Hyporesponsiveness of natural killer cells and impaired inflammatory responses in critically ill patients

Minkyung Kim, Minjoo Kim, Hana Jeong, Jey Sook Chae, Young Sam Kim, Jae Gil Lee, Younsoo Cho, Jong Ho Lee, Minkyung Kim, Minjoo Kim, Hana Jeong, Jey Sook Chae, Young Sam Kim, Jae Gil Lee, Younsoo Cho, Jong Ho Lee

Abstract

Background: To investigate natural killer (NK) cell activity, circulating cytokine level and peripheral blood mononuclear cell (PBMC) cytokine production status in critically ill patients.

Methods: Blood samples were collected <24 h after admission from 24 intensive care unit (ICU) patients and 24 age-, sex-, and body mass index (BMI)-matched healthy controls. Serum cytokine concentrations and cytokine production by PBMCs and lipopolysaccharide (LPS)-stimulated PBMCs were measured.

Results: The ICU group showed lower NK cell activity than the controls under all conditions and an absence of interferon (IFN)-γ. After adjusting for triglycerides, LDL- and HDL-cholesterol, and glucose, the ICU group exhibited lower serum levels of albumin and interleukin (IL)-12 and higher leukocyte counts and hs-CRP and IL-6 levels than the controls. Non-stimulated PBMCs from ICU patients secreted significantly greater amounts of IL-6 and IL-1β than the controls; however, the production of IL-6, TNF-α and IL-1β in response to LPS stimulation was significantly lower in the ICU group.

Conclusions: Significant reductions in NK cell activity and serum IL-12 level, an absence of serum IFN-γ, and decreased cytokine production from LPS-stimulated PBMCs indicate the hyporesponsiveness of NK cells and an impaired early phase inflammatory response in critically ill patients (ClinicalTrials.gov NCT02565589 :). Retrospectively registered; October 1, 2015.

Keywords: Cytokine; ICU; Inflammatory response; NK cell; PBMC.

Conflict of interest statement

Ethics approval and consent to participate

Written informed consent was provided by a close family member. This investigation was approved by the Institutional Review Board at Yonsei University Severance Hospital (Seoul, Korea; Approval Number: 4–2013-0902) and conducted according to the Helsinki Declaration.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Correlation matrix among clinical and inflammatory parameters in control and ICU patient groups. Correlations were obtained by deriving Pearson’s correlation coefficient. Red denotes a positive correlation, and blue denotes a negative correlation

References

    1. Andaluz-Ojeda D, Iglesias V, Bobillo F, et al. Early levels in blood of immunoglobulin M and natural killer cells predict outcome in nonseptic critically ill patients. J Crit Care. 2013;28:1110. doi: 10.1016/j.jcrc.2013.06.007.
    1. Adib-Conquy M, Cavaillon JM. Compensatory anti-inflammatory response syndrome. Thromb Haemost. 2009;101:36–47.
    1. Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med. 2013;369:840–851. doi: 10.1056/NEJMra1208623.
    1. Ward NS, Casserly B, Ayala A. The compensatory anti-inflammatory response syndrome (CARS) in critically ill patients. Clin Chest Med. 2008;29:617–625. doi: 10.1016/j.ccm.2008.06.010.
    1. Monneret G, Venet F, Kullberg BJ, Netea MG. ICU-acquired immunosuppression and the risk for secondary fungal infections. Med Mycol. 2011;49(Suppl 1):S17–S23. doi: 10.3109/13693786.2010.509744.
    1. Opal SM. Immunologic alterations and the pathogenesis of organ failure in the ICU. Semin Respir Crit Care Med. 2011;32:569–580. doi: 10.1055/s-0031-1287865.
    1. Marshall JC, Charbonney E, Gonzalez PD. The immune system in critical illness. Clin Chest Med. 2008;29:605–616. doi: 10.1016/j.ccm.2008.08.001.
    1. Spolarics Z, Siddiqi M, Siegel JH, et al. Depressed interleukin-12-producing activity by monocytes correlates with adverse clinical course and a shift toward Th2-type lymphocyte pattern in severely injured male trauma patients. Crit Care Med. 2003;31:1722–1729. doi: 10.1097/01.CCM.0000063579.43470.AA.
    1. Weighardt H, Heidecke CD, Westerholt A, et al. Impaired monocyte IL-12 production before surgery as a predictive factor for the lethal outcome of postoperative sepsis. Ann Surg. 2002;235:560–567. doi: 10.1097/00000658-200204000-00015.
    1. Kimura F, Shimizu H, Yoshidome H, Ohtsuka M, Miyazaki M. Immunosuppression following surgical and traumatic injury. Surg Today. 2010;40:793–808. doi: 10.1007/s00595-010-4323-z.
    1. Wu HP, Shih CC, Chu CM, et al. Effect of interleukin-17 on in vitro cytokine production in healthy controls and patients with severe sepsis. J Formos Med Assoc. 2015;114:1250–1257. doi: 10.1016/j.jfma.2014.09.009.
    1. Wu HP, Shih CC, Lin CY, Hua CC, Chuang DY. Serial increase of IL-12 response and human leukocyte antigen-DR expression in severe sepsis survivors. Crit Care. 2011;15:R224. doi: 10.1186/cc10464.
    1. Hamers L, Kox M, Pickkers P. Sepsis-induced immunoparalysis: mechanisms, markers, and treatment options. Minerva Anestesiol. 2015;81:426–439.
    1. Drewry AM, Ablordeppey EA, Murray ET, et al. Comparison of monocyte human leukocyte antigen-DR expression and stimulated tumor necrosis factor alpha production as outcome predictors in severe sepsis: a prospective observational study. Crit Care. 2016;20:334. doi: 10.1186/s13054-016-1505-0.
    1. Hall MW, Knatz NL, Vetterly C, et al. Immunoparalysis and nosocomial infection in children with multiple organ dysfunction syndrome. Intensive Care Med. 2011;37:525–532. doi: 10.1007/s00134-010-2088-x.
    1. Hall MW, Geyer SM, Guo CY, et al. Innate immune function and mortality in critically ill children with influenza: a multicenter study. Crit Care Med. 2013;41:224–236. doi: 10.1097/CCM.0b013e318267633c.
    1. Mandal A, Viswanathan C. Natural killer cells: in health and disease. Hematol Oncol Stem Cell Ther. 2015;8:47–55. doi: 10.1016/j.hemonc.2014.11.006.
    1. Huntington ND, Vosshenrich CA, Di Santo JP. Developmental pathways that generate natural-killer-cell diversity in mice and humans. Nat Rev Immunol. 2007;7:703–714. doi: 10.1038/nri2154.
    1. Badgwell B, Parihar R, Magro C, Dierksheide J, Russo T, Carson WE., 3rd Natural killer cells contribute to the lethality of a murine model of Escherichia Coli infection. Surgery. 2002;132:205–212. doi: 10.1067/msy.2002.125311.
    1. Etogo AO, Nunez J, Lin CY, Toliver-Kinsky TE, Sherwood ER. NK but not CD1-restricted NKT cells facilitate systemic inflammation during polymicrobial intra-abdominal sepsis. J Immunol. 2008;180:6334–6345. doi: 10.4049/jimmunol.180.9.6334.
    1. Forel JM, Chiche L, Thomas G, et al. Phenotype and functions of natural killer cells in critically-ill septic patients. PLoS One. 2012;7:e50446. doi: 10.1371/journal.pone.0050446.
    1. Knaus WA, Draper EA, Wagner DP, Zimmerman JE. APACHE II: a severity of disease classification system. Crit Care Med. 1985;13:818–829. doi: 10.1097/00003246-198510000-00009.
    1. Dellinger RP, Carlet JM, Masur H, et al. Surviving sepsis campaign guidelines for management of severe sepsis and septic shock. Crit Care Med. 2004;32:858–873. doi: 10.1097/01.CCM.0000117317.18092.E4.
    1. Dellinger RP, Levy MM, Carlet JM, et al. Surviving se psis campaign: international guidelines for management of severe sepsis and septic shock: 2008. Crit Care Med. 2008;36:296–327. doi: 10.1097/01.CCM.0000298158.12101.41.
    1. Lee JG, Kim YS, Lee YJ, et al. Effect of immune-enhancing Enteral nutrition enriched with or without Beta-Glucan on Immunomodulation in critically ill patients. Nutrients. 2016;8:336. doi: 10.3390/nu8060336.
    1. Kjaergaard AG, Nielsen JS, Tønnesen E, Krog J. Expression of NK cell and monocyte receptors in critically ill patients--potential biomarkers of sepsis. Scand J Immunol. 2015;81:249–258. doi: 10.1111/sji.12272.
    1. Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, Yokoyama WM, Ugolini S. Innate or adaptive immunity? The example of natural killer cells. Science. 2011;331:44–49. doi: 10.1126/science.1198687.
    1. Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev Immunol. 2003;3:133–146. doi: 10.1038/nri1001.
    1. Stanilova SA, Karakolev ZT, Dimov GS, et al. High interleukin 12 and low interleukin 10 production after in vitro stimulation detected in sepsis survivors. Intensive Care Med. 2005;31:401–407. doi: 10.1007/s00134-005-2575-7.
    1. Landelle C, Lepape A, Voirin N, et al. Low monocyte human leukocyte antigen-DR is independently associated with nosocomial infections after septic shock. Intensive Care Med. 2010;36:1859–1866. doi: 10.1007/s00134-010-1962-x.
    1. Calder PC, Jensen GL, Koletzko BV, Singer P, Wanten GJ. Lipid emulsions in parenteral nutrition of intensive care patients: current thinking and future directions. Intensive Care Med. 2010;36:735–749. doi: 10.1007/s00134-009-1744-5.
    1. Ott J, Hiesgen C, Mayer K. Lipids in critical care medicine. Prostaglandins Leukot Essent Fatty Acids. 2011;85:267–273. doi: 10.1016/j.plefa.2011.04.011.
    1. Barros KV, Cassulino AP, Schalch L, et al. Pharmaconutrition: acute fatty acid modulation of circulating cytokines in elderly patients in the ICU. JPEN J Parenter Enteral Nutr. 2014;38:467–474. doi: 10.1177/0148607113480183.
    1. Giger U, Büchler M, Farhadi J, et al. Preoperative immunonutrition suppresses perioperative inflammatory response in patients with major abdominal surgery-a randomized controlled pilot study. Ann Surg Oncol. 2007;14:2798–2806. doi: 10.1245/s10434-007-9407-7.
    1. Damas P, Ledoux D, Nys M, et al. Cytokine serum level during severe sepsis in human IL-6 as a marker of severity. Ann Surg. 1992;215:356–362. doi: 10.1097/00000658-199204000-00009.
    1. Shaw AC, Joshi S, Greenwood H, Panda A, Lord JM. Aging of the innate immune system. Curr Opin Immunol. 2010;22:507–513. doi: 10.1016/j.coi.2010.05.003.
    1. Valente SA, Fallon WF, Jr, Alexander TS, et al. Immunologic function in the elderly after injury--the neutrophil and innate immunity. J Trauma. 2009;67:968–974. doi: 10.1097/TA.0b013e3181b84279.
    1. Brüünsgaard H, Pedersen BK. Age-related inflammatory cytokines and disease. Immunol Allergy Clin N Am. 2003;23:15–39. doi: 10.1016/S0889-8561(02)00056-5.
    1. Gomez CR, Karavitis J, Palmer JL, et al. Interleukin-6 contributes to age-related alteration of cytokine production by macrophages. Mediat Inflamm. 2010;2010:475139. doi: 10.1155/2010/475139.
    1. Ottinger ME, Monaghan SF, Gravenstein S, Cioffi WG, Ayala A, Heffernan DS. The geriatric cytokine response to trauma: time to consider a new threshold. Surg Infect. 2014;15:800–805. doi: 10.1089/sur.2013.235.
    1. Jing Y, Shaheen E, Drake RR, Chen N, Gravenstein S, Deng Y. Aging is associated with a numerical and functional decline in plasmacytoid dendritic cells, whereas myeloid dendritic cells are relatively unaltered in human peripheral blood. Hum Immunol. 2009;70:777–784. doi: 10.1016/j.humimm.2009.07.005.

Source: PubMed

3
Suscribir