Prague Med. Rep. 2018, 119, 9-29

https://doi.org/10.14712/23362936.2018.2

Sepsis Diagnostics in the Era of “Omics” Technologies

Miroslav Průcha1, Roman Zazula2, Stefan Russwurm3

1Department of Clinical Biochemistry, Haematology and Immunology, Na Homolce Hospital, Prague, Czech Republic
2Department of Anesthesiology and Intensive Care, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
3Department of Anesthesiology and Intensive Care, University Hospital Jena, Jena, Germany

Received December 5, 2017
Accepted March 19, 2018

References

1. Adhikari, N. K., Fowle, R. A., Bhagwanjee, S., Rubenfeld, G. D. (2010) Critical care and the global burden of critical illness in adults. Lancet 376, 1339–1346. <https://doi.org/10.1016/S0140-6736(10)60446-1>
2. Akira, S., Uematsu, S., Takeuchi, O. (2006) Pathogen recognition and innate immunity. Cell 124, 783–801. <https://doi.org/10.1016/j.cell.2006.02.015>
3. Ambroggio, L., Florin, T. A., Shah, S. S., Ruddy, R., Yeomans, L., Trexel, J., Stringer, K. A. (2017) Emerging biomarkers of illness severity: Urinary metabolites associated with sepsis and necrotizing methicillin-resistant Staphylococcus aureus pneumonia. Pharmacotherapy 37, 1033–1042. <https://doi.org/10.1002/phar.1973> <PubMed>
4. Angus, D. C., Linde-Zwirble, W. T., Lidicker, J., Clermont, G., Garcillo, J., Pinsky, M. R. (2001) Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit. Care Med. 29, 1303–1310. <https://doi.org/10.1097/00003246-200107000-00002>
5. Arcaroli, J., Silva, E., Maloney, J. P., He, Q., Svetkauskaite, D., Murphy, J. R., Abraham, E. (2006) Variant IRAK-1 haplotype is associated with increased nuclear factor-kappa B activation and worse outcomes in sepsis. Am. J. Respir. Crit. Care Med. 173, 1335–1341. <https://doi.org/10.1164/rccm.200603-341OC> <PubMed>
6. Ayala, A., Lomas, J. L., Grutkoski, P. S., Grutkoski, P. S., Chung, C. S. (2003) Pathological aspects of apoptosis in severe sepsis and shock? Int. J. Biochem. Cell Biol. 35, 715–720. <https://doi.org/10.1016/S1357-2725(02)00099-7>
7. Becker, K. L., Snider, R., Nylen, E. S. (2008) Procalcitonin assay in systemic inflammation, infection, and sepsis: clinical utility and limitations. Crit. Care Med. 6, 941–952. <https://doi.org/10.1097/CCM.0B013E318165BABB>
8. Bierne, H., Hamon, M., Cossart, P. (2012) Epigenetics and bacterial infections. Cold Spring Harb. Perspect. Med. 2, a010272. <https://doi.org/10.1101/cshperspect.a010272> <PubMed>
9. Biron, B. M., Ayala, A., Lomas-Neira, L. J. (2015) Biomarkers for sepsis: What is and what might be? Biomark. Insights 10(S4), 7–17.
10. Biswas, S. K., Bist, P., Dhillon, M. K., Kajiji, T., del Fresno, C., Yamamoto, M., Lopez-Collazo, E., Akira, S., Tergaonkar, V. (2007) Role for MyD88-independent, TRIF pathway in lipid A/TLR4-induced endotoxin tolerance. J. Immunol. 179, 4083–4092. <https://doi.org/10.4049/jimmunol.179.6.4083>
11. Bombard, Y., Bach, P. B., Offit, K. (2013) Translating genomics in cancer care. J. Natl. Compr. Canc. Netw. 11, 1343–1353. <https://doi.org/10.6004/jnccn.2013.0158>
12. Boomer, J. S., To, K., Chang, K. C., Takasu, O., Osborne, D. F., Walton, A. H., Bricker, T. L., Jarman, S. D., Kreisel, D., Krupnick, A. S., Srivastava, A., Swanson, P. E., Green, J. M., Hotchkiss, R. S. (2011) Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA 306, 2594–2605. <https://doi.org/10.1001/jama.2011.1829> <PubMed>
13. Brunkhorst, F. M., Reinhart, K. (2005) German competence network for the study of severe sepsis and septic shock (SepNet). ICU Manag. Pract. 5.
14. Buhimschi, C. S., Bhandari, V., Dulay, A. T., Nayeri, U. A., Abdel-Razeq, S. S., Pettker, C. M., Thung, S., Zhao, G., Han, Y. W., Bizzarro, M., Buhimschi, I. A. (2011) Proteomics mapping of cord blood identifies haptoglobin “switch-on” pattern as biomarker of early-onset neonatal sepsis in preterm newborns. PLoS One 6, e26111. <https://doi.org/10.1371/journal.pone.0026111> <PubMed>
15. Bustamante, J., Boisson-Dupuisa, S., Abela, L., Casanova, J. L. (2014) Mendelian susceptibility to mycobacterial disease: Genetic, immunological, and clinical features of inborn errors of IFN-γ immunity. Semin. Immunol. 26, 454–470. <https://doi.org/10.1016/j.smim.2014.09.008> <PubMed>
16. Calandra, T., Echtenacher, B., Roy, D. L., Pugin, J., Metz, N., Hültner, L., Heumann, D., Männel, D., Bucala, R., Glauser, M. P. (2000) Protection from septic shock by neutralization of macrophage migration inhibitory factor. Nat. Med. 6, 164–170. <https://doi.org/10.1038/72262>
17. Calvano, S. E., Xiao, W., Richards, D. R. (2005) A network-based analysis of systemic inflammation in humans. Nature 437, 1032–1037. <https://doi.org/10.1038/nature03985>
18. Carpio, R., Zapata, J., Spanuth, E., Hess, G. (2015) Utility of presepsin (sCD14-ST) as a diagnostic and prognostic marker of sepsis in the emergency department. Clin. Chim. Acta 450, 169–175. <https://doi.org/10.1016/j.cca.2015.08.013>
19. Cavaillon, J. M., Adib-Conquy, M., Fitting, C., Adrie, C., Payen, D. (2003) Cytokine cascade in sepsis. Scand. J. Infect. Dis. 35, 535–544. <https://doi.org/10.1080/00365540310015935>
20. Cazalis, M. A., Lepape, A., Venet, F., Frager, F., Mougin, B., Vallin, H., Paye, M., Pachot, A., Monneret, G. (2014) Early and dynamic changes in gene expression in septic shock patients: a genome-wide approach. Intensive Care Med. Exp. 2, 20–37. <https://doi.org/10.1186/s40635-014-0020-3> <PubMed>
21. Chien, J. W., Boeckh, M. J., Hansen, J. A., Clark, J. G. (2008) Lipopolysaccharide binding protein promoter variants influence the risk for gram-negative bacteremia and mortality after allogeneic hematopoietic cell transplantation. Blood 111, 2462–2469. <https://doi.org/10.1182/blood-2007-09-101709> <PubMed>
22. Ciardiello, F., Arnold, D., Casali, P. G., Cervantes, J. Y., Douillard, A., Eggermont, A., Eniu, K., McGregor, S., Piccart, P. M., Popescu, R., Van Cutsem, E., Zielinski, R., Stahel, R. (2014) Delivering precision medicine in oncology today and in future-the promise and challenges of personalised cancer medicine: a position paper by the European Society for Medical Oncology (ESMO). Ann. Oncol. 25, 1673–1678. <https://doi.org/10.1093/annonc/mdu217>
23. Cobb, J. P., Laramie, J. M., Stormo, G., Morrissey, J. J., Shannon, W. D., Qiu, Y., Karl, I. E., Buchman, T. G., Hotchkiss, R. S. (2002) Sepsis gene expression profiling: Murine splenic compared with hepatic responses determined by using complementary DNA microarrays. Crit. Care Med. 30, 2711–2721. <https://doi.org/10.1097/00003246-200212000-00016>
24. Dahm, R. (2010) From discovering to understanding. EMBO Rep. 11, 153–160. <https://doi.org/10.1038/embor.2010.14> <PubMed>
25. Davenport, E. E., Burnham, K. L., Radhakrishnan, J., Humburg, P., Hutton, P., Mills, T. C., Rautanen, A., Gordon, A. C., Garrard, C., Hill, A. V., Hinds, C. J., Knight, J. C. (2016) Genomic landscape of the individual host response and outcomes in sepsis: a prospective cohort study. Lancet Respir. Med. 4, 259–271. <https://doi.org/10.1016/S2213-2600(16)00046-1> <PubMed>
26. De La Rica, A. S., Gilsanz, F., Maseda, E. (2016) Epidemiologic trends of sepsis in western countries. Ann. Transl. Med. 4, 325–331. <https://doi.org/10.21037/atm.2016.08.59> <PubMed>
27. del Fresno, C., Gómez-Piña, V., Lores, V., Soares-Schanoski, A., Fernández-Ruiz, I., Rojo, B., Alvarez-Sala, R., Caballero-Garrido, E., García, F., Veliz, T., Arnalich, F., Fuentes-Prior, P., García-Río, F., López-Collazo, E. (2008) Monocytes from cystic fibrosis patients are locked in an LPS tolerance state: down-regulation of TREM-1 as putative underlying mechanism. PLoS One 3, e2667. <https://doi.org/10.1371/journal.pone.0002667> <PubMed>
28. del Fresno, C., García-Rio, F., Gómez-Piña, V., Soares-Schanoski, A., Fernández-Ruíz, I., Jurado, T., Kajiji, T., Shu, C., Marín, E., Gutierrez del Arroyo, A., Prados, C., Arnalich, F., Fuentes-Prior, P., Biswas, S. K., López-Collazo, E. (2009) Potent phagocytic activity with impaired antigen presentation identifying lipopolysaccharide-tolerant human monocytes: demonstration in isolated monocytes from cystic fibrosis patients. J. Immunol. 182, 6494–6507. <https://doi.org/10.4049/jimmunol.0803350>
29. Dong, X., Weng, Z. (2013) The correlation between histone modifications and gene expression. Epigenomics 5, 113–116. <https://doi.org/10.2217/epi.13.13> <PubMed>
30. Esper, A., Martin, G. S. (2007) Is severe sepsis increasing in incidence and severity? Crit. Care Med. 35, 1414–1415. <https://doi.org/10.1097/01.CCM.0000262946.68003.21>
31. Esteller, M. (2007) Cancer epigenomics: DNA methylomes and histone-modification maps. Nat. Rev. Genet. 8, 286–298. <https://doi.org/10.1038/nrg2005>
32. Everett, J. R. (2016) From metabonomics to pharmacometabonomics: the role of metabolic profiling in personalized medicine. Front. Pharmacol. 7, 297. <https://doi.org/10.3389/fphar.2016.00297> <PubMed>
33. Fang, X. M., Schröder, S., Hoeft, A., Stuber, F. (1999) Comparison of two polymorphisms of the interleukin-1 gene family: interleukin-1 receptor antagonist polymorphism contributes to susceptibility to severe sepsis. Crit. Care Med. 27, 1330–1334. <https://doi.org/10.1097/00003246-199907000-00024>
34. Feezor, R. J., Cheng, A., Paddock, H. N., Baker, H. V., Moldawer, L. L. (2005) Functional genomics and gene expression profiling in sepsis: beyond class prediction. Clin. Infect. Dis. 41, S427–S435. <https://doi.org/10.1086/431993>
35. Ferrario, M., Cambiaghi, A., Brunelli, L., Giordano, S., Caironi, P., Guatteri, L., Raimondi, F., Gattinoni, L., Latini, R., Masson, S., Ristagno, G., Pastorelli, R. (2016) Mortality prediction in patients with severe septic shock: a pilot study using a target metabolomics approach. Sci. Rep. 6, 20391. <https://doi.org/10.1038/srep20391> <PubMed>
36. Fessenden, M. (2016) Metabolomics: small molecules, single cells. Nature 540, 153–155. <https://doi.org/10.1038/540153a>
37. Feterowski, C., Emmanuilidis, K., Miethke, T., Gerauer, K., Rump, M., Ulm, K., Holzmann, B., Weighardt, H. (2003) Effects of functional Toll-like receptor-4 mutations on the immune response to human and experimental sepsis. Immunology 109, 426–431. <https://doi.org/10.1046/j.1365-2567.2003.01674.x> <PubMed>
38. Fiusa, M. M., Carvalho, B. S., Hubert, R. M., Souza, W., Lopes-Cendes, I., Annichino-Bizzacchi, J. M., De Paula, E. V. (2014) A meta-analysis of gene expression studies in severe sepsis and septic shock. Blood 124, 2741.
39. Garcia-Simon, M., Morales, J. M., Modesto-Alapont, V., Gonzalez-Marrachelli, V., Vento-Rehues, R., Jorda-Miñana, A., Blanquer-Olivas, J., Monleon, D. (2015) Prognosis biomarkers of severe sepsis and septic shock by 1H NMR urine metabolomics in the intensive care unit. PLoS One 10, e0140993. <https://doi.org/10.1371/journal.pone.0140993> <PubMed>
40. Gawad, C., Koh, W., Quake, S. R. (2016) Single-cell genome sequencing: current state of the science. Nat. Rev. Genet. 17, 175–188. <https://doi.org/10.1038/nrg.2015.16>
41. Geiger, T., Velic, A., Macek, B., Lundberg, E., Kampf, C., Nagaraj, N., Uhlen, M., Cox, J., Mannet, M. (2013) Initial quantitative proteomic map of 28 mouse tissues using the SILAC mouse. Mol. Cell. Proteomics 12, 1709–1722. <https://doi.org/10.1074/mcp.M112.024919> <PubMed>
42. Ginsburg, G. S., Willard, H. F. (2009) Genomic and personalized medicine: foundations and applications. Transl. Res. 154, 277–287. <https://doi.org/10.1016/j.trsl.2009.09.005>
43. Gocek, E., Moulas, A. N., Studzinski, G. P. (2014) Non-receptor protein tyrosine kinases signaling pathways in normal and cancer cells. Crit. Rev. Clin. Lab. Sci. 51, 125–137. <https://doi.org/10.3109/10408363.2013.874403>
44. Gootenberg, J. S., Abudayyeh, O. O., Lee, J. W., Essletzbichler, P., Dy, A. J., Joung, J., Verdine, V., Donghia, N., Daringer, N. M., Freije, C. A., Myhrvold, C., Bhattacharyya, R. P., Livny, J., Regev, A., Koonin, E. V., Hung, D. T., Sabeti, P. C., Collins, J. J., Zhang, F. (2017) Nucleic acid detection with CRISPR-Cas13a/C2c2. Science 356, 438–442. <https://doi.org/10.1126/science.aam9321> <PubMed>
45. Grealy, R., White, M., Stordeur, P., Kelleher, D., Doherty, D. G., McManus, R., Ryan, T. (2013) Characterising cytokine gene expression signatures in patients with severe sepsis. Mediators Inflamm. 2013, 164246. <https://doi.org/10.1155/2013/164246> <PubMed>
46. Hazeldine, J., Hampson, P., Lord, J. M. (2016) The diagnostic and prognostic value of systems biology research in major traumatic and thermal injury: a review. Burns Trauma 4, 33–44. <https://doi.org/10.1186/s41038-016-0059-3> <PubMed>
47. Hermans, P. W., Hibberd, M. L., Booy, R., Daramola, O., Hazelzet, J. A., de Groot, R., Levin, M. (1999) 4G/5G promoter polymorphism in the plasminogen-activator-inhibitor-1 gene and outcome of meningococcal disease. Meningococcal Research Group. Lancet 354, 556–560. <https://doi.org/10.1016/S0140-6736(99)02220-5>
48. Hinrichs, C., Kotsch, K., Buchwald, S., Habicher, M., Saak, N., Gerlach, H., Volk, H. D., Keh, D. (2010) Perioperative gene expression analysis for prediction of postoperative sepsis. Clin. Chem. 56, 613–622. <https://doi.org/10.1373/clinchem.2009.133876>
49. Hotchkiss, R. S., Monneret, G., Payen, D. (2013) Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat. Rev. Immunol. 13, 862–874. <https://doi.org/10.1038/nri3552> <PubMed>
50. Huan, T., Forsberg, E. M., Rinehart, D., Johnson, C. H., Ivanisevic, J., Benton, H. P., Fang, M., Aisporna, A., Hilmers, B., Poole, F. L., Thorgersen, M. P., Adams, M. W. W., Krantz, G., Fields, M. W., Robbins, P. D., Niedernhofer, L. J., Ideker, T., Majumder, E. L., Wall, J. D., Rattray, N. J. W., Goodacre, R., Lairson, L. L., Siuzdak, G. (2017) Systems biology guided by XCMS online metabolomics. Nat. Methods 14, 461–462. <https://doi.org/10.1038/nmeth.4260> <PubMed>
51. Huang, W., Tang, Y., Li, L. (2010) HMGB1, a potent proinflammatory cytokine in sepsis. Cytokine 51, 119–126. <https://doi.org/10.1016/j.cyto.2010.02.021>
52. Hubacek, J. A., Stüber, F., Fröhlich, D., Book, M., Wetegrove, S., Ritter, M., Rothe, G., Schmitz, G. (2001) Gene variants of the bactericidal/permeability increasing protein and lipopolysaccharide binding protein in sepsis patients: gender-specific genetic predisposition to sepsis. Crit. Care Med. 29, 557–561. <https://doi.org/10.1097/00003246-200103000-00015>
53. Huttlin, E. L., Jedrychowski, M. P., Elias, J. E., Goswami, T., Rad, R., Beausoleil, S. A., Villén, J., Haas, W., Sowa, M. E., Gygi, S. P. (2010) A tissue-specific atlas of mouse protein phosphorylation and expression. Cell 143, 1174–1189. <https://doi.org/10.1016/j.cell.2010.12.001> <PubMed>
54. Hynes, S. O., Pang, B., James, J. A., Maxwell, P., Salto-Tellez, M. (2017) Tissue-based next generation sequencing: application in a universal healthcare system. Br. J. Cancer 116, 553–560. <https://doi.org/10.1038/bjc.2016.452> <PubMed>
55. Jawad, I., Luksic, I., Rafnsson, S. B. (2012) Assessing available information on the burden of sepsis: global estimates of incidence, prevalence and mortality. J. Glob. Health 2, 010404. <https://doi.org/10.7189/jogh.01.010404>
56. Johnson, S. B., Lissauer, M., Bochicchio, G. V., Moore, R., Cross, A. S., Scalea, T. M. (2007) Gene expression profiles differentiate between sterile SIRS and early sepsis. Ann. Surg. 245, 611–621. <https://doi.org/10.1097/01.sla.0000251619.10648.32> <PubMed>
57. Kaddurah-Daouk, R., Weinshilboum, R.; Pharmacometabolomics Research Network (2015) Metabolomic signatures for drug response phenotypes: Pharmacometabolomics enables precision medicine. Clin. Pharmacol. Ther. 98, 71–75. <https://doi.org/10.1002/cpt.134> <PubMed>
58. Kauppi, A. M., Edin, A., Ziegler, I., Mölling, P., Sjösted, A., Gylfe, A., Strålin, K., Johansson, A. (2016) Metabolites in blood for prediction of bacteremic sepsis in the emergency room. PLoS One 11, e0147670. <https://doi.org/10.1371/journal.pone.0147670> <PubMed>
59. Khor, C. C., Chapman, S. J., Vannberg, F. O., Dunne, A., Murphy, C., Ling, E. Y., Frodsham, A. J., Walley, A. J., Kyrieleis, O., Khan, A., Aucan, C., Segal, S., Moore, C. E., Knox, K., Campbell, S. J., Lienhardt, C., Scott, A., Aaby, P., Sow, O. Y., Grignani, R. T., Sillah, J., Sirugo, G., Peshu, N., Williams, T. N., Maitland, K., Davies, R. J., Kwiatkowski, D. P., Day, N. P., Yala, D., Crook, D. W., Marsh, K., Berkley, J. A., O’Neill, L. A., Hill, A. V. (2007) A Mal functional variant is associated with protection against invasive pneumococcal disease, bacteremia, malaria and tuberculosis. Nat. Genet. 39, 523–528. <https://doi.org/10.1038/ng1976> <PubMed>
60. Kono, H., Rock, K. L. (2008) How dying cells alert the immune system to danger. Nat. Rev. Immunol. 8, 279–289. <https://doi.org/10.1038/nri2215> <PubMed>
61. Kosmides, A. K., Kamisoglu, K., Calvano, S. E., Corbett, S. A., Androulakis, I. P. (2013) Metabolomic fingerprinting: challenges and opportunities. Crit. Rev. Biomed. Eng. 41, 205–221. <https://doi.org/10.1615/CritRevBiomedEng.2013007736>
62. Kumar, A. T., Sudhir, U., Punith, K., Kumar, R., Kumar, V. N., Rao, M. Y. (2009a) Cytokine profile in elderly patients with sepsis. Indian J. Crit. Care Med. 13, 74–78.
63. Kumar, A., Ellis, P., Arabi, Y., Roberts, D., Light, B., Parrillo, J. E., Dodek, P., Wood, G., Kumar, A., Simon, D., Peters, C., Ahsan, M., Chateau, D.; Cooperative Antimicrobial Therapy of Septic Shock Database Research Group (2009b) Initiation of inappropriate antimicrobial therapy results in a fivefold reduction of survival in human septic shock. Chest 136, 1237–1248. <https://doi.org/10.1378/chest.09-0087>
64. Kumar, G., Kumar, A., Taneja, A., Kaleekal, T., Tarima, S., McGinley, E., Jimenez, E., Mohan, A., Khan, R. A., Whittle, J., Jacobs, E., Nanchal, R.; Milwaukee Initiative in Critical Care Outcomes Research (MICCOR) Group of Investigators (2011) Nationwide trends of severe sepsis in the 21st century (2000–2007). Chest 140, 1223–1231. <https://doi.org/10.1378/chest.11-0352>
65. Lander, E. S., Linton, L. M., Birren, B., Nusbaum, C., Zody, M. C., Baldwin, J., Devon, K., Dewar, K., Doyle, M., FitzHugh, W., Funke, R., Gage, D., Harris, K., Heaford, A., Howland, J., Kann, L., Lehoczky, J., LeVine, R., McEwan, P., McKernan, K., Meldrim, J., Mesirov, J. P., Miranda, C., Morris, W., Naylor, J., Raymond, C., Rosetti, M., Santos, R., Sheridan, A., Sougnez, C., Stange-Thomann, Y., Stojanovic, N., Subramanian, A., Wyman, D., Rogers, J., Sulston, J., Ainscough, R., Beck, S., Bentley, D., Burton, J., Clee, C., Carter, N., Coulson, A., Deadman, R., Deloukas, P., Dunham, A., Dunham, I., Durbin, R., French, L., Grafham, D., Gregory, S., Hubbard, T., Humphray, S., Hunt, A., Jones, M., Lloyd, C., McMurray, A., Matthews, L., Mercer, S., Milne, S., Mullikin, J. C., Mungall, A., Plumb, R., Ross, M., Shownkeen, R., Sims, S., Waterston, R. H., Wilson, R. K., Hillier, L. W., McPherson, J. D., Marra, M. A., Mardis, E. R., Fulton, L. A., Chinwalla, A. T., Pepin, K. H., Gish, W. R., Chissoe, S. L., Wendl, M. C., Delehaunty, K. D., Miner, T. L., Delehaunty, A., Kramer, J. B., Cook, L. L., Fulton, R. S., Johnson, D. L., Minx, P. J., Clifton, S. W., Hawkins, T., Branscomb, E., Predki, P., Richardson, P., Wenning, S., Slezak, T., Doggett, N., Cheng, J. F., Olsen, A., Lucas, S., Elkin, C., Uberbacher, E., Frazier, M., Gibbs, R. A., Muzny, D. M., Scherer, S. E., Bouck, J. B., Sodergren, E. J., Worley, K. C., Rives, C. M., Gorrell, J. H., Metzker, M. L., Naylor, S. L., Kucherlapati, R. S., Nelson, D. L., Weinstock, G. M., Sakaki, Y., Fujiyama, A., Hattori, M., Yada, T., Toyoda, A., Itoh, T., Kawagoe, C., Watanabe, H., Totoki, Y., Taylor, T., Weissenbach, J., Heilig, R., Saurin, W., Artiguenave, F., Brottier, P., Bruls, T., Pelletier, E., Robert, C., Wincker, P., Smith, D. R., Doucette-Stamm, L., Rubenfield, M., Weinstock, K., Lee, H. M., Dubois, J., Rosenthal, A., Platzer, M., Nyakatura, G., Taudien, S., Rump, A., Yang, H., Yu, J., Wang, J., Huang, G., Gu, J., Hood, L., Rowen, L., Madan, A., Qin, S., Davis, R. W., Federspiel, N. A., Abola, A. P., Proctor, M. J., Myers, R. M., Schmutz, J., Dickson, M., Grimwood, J., Cox, D. R., Olson, M. V., Kaul, R., Raymond, C., Shimizu, N., Kawasaki, K., Minoshima, S., Evans, G. A., Athanasiou, M., Schultz, R., Roe, B. A., Chen, F., Pan, H., Ramser, J., Lehrach, H., Reinhardt, R., McCombie, W. R., de la Bastide, M., Dedhia, N., Blöcker, H., Hornischer, K., Nordsiek, G., Agarwala, R., Aravind, L., Bailey, J. A., Bateman, A., Batzoglou, S., Birney, E., Bork, P., Brown, D. G., Burge, C. B., Cerutti, L., Chen, H. C., Church, D., Clamp, M., Copley, R. R., Doerks, T., Eddy, S. R., Eichler, E. E., Furey, T. S., Galagan, J., Gilbert, J. G., Harmon, C., Hayashizaki, Y., Haussler, D., Hermjakob, H., Hokamp, K., Jang, W., Johnson, L. S., Jones, T. A., Kasif, S., Kaspryzk, A., Kennedy, S., Kent, W. J., Kitts, P., Koonin, E. V., Korf, I., Kulp, D., Lancet, D., Lowe, T. M., McLysaght, A., Mikkelsen, T., Moran, J. V., Mulder, N., Pollara, V. J., Ponting, C. P., Schuler, G., Schultz, J., Slater, G., Smit, A. F., Stupka, E., Szustakowki, J., Thierry-Mieg, D., Thierry-Mieg, J., Wagner, L., Wallis, J., Wheeler, R., Williams, A., Wolf, Y. I., Wolfe, K. H., Yang, S. P., Yeh, R. F., Collins, F., Guyer, M. S., Peterson, J., Felsenfeld, A., Wetterstrand, K. A., Patrinos, A., Morgan, M. J., de Jong, P., Catanese, J. J., Osoegawa, K., Shizuya, H., Choi, S., Chen, Y. J., Szustakowki, J.; International Human Genome Sequencing Consortium (2001) Initial sequencing and analysis of the human genome. Nature 409, 860–921. <https://doi.org/10.1038/35057062>
66. Leentjens, J., Kox, M., van der Hoeven, J. G., Netea, M. G., Pickkers, P. (2013) Immunotherapy for the adjunctive treatment of sepsis: from immunosuppression to immunostimulation. Time for a paradigm change? Am. J. Respir. Crit. Care Med. 187, 1287–1293. <https://doi.org/10.1164/rccm.201301-0036CP>
67. Leliefeld, H. F., Wessels, C. M., Leenan, L. P., Koenderman, L., Pillay, J. (2016) The role of neutrophils in immune dysfunction during severe inflammation. Crit. Care 20, 73–82. <https://doi.org/10.1186/s13054-016-1250-4> <PubMed>
68. Li, Y. R., Kauffman, J. M. (2014) Molecular medicine of rheumatoid arthritis: From molecular pathophysiology to novel therapeutics and evidence-based practice. Ann. Orthop. Rheumatol. 2, 1014–1024.
69. Lissauer, M. E., Johnson, S. B., Bochicchio, G. V., Carinda, J., Feild, A. S., Cross, J. D., Hasday, C. C., Whiteford, W. A., Nussbaumer, M. T., Scalea, T. (2009) Differential expression of Toll-like receptor genes: sepsis compared with sterile inflammation 1 day before sepsis diagnosis. Shock 31, 238–244. <https://doi.org/10.1097/SHK.0b013e3181834991>
70. López-Collazo, E., Fuentes-Prior, P., Arnalich, F., del Fresno, C. (2006) Pathophysiology of interleukin-1 receptor-associated kinase-M: implications in refractory state. Curr. Opin. Infect. Dis. 19, 237–244. <https://doi.org/10.1097/01.qco.0000224817.35105.7d>
71. Malmström, E., Kilsgård, O., Hauri, S., Smeds, E., Herwald, H., Malmström, L., Malmström, J. (2016) Large-scale inference of protein tissue origin in gram-positive sepsis plasma using quantitative targeted proteomics. Nat. Commun. 7, 10261. <https://doi.org/10.1038/ncomms10261> <PubMed>
72. Man, M., Close, S. L., Shaw, A. D., Bernard, G. R., Douglas, I. S., Kaner, R. J. (2013) Beyond single-marker analyses: Mining whole genome scans for insights into treatment responses in severe sepsis. Pharmacogenomics J. 13, 218–226. <https://doi.org/10.1038/tpj.2012.1>
73. Mansur, A., Liese, B., Steinau, M., Ghadimi, M., Bergmann, I., Tzvetkov, M., Popov, A. F., Beissbarth, T., Bauer, M., Hinz, J. (2015) The CD14 rs2569190 TT genotype is associated with an improved 30-days survival in patients with sepsis: a prospective observational study. PLoS One 10, e0127761. <https://doi.org/10.1371/journal.pone.0127761> <PubMed>
74. Marshall, J. C. (2014) Why have clinical trials in sepsis failed? Trends Mol. Med. 20, 195–203. <https://doi.org/10.1016/j.molmed.2014.01.007>
75. Martin, G. S. (2012) Sepsis, severe sepsis and septic shock: Changes in incidence, pathogens and outcomes. Expert Rev. Anti Infect. Ther. 10, 701–706. <https://doi.org/10.1586/eri.12.50> <PubMed>
76. Martin, G., Brunkhorst, F. M., Janes, J. M., Reinhart, K., Sundin, D. P., Garnett, K., Beale, R. (2009) The international PROGRESS registry of patients with severe sepsis: drotrecogin alfa (activated) use and patients outcomes. Crit. Care 13, R103. <https://doi.org/10.1186/cc7936> <PubMed>
77. Medvedev, A. E., Lentschat, A., Kuhns, D. B., Blanco, J. C., Salkowski, C., Zhang, S., Arditi, M., Gallin, J. I., Vogel, S. N. (2003) Distinct mutations in IRAK-4 confer hyporesponsiveness to lipopolysaccharide and interleukin-1 in a patient with recurrent bacterial infections. J. Exp. Med. 198, 521–531. <https://doi.org/10.1084/jem.20030701> <PubMed>
78. Medzhitov, R. (2001) Toll-like receptors and innate immunity. Nat. Rev. Immunol. 1, 135–139. <https://doi.org/10.1038/35100529>
79. Mesri, M. (2014) Advances in proteomic technologies and its contribution to the field of cancer. Adv. Med. 2014, 238045. <https://doi.org/10.1155/2014/238045> <PubMed>
80. Ng, P. C., Kirkness, E. F. (2010) Whole genome sequencing. Methods Mol. Biol. 628, 215–226. <https://doi.org/10.1007/978-1-60327-367-1_12>
81. Palmer, C., Diehn, M., Alizadeh, A. A., Brown, P. O. (2006) Cell-type specific gene expression profiles of leukocytes in human peripheral blood. BMC Genomics 7, 115–130. <https://doi.org/10.1186/1471-2164-7-115> <PubMed>
82. Parnell, G. P., McLean, A. S., Booth, D. R., Armstrong, N. J., Nalos, M., Huang, S. J., Manak, J., Tang, W., Tam, O. Y., Chan, S., Tang, B. M. (2012) A distinct influenza infection signature in the blood transcriptome of patients with severe community-acquired pneumonia. Crit. Care 16, R157. <https://doi.org/10.1186/cc11477> <PubMed>
83. Parnell, G. P., Tang, B. M., Nalos, M., Armstrong, N. J., Huang, S. J., Booth, D. R., McLean, A. S. (2013) Identifying key regulatory genes in the whole blood of septic patients to monitor underlying immune dysfunctions. Shock 40, 166–174. <https://doi.org/10.1097/SHK.0b013e31829ee604>
84. Parsons, S. A., Mewburn, J. D., Truesdell, P., Greer, P. A. (2007) The Fps/Fes kinase regulates leucocyte recruitment and extravasation during inflammation. Immunology 122, 542–550. <https://doi.org/10.1111/j.1365-2567.2007.02670.x> <PubMed>
85. Patel, S. J., Sanjana, N. E., Kishton, R. J., Eidizadeh, A., Vodnala, S. K., Cam, M., Gartner, J. J., Jia, L., Steinberg, S. M., Yamamoto, T. N., Merchant, A. S., Mehta, G. U., Chichura, A., Shalem, O., Tran, E., Eil, R., Sukumar, M., Guijarro, E. P., Day, C. P., Robbins, P., Feldman, S., Merlino, G., Zhang, F., Restifo, N. P. (2017) Identification of essential genes for cancer immunotherapy. Nature 548, 537–542. <https://doi.org/10.1038/nature23477> <PubMed>
86. Patti, G. J., Yanes, O., Siuzdak, G. (2012) Innovation: Metabolomics: The apogee of the omics trilogy. Nat. Rev. Mol. Cell Biol. 13, 263–269. <https://doi.org/10.1038/nrm3314> <PubMed>
87. Paugam-Burtz, C., Albuquerque, M., Baron, G., Bert, F., Voitot, H., Delefosse, D., Dondero, F., Sommacale, D., Francoz, C., Hanna, N., Belghiti, J., Ravaud, P., Bedossa, P., Mantz, J., Paradis, V. (2010) Plasma proteome to look for diagnostic biomarkers of early bacterial sepsis after liver transplantation: a preliminary study. Anesthesiology 112, 926–935. <https://doi.org/10.1097/ALN.0b013e3181d049f0>
88. Phillips, T. (2008) The role of methylation in gene expression. Nature Education 1, 116.
89. Prucha, M., Ruryk, A., Boriss, H., Möller, E., Zazula, R., Herold, I., Claus, R. A., Reinhart, K. A., Deigner, P., Russwurm, S. (2004) Expression profiling: Toward an application in sepsis diagnostics. Shock 22, 29–33. <https://doi.org/10.1097/01.shk.0000129199.30965.02>
90. Prucha, M., Bellingan, G., Zazula, R. (2015) Sepsis biomarkers. Clin. Chim. Acta 440, 97–104. <https://doi.org/10.1016/j.cca.2014.11.012>
91. Puskarich, M. A., Finkel, M. A., Karnowsky, A., Jones, A. E., Trexel, J., Harris, B. N., Stringer, K. A. (2015) Pharmacometabolomics of l-carnitine treatment response phenotypes in patients with septic shock. Ann. Am. Thorac. Soc. 12, 46–56. <https://doi.org/10.1513/AnnalsATS.201409-415OC> <PubMed>
92. Rattray, N. J., Kaddurah Daouk, R. (2017) Pharmacometabolomics and precision medicine special issue editorial. Metabolomics 13, 59. <https://doi.org/10.1007/s11306-017-1191-1>
93. Rautanen, A., Mills, T. C., Gordon, A. C., Hutton, P., Steffens, M., Nuamah, R., Chiche, J. D., Parks, T., Chapman, S. J., Davenport, E. E., Elliott, K. S., Bion, J., Lichtner, P., Meitinger, T., Wienker, T. F., Caulfield, M. J., Mein, C., Bloos, F., Bobek, I., Cotogni, P., Sramek, V., Sarapuu, S., Kobilay, M., Ranieri, V. M., Rello, J., Sirgo, G., Weiss, Y. G., Russwurm, S., Schneider, E. M., Reinhart, K., Holloway, P. A., Knight, J. C., Garrard, C. S., Russell, J. A., Walley, K. R., Stüber, F., Hill, A. V., Hinds, C. J.; ESICM/ECCRN GenOSept Investigators (2015) Genome-wide association study of survival from sepsis due to pneumonia: an observational cohort study. Lancet Respir. Med. 3, 53–60. <https://doi.org/10.1016/S2213-2600(14)70290-5> <PubMed>
94. Rhee, C., Dantes, R., Epstein, L., Murphy, D. J., Seymour, C. W., Iwashyna, T. J., Kadri, S. S., Angus, D. C., Danner, R. L., Fiore, A. E., Jernigan, J. A., Martin, G. S., Septimus, E., Warren, D. K., Karcz, A., Chan, C., Menchaca, J. T., Wang, R., Gruber, S., Klompas, M.; CDC Prevention Epicenter Program (2017) Incidence and trends of sepsis in US hospitals using clinical vs. claims data, 2009–2014. JAMA 318, 1241–1249. <https://doi.org/10.1001/jama.2017.13836> <PubMed>
95. Sager, R., Kutz, A., Mueller, B., Schuetz, P. (2017) Procalcitonin-guided diagnosis and antibiotic stewardship revisited. BMC Med. 15, 15–26. <https://doi.org/10.1186/s12916-017-0795-7> <PubMed>
96. Sampson, D. L., Fox, B. A., Yager, T. D., Bhide, S., Cermelli, L. C., McHugh, T. A., Seldon, R. A., Brandon, E., Sullivan, J. J., Zimmerman, M., Noursadeghi, R. B. (2017) A four-biomarker blood signature discriminates systemic inflammation due to viral infection versus other etiologies. Sci. Rep. 7, 2914. <https://doi.org/10.1038/s41598-017-02325-8> <PubMed>
97. Sapru, A., Liu, K. D., Wiemels, J., Hansen, H., Pawlikowska, L., Poon, A., Jorgenson, E., Witte, J. S., Calfee, C. S., Ware, L. B., Matthay, M. A.; NHLBI ARDS Network (2016) Association of common genetic variation in the protein C pathway genes with clinical outcomes in acute respiratory distress syndrome. Crit. Care 20, 151–161. <https://doi.org/10.1186/s13054-016-1330-5> <PubMed>
98. Scherag, A., Schöneweck, F., Kesselmeier, M., Taudien, S., Platzer, M., Felder, M., Sponholz, C., Rautanen, A., Hill, A. V. S., Hinds, C. J., Hossain, H., Suttorp, N., Kurzai, O., Slevogt, H., Giamarellos-Bourboulis, E. J., Armaganidis, A., Trips, E., Scholz, M., Brunkhorst, F. M. (2016) Genetic factors of the disease course after sepsis: a genome-wide study for 28 day mortality. EBioMedicine 12, 239–246. <https://doi.org/10.1016/j.ebiom.2016.08.043> <PubMed>
99. Schnegelsberg, A., Mackenhauer, J., Nibro, H. L., Dreyer, P., Koch, K., Kirkengaard, H. (2016) Impact of socioeconomic status on mortality and unplanned readmission in septic intensive care unit patients. Acta Anaesthesiol. Scand. 60, 465–475. <https://doi.org/10.1111/aas.12644>
100. Scicluna, B. P., van Vught, L. A., Zwinderman, A. H., Wiewel, M. A., Davenport, E. E., Burnham, K. L., Nürnberg, P., Schultz, M. J., Horn, J., Cremer, O. L., Bonten, M. J., Hinds, C. J., Wong, H. R., Knight, J. C., van der Poll, T.; MARS Consortium (2017) Classification of patients with sepsis according to blood genomic endotype: a prospective cohort study. Lancet Respir. Med. 5, 816–826. <https://doi.org/10.1016/S2213-2600(17)30294-1>
101. Seok, J., Warren, H. S., Cuenca, A. G., Mindrinos, M. N., Baker, H. V., Xu, W., Richards, D. R., McDonald-Smith, G. P., Gao, H., Hennessy, L., Finnerty, C. C., López, C. M., Honari, S., Moore, E. E., Minei, J. P., Cuschieri, J., Bankey, P. E., Johnson, J. L., Sperry, J., Nathens, A. B., Billiar, T. R., West, M. A., Jeschke, M. G., Klein, M. B., Gamelli, R. L., Gibran, N. S., Brownstein, B. H., Miller-Graziano, C., Calvano, S. E., Mason, P. H., Cobb, J. P., Rahme, L. G., Lowry, S. F., Maier, R. V., Moldawer, L. L., Herndon, D. N., Davis, R. W., Xiao, W., Tompkins, R. G.; Inflammation and Host Response to Injury, Large Scale Collaborative Research Program (2013) Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc. Natl. Acad. Sci. U. S. A. 110, 3507–3512. <https://doi.org/10.1073/pnas.1222878110> <PubMed>
102. Serkova, N. J., Standiford, T. J., Stringer, K. A. (2011) The emerging field of quantitative blood metabolomics for biomarker discovery in critical illnesses. Am. J. Respir. Crit. Care Med. 184, 647–655. <https://doi.org/10.1164/rccm.201103-0474CI> <PubMed>
103. Shen, H. N., Lu, C. L., Yang, H. H. (2010) Epidemiologic trend of severe sepsis in Taiwan from 1997 through 2006. Chest 138, 298–304. <https://doi.org/10.1378/chest.09-2205>
104. Sjöholm, K., Kilsgård, O., Teleman, J., Happonen, L., Malmström, L., Malmström, J. (2017) Targeted proteomics and absolute protein quantification for the construction of a stoichiometric host-pathogen surface density model. Mol. Cell. Proteomics 16, S29–S41. <https://doi.org/10.1074/mcp.M116.063966> <PubMed>
105. Smolen, J. S., Aletaha, D., McInnes, I. B. (2016) Rheumatoid arthritis. Lancet 388, 2023–2038. <https://doi.org/10.1016/S0140-6736(16)30173-8>
106. Sørensen, T. I., Nielsen, G. G., Andersen, P. K., Teasdale, T. W. (1988) Genetic and environmental influences on premature death in adult adoptees. N. Engl. J. Med. 318, 727–732. <https://doi.org/10.1056/NEJM198803243181202>
107. Srinivasan, L., Page, G., Kirpalani, H., Murray, J. C., Das, A., Higgins, R. D., Carlo, W. A., Bell, E. F., Goldberg, R. N., Schibler, K., Sood, B. G., Stevenson, D. K., Stoll, B. J., Van Meurs, K. P., Johnson, K. J., Levy, J., McDonald, S. A., Zaterka-Baxter, K. M., Kennedy, K. A., Sánchez, P. J., Duara, S., Walsh, M. C., Shankaran, S., Wynn, J. L., Cotton, C. N.; Eunice Kennedy Shriver National Institute of Child Health and Human Development Neonatal Research Network (2017) Genome-wide association study of sepsis in extremely premature infants. Arch. Dis. Child. Fetal Neonatal Ed. 102, F439–F445. <https://doi.org/10.1136/archdischild-2016-311545> <PubMed>
108. Stephens, K. E., Miaskowski, C. A., Levine, J. D., Pullinger, C. R., Aouizerat, B. E. (2013) Epigenetic regulation and measurement of epigenetic changes. Biol. Res. Nurs. 5, 373–381. <https://doi.org/10.1177/1099800412444785> <PubMed>
109. Stüber, F., Petersen, M., Bokelmann, F., Schade, U. (1996) A genomic polymorphism with in the tumor necrosis factor locus influences plasma tumor necrosis factor-alpha concentrations and outcome of patients with severe sepsis. Crit. Care Med. 24, 381–384. <https://doi.org/10.1097/00003246-199603000-00004>
110. Su, L., Cao, L., Zhou, R., Jiang, Z., Xiao, K., Kong, W., Wang, H., Deng, J., Wen, B., Tan, F., Zhang, Y., Xie, L. (2013) Identification of novel biomarkers for sepsis prognosis via urinary proteomic analysis using iTRAQ labeling and 2D-LC-MS/MS. PLos One 8, e54237. <https://doi.org/10.1371/journal.pone.0054237> <PubMed>
111. Su, L., Huang, Y., Zhu, Y., Xia, L., Wang, R., Xiao, K., Wang, H., Yan, P., Wen, B., Cao, L., Meng, N., Luan, H., Liu, C., Li, X., Xie, L. (2014) Discrimination of sepsis stage metabolic profiles with an LC/MS-MS-based metabolomics approach. BMJ Open Respir. Res. 1, e000056. <https://doi.org/10.1136/bmjresp-2014-000056> <PubMed>
112. Sutherland, A. M., Walley, K. R., Russell, J. A. (2005) Polymorphisms in CD14, manose binding lectin, and Toll-like receptor-2 are associated with increased prevalence of infection in critically ill adults. Crit. Care Med. 33, 638–644. <https://doi.org/10.1097/01.CCM.0000156242.44356.C5>
113. Sweeney, T. E., Khatri, P. (2017) Benchmarking sepsis gene expression diagnostics using public data. Crit. Care Med. 45, 1–10. <https://doi.org/10.1097/CCM.0000000000002021> <PubMed>
114. Sweeney, T. E., Shidham, A., Wong, H. R., Khatri, P. (2015) A comprehensive time-course-based multicohort analysis of sepsis and sterile inflammation reveals a robust diagnostic gene set. Sci. Transl. Med. 7, 287ra71. <https://doi.org/10.1126/scitranslmed.aaa5993> <PubMed>
115. Talwar, S. (2006) Gene expression profiles of peripheral blood leukocytes after endotoxin challenge in humans. Physiol. Genomics 25, 203–215. <https://doi.org/10.1152/physiolgenomics.00192.2005> <PubMed>
116. Tang, B. M., McLean, A. S., Dawes, I. W., Huang, S. J., Cowley, M. J., Lin, R. C. (2008) Gene-expression profiling of gram-positive and gram-negative sepsis in critically ill patients. Crit. Care Med. 36, 1125–1128. <https://doi.org/10.1097/CCM.0b013e3181692c0b>
117. Toft, P., Tønnesen, E. (2011) Immune-modulating interventions in critically ill septic patients: pharmacological options. Expert Rev. Clin. Pharmacol. 4, 491–501. <https://doi.org/10.1586/ecp.11.25>
118. van der Poll, T., Opal, S. M. (2008) Host-pathogen interactions in sepsis. Lancet Infect. Dis. 8, 32–43. <https://doi.org/10.1016/S1473-3099(07)70265-7>
119. van Vught, L. A., Wiewel, M. A., Hoogendijk, A. J., Frencken, J. F., Scicluna, B. P., Klein, K., Klouwenberg, P. M. C., Zwinderman, A. H., Lutter, R., Horn, J., Schultz, M. J., Bonten, M. M. J., Cremer, O. L., van der Poll, T. (2017) The host response in patients with sepsis developing intensive care unit-acquired secondary infections. Am. J. Respir. Crit. Care Med. 196, 458–470. <https://doi.org/10.1164/rccm.201606-1225OC>
120. Venter, J. C., Adams, M. D., Myers, E. W., Li, P. W., Mural, R. J. (2001) The sequence of the genome. Science 291, 1304–1351. <https://doi.org/10.1126/science.1058040>
121. Vincent, J. L., Sakr, Y., Sprung, C. L., Gerlach, H., Ranieri, V. M. (2006) Sepsis in European intensive care units: results of the SOAP study. Crit. Care Med. 34, 344–353. <https://doi.org/10.1097/01.CCM.0000194725.48928.3A>
122. Wang, Z. Z., Shi, K., Peng, J. (2017) Serologic testing of a panel of five antibodies in inflammatory bowel diseases: diagnostic value and correlation with disease phenotype. Biomed. Rep. 6, 401–410. <https://doi.org/10.3892/br.2017.860> <PubMed>
123. Watson, J. D., Crick, F. H. (1953) Genetical implications of the structure of deoxyribonucleic acid. Nature 171, 737–738. <https://doi.org/10.1038/171737a0>
124. Weiser, M., Simon, J. M., Kochar, B., Tovar, A., Israel, J. W., Robinson, A., Gipson, G. R., Schaner, M. S., Herfarth, H. H., Sartor, R. B., McGovern, D. P., Rahbar, R., Sadiq, T. S., Koruda, M. J., Furey, T. S., Sheikh, S. Z. (2018) Molecular classification of Crohn’s disease reveals two clinically relevant subtypes. Gut 67, 36–42. <https://doi.org/10.1136/gutjnl-2016-312518> <PubMed>
125. Wiel, E., Lebuffe, G., Vallet, B. (2002) Bacterial CpG DNA in septic shock. In: Yearbook of Intensive Care and Emergency Medicine. Vincent, J. L., Editor, pp. 388–398, Springer, Berlin.
126. Xiao, W., Mindrinos, M. N., Seok, J., Cuschieri, J., Cuenca, A. J., Gao, H., Hayden, D., Hennessy, L., Moore, E. E., Minei, J. P., Bankey, P. E., Johnson, J. L., Sperry, J., Nathens, A. B., Billiar, T. R., West, M. A., Brownstein, B. H., Mason, P. H., Baker, H. V., Finnerty, C. C., Jeschke, M. G., López, M. C., Klein, M. B., Gamelli, R. L., Gibran, N. S., Arnoldo, B., Xu, W., Zhang, Y., Calvano, S. E., McDonald-Smith, G. P., Schoenfeld, D. A., Storey, J. D., Cobb, J. P., Warren, H. S., Moldawer, L. L., Herndon, D. N., Lowry, S. F., Maier, R. V., Davis, R. W., Tompkins, R. G.; Inflammation and Host Response to Injury Large-Scale Collaborative Research Program (2011) A genomic storm in critically injured humans. J. Exp. Med. 208, 2581–2590. <https://doi.org/10.1084/jem.20111354> <PubMed>
127. Yu, K. H., Snyder, M. (2016) Omics profiling in precision oncology. Mol. Cell. Proteomics 16, 2525–2536. <https://doi.org/10.1074/mcp.O116.059253> <PubMed>
128. Yu, S. L., Chen, H. W., Yang, P. C., Peck, K., Tsai, M. H., Chen, J. J., Lin, F. Y. (2004) Differential gene expression in gram-negative and gram-positive sepsis. Am. J. Respir. Crit. Care Med. 169, 1135–1143. <https://doi.org/10.1164/rccm.200211-1278OC>
129. Zhang, X., Liu, D., Liu, Y. N., Wang, R., Xie, L. X. (2015) The accuracy of presepsin (sCD14-ST) for the diagnosis of sepsis in adults: a meta analysis. Crit. Care 19, 323–334. <https://doi.org/10.1186/s13054-015-1032-4> <PubMed>
130. Zimmerman, J. J., Sullivan, E., Yager, T. D., Cheng, C., Permut, L., Cermelli, S., McHugh, L., Sampson, D., Seldon, T., Brandon, R. B., Brandon, R. A. (2017) Diagnostic accuracy of a host gene expression signature that discriminates clinical severe sepsis syndrome and infection-negative systemic inflammation among critically ill children. Crit. Care Med. 45, e418–e425. <https://doi.org/10.1097/CCM.0000000000002100>
front cover

ISSN 1214-6994 (Print) ISSN 2336-2936 (Online)

Archive