unification of biology. The gene ontology consortium. Nat Genet. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

30 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar : PubMed/NCBI

31 

da Huang W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Liu J, Li SC and Luo X: Iterative reweighted noninteger norm regularizing SVM for gene expression data classification. Comput Math Methods Med. 2013:7684042013. View Article : Google Scholar : PubMed/NCBI

33 

Jawad I, Lukšić I and Rafnsson SB: Assessing available information on the burden of sepsis: Global estimates of incidence, prevalence and mortality. J Glob Health. 2:0104042012. View Article : Google Scholar : PubMed/NCBI

34 

Geörg M, Maudsdotter L, Tavares R and Jonsson AB: Meningococcal resistance to antimicrobial peptides is mediated by bacterial adhesion and host cell RhoA and Cdc42 signalling. Cell Microbiol. 15:1938–1954. 2013.PubMed/NCBI

35 

Rauch PJ, Chudnovskiy A, Robbins CS, Weber GF, Etzrodt M, Hilgendorf I, Tiglao E, Figueiredo JL, Iwamoto Y, Theurl I, et al: Innate response activator B cells protect against microbial sepsis. Science. 335:597–601. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Durand M and Thomas SL: Incidence of infections in patients with giant cell arteritis: A cohort study. Arthritis Care Res (Hoboken). 64:581–588. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Metukuri MR, Reddy CM, Reddy PR and Reddanna P: Bacterial LPS-mediated acute inflammation-induced spermatogenic failure in rats: Role of stress response proteins and mitochondrial dysfunction. Inflammation. 33:235–243. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Sista F, Schietroma M, Santis GD, Mattei A, Cecilia EM, Piccione F, Leardi S, Carlei F and Amicucci G: Systemic inflammation and immune response after laparotomy vs laparoscopy in patients with acute cholecystitis, complicated by peritonitis. World J Gastrointest Surg. 5:73–82. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Benarafa C: The SerpinB1 knockout mouse a model for studying neutrophil protease regulation in homeostasis and inflammation. Methods Enzymol. 499:135–148. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Ormsby T, Schlecker E, Ferdin J, Tessarz AS, Angelisová P, Köprülü AD, Borte M, Warnatz K, Schulze I, Ellmeier W, Horejsí V and Cerwenka A: Btk is a positive regulator in the TREM-1/DAP12 signaling pathway. Blood. 118:936–945. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Ganter U, Bauer J, Schulz-Huotari C, Gebicke-Haerter PJ, Beeser H and Gerok W: Repression of alpha 2-macroglobulin and stimulation of alpha 1-proteinase inhibitor synthesis in human mononuclear phagocytes by endotoxin. Eur J Biochem. 169:13–20. 1987. View Article : Google Scholar : PubMed/NCBI

42 

Buttenschoen K, Buttenschoen DC, Berger D, Vasilescu C, Schafheutle S, Goeltenboth B, Seidelmann M and Beger HG: Endotoxemia and acute-phase proteins in major abdominal surgery. Am J Surg. 181:36–43. 2001. View Article : Google Scholar : PubMed/NCBI

43 

Su L, Zhou R, Liu C, Wen B, Xiao K, Kong W, Tan F, Huang Y, Cao L and Xie L: Urinary proteomics analysis for sepsis biomarkers with iTRAQ labeling and two-dimensional liquid chromatography-tandem mass spectrometry. J Trauma Acute Care Surg. 74:940–945. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Butchar JP, Mehta P, Justiniano SE, Guenterberg KD, Kondadasula SV, Mo X, Chemudupati M, Kanneganti TD, Amer A and Muthusamy N: Reciprocal regulation of activating and inhibitory Fc{gamma} receptors by TLR7/8 activation: implications for tumor immunotherapy. Clin Cancer Res. 16:2065–2075. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Hu WC: Sepsis is a syndrome with hyperactivity of TH17-like innate immunity and hypoactivity of adaptive immunity. ArXiv Preprint ArXiv:. 1311:47472013.

46 

da Pinheiro Silva F, Aloulou M, Skurnik D, Benhamou M, Andremont A, Velasco IT, Chiamolera M, Verbeek JS, Launay P and Monteiro RC: CD16 promotes Escherichia coli sepsis through an FcR gamma inhibitory pathway that prevents phagocytosis and facilitates inflammation. Nat Med. 13:1368–1374. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Iacobone E, Bailly-Salin J, Polito A, Friedman D, Stevens RD and Sharshar T: Sepsis-associated encephalopathy and its differential diagnosis. Crit Care Med. 37:(Suppl 10). S331–S336. 2009. View Article : Google Scholar : PubMed/NCBI

48 

Hsu AA, Fenton K, Weinstein S, Carpenter J, Dalton H and Bell MJ: Neurological injury markers in children with septic shock. Pediatr Crit Care Med. 9:245–251. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Vodounon CA, Chabi CB, Skibo YV, Ezin V, Aikou N, Kotchoni SO, Akpona SA, Babamoussa L and Abramova ZI: Influence of the programmed cell death of lymphocytes on the immunity of patients with atopic bronchial asthma. Allergy Asthma Clin Immunol. 10:1–11. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Perl M, Chung CS, Swan R and Ayala A: Role of programmed cell death in the immunopathogenesis of sepsis. Drug Discov Today Dis Mech. 4:223–230. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Kurko J, Vähä-Mäkilä M, Tringham M, Tanner L, Paavanen-Huhtala S, Saarinen M, Näntö-Salonen K, Simell O, Niinikoski H and Mykkänen J: Dysfunction in macrophage toll-like receptor signaling caused by an inborn error of cationic amino acid transport. Mol Immunol. 67:416–425. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Bermpohl D, Halle A, Freyer D, Dagand E, Braun JS, Bechmann I, Schröder NW and Weber JR: Bacterial programmed cell death of cerebral endothelial cells involves dual death pathways. J Clin Invest. 115:1607–1615. 2005. View Article : Google Scholar : PubMed/NCBI

53 

Bergsbaken T, Fink SL and Cookson BT: Pyroptosis: Host cell death and inflammation. Nat Rev Microbiol. 7:99–109. 2009. View Article : Google Scholar : PubMed/NCBI

54 

Into T, Kiura K, Yasuda M, Kataoka H, Inoue N, Hasebe A, Takeda K, Akira S and Shibata K: Stimulation of human Toll-like receptor (TLR) 2 and TLR6 with membrane lipoproteins of Mycoplasma fermentans induces apoptotic cell death after NF-kappa B activation. Cell Microbiol. 6:187–199. 2004. View Article : Google Scholar : PubMed/NCBI

55 

Armstrong L, Medford AR, Hunter KJ, Uppington KM and Millar AB: Differential expression of Toll-like receptor (TLR)-2 and TLR-4 on monocytes in human sepsis. Clin Exp Immunol. 136:312–319. 2004. View Article : Google Scholar : PubMed/NCBI

56 

Menasche G, Feldmann J, Houdusse A, Desaymard C, Fischer A, Goud B and De SBG: Biochemical and functional characterization of Rab27a mutations occurring in Griscelli syndrome patients. Blood. 101:2736–2742. 2003. View Article : Google Scholar : PubMed/NCBI

57 

Ménasché G, Pastural E, Feldmann J, Certain S, Ersoy F, Dupuis S, Wulffraat N, Bianchi D, Fischer A, Le Deist F and de Saint Basile G: Mutations in RAB27A cause Griscelli syndrome associated with haemophagocytic syndrome. Nat Genet. 25:173–176. 2000. View Article : Google Scholar : PubMed/NCBI

58 

Johnson JL, Hong H, Monfregola J and Catz SD: Increased survival and reduced neutrophil infiltration of the liver in Rab27a- but not Munc13-4-deficient mice in lipopolysaccharide-induced systemic inflammation. Infect Immun. 79:3607–3618. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 13 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang Y and Yang Y: Identification of potential biomarkers of sepsis using bioinformatics analysis. Exp Ther Med 13: 1689-1696, 2017
APA
Yang, Y., & Yang, Y. (2017). Identification of potential biomarkers of sepsis using bioinformatics analysis. Experimental and Therapeutic Medicine, 13, 1689-1696. https://doi.org/10.3892/etm.2017.4178
MLA
Yang, Y., Li, L."Identification of potential biomarkers of sepsis using bioinformatics analysis". Experimental and Therapeutic Medicine 13.5 (2017): 1689-1696.
Chicago
Yang, Y., Li, L."Identification of potential biomarkers of sepsis using bioinformatics analysis". Experimental and Therapeutic Medicine 13, no. 5 (2017): 1689-1696. https://doi.org/10.3892/etm.2017.4178