Sepsis-induced long-term immune paralysis--results of a descriptive, explorative study

C Arens, S A Bajwa, C Koch, B H Siegler, E Schneck, A Hecker, S Weiterer, C Lichtenstern, M A Weigand, F Uhle, C Arens, S A Bajwa, C Koch, B H Siegler, E Schneck, A Hecker, S Weiterer, C Lichtenstern, M A Weigand, F Uhle

Abstract

Background: Long-lasting impairment of the immune system is believed to be the underlying reason for delayed deaths after surviving sepsis. We tested the hypothesis of persisting changes to the immune system in survivors of sepsis for the first time.

Methods: In our prospective, cross-sectional pilot study, eight former patients who survived catecholamine-dependent sepsis and eight control individuals matched for age, sex, diabetes and renal insufficiency were enrolled. Each participant completed a questionnaire concerning morbidities, medications and infection history. Peripheral blood was collected for determination of i) immune cell subsets (CD4(+), CD8(+) T cells; CD25(+) CD127(-) regulatory T cells; CD14(+) monocytes), ii) cell surface receptor expression (PD-1, BTLA, TLR2, TLR4, TLR5, Dectin-1, PD-1 L), iii) HLA-DR expression, and iv) cytokine secretion (IL-6, IL10, TNF-α, IFN-γ) of whole blood stimulated with either α-CD3/28, LPS or zymosan.

Results: After surviving sepsis, former patients presented with increased numbers of clinical apparent infections, including those typically associated with an impaired immune system. Standard inflammatory markers indicated a low-level inflammatory situation in former sepsis patients. CD8(+) cell surface receptor as well as monocytic HLA-DR density measurements showed no major differences between the groups, while CD4(+) T cells tended towards two opposed mechanisms of negative immune cell regulation via PD-1 and BTLA. Moreover, the post-sepsis group showed alterations in monocyte surface expression of distinct pattern recognition receptors; most pronouncedly seen in a decrease of TLR5 expression. Cytokine secretion in response to important activators of both the innate (LPS, zymosan) and the adaptive immune system (α-CD3/28) seemed to be weakened in former septic patients.

Conclusions: Cytokine secretion as a reaction to different activators of the immune system seemed to be comprehensively impaired in survivors of sepsis. Among others, this could be based on trends in the downregulation of distinct cell surface receptors. Based on our results, the conduct of larger validation studies seems feasible, aiming to characterize alterations and to find potential therapeutic targets to engage.

Keywords: Immune system; Immunocompromised; Immunology; Sepsis.

Figures

Fig. 1
Fig. 1
Characterization of immune cells subsets. Peripheral blood was collected from eight survivors of sepsis and matched controls and analyzed by flow cytometry to determine the percentage of circulating CD4+ and CD8+ subsets of CD3+ T cells, the amount of regulatory T cells (CD25+ CD127- Treg) as a fraction of all CD4+ cells, and CD14+ monocytes of all leucocytes. The markers used are described in “Methods”. Each data point represents an individual patient. Horizontal lines indicate median values
Fig. 2
Fig. 2
Determination of the pattern of expression of specific receptors on CD4+ and CD8+ T cells. Peripheral blood was collected from eight survivors of sepsis and matched controls and stained for the indicated markers, followed by determination of percent positive relative to isotype control staining (bottom line) and quantification of fluorescence intensity by molecules of equivalent soluble fluorochrome (MESF) calculation (upper line) as described in “Methods”. Each data point represents an individual patient. Horizontal lines indicate median values. PD programmed cell death, BTLA B- and T-lymphocyte attenuator
Fig. 3
Fig. 3
Determination of expression pattern of specific receptors on monocytes. Peripheral blood was collected from eight survivors of sepsis and matched controls and stained for the indicated markers, followed by determination of percent positive relative to isotype control staining (bottom line) and quantification of fluorescence intensity by molecules of equivalent soluble fluorochrome (MESF) calculation (upper line) as described in “Methods”. Each data point represents an individual patient. Horizontal lines indicate median values. TLR toll-like receptor, PD-1 L programmed cell death ligand
Fig. 4
Fig. 4
Determination of human leukocyte antigen (HLA)-DR expression on monocytes. Peripheral blood was collected from eight survivors of sepsis and matched controls and HLA-DR expression was measured. Each data point represents an individual patient. Horizontal lines indicate median values
Fig. 5
Fig. 5
Cytokine secretion of ex-vivo-stimulated whole blood. Peripheral blood was collected from eight survivors of sepsis (S) and matched controls (C). Whole blood was stimulated with either CD3/28 antibody (α-CD3/28), lipopolysaccharide (LPS) or Zymosan. Supernatants were harvested after 24 h and TNF-α, interferon-γ (IFN-γ), and IL-6 and IL-10 were measured by enzyme-linked immunosorbent assay. Each data point represents an individual patient. Horizontal lines indicate median values. CD cluster of differentiation

References

    1. Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR. Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med. 2001;29:1303–10. doi: 10.1097/00003246-200107000-00002.
    1. Vincent J-L, Sakr Y, Sprung CL, Ranieri VM, Reinhart K, Gerlach H, et al. Sepsis in European intensive care units: results of the SOAP study. Crit Care Med. 2006;34:344–53. doi: 10.1097/01.CCM.0000194725.48928.3A.
    1. Engel C, Brunkhorst FM, Bone H-G, Brunkhorst R, Gerlach H, Grond S, et al. Epidemiology of sepsis in Germany: results from a national prospective multicenter study. Intensive Care Med. 2007;33:606–18. doi: 10.1007/s00134-006-0517-7.
    1. Bone RC, Balk RA, Cerra FB, Dellinger RP, Fein AM, Knaus WA, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest. 1992;101:1644–55. doi: 10.1378/chest.101.6.1644.
    1. Bone RC, Grodzin CJ, Balk RA. Sepsis: a new hypothesis for pathogenesis of the disease process. Chest. 1997;112:235–43. doi: 10.1378/chest.112.1.235.
    1. Bone RC. Toward a theory regarding the pathogenesis of the systemic inflammatory response syndrome: what we do and do not know about cytokine regulation. Crit Care Med. 1996;24:163–72. doi: 10.1097/00003246-199601000-00026.
    1. Muenzer JT, Davis CG, Chang K, Schmidt RE, Dunne WM, Coopersmith CM, et al. Characterization and modulation of the immunosuppressive phase of sepsis. Infect Immun. 2010;78:1582–92. doi: 10.1128/IAI.01213-09.
    1. Hotchkiss RS, Nicholson DW. Apoptosis and caspases regulate death and inflammation in sepsis. Nat Rev Immunol. 2006;6:813–22. doi: 10.1038/nri1943.
    1. Monneret G, Debard A-L, Venet F, Bohe J, Hequet O, Bienvenu J, et al. Marked elevation of human circulating CD4 + CD25+ regulatory T cells in sepsis-induced immunoparalysis. Crit Care Med. 2003;31:2068–71. doi: 10.1097/01.CCM.0000069345.78884.0F.
    1. Boomer JS, Green JM, Hotchkiss RS. The changing immune system in sepsis: is individualized immuno-modulatory therapy the answer? Virulence. 2014;5:45–56. doi: 10.4161/viru.26516.
    1. Otto GP, Sossdorf M, Claus RA, Rödel J, Menge K, Reinhart K, et al. The late phase of sepsis is characterized by an increased microbiological burden and death rate. Crit Care. 2011;15:R183. doi: 10.1186/cc10332.
    1. Monneret G, Venet F, Kullberg B-J, Netea MG. ICU-acquired immunosuppression and the risk for secondary fungal infections. Med Mycol. 2011;49(Suppl 1):S17–23. doi: 10.3109/13693786.2010.509744.
    1. Brenner T, Rosenhagen C, Hornig I, Schmidt K, Lichtenstern C, Mieth M, et al. Viral infections in septic shock (VISS-trial)-crosslinks between inflammation and immunosuppression. J Surg Res. 2012;176:571–82. doi: 10.1016/j.jss.2011.10.020.
    1. Dellinger RP, Levy MM, Rhodes A, Annane D, Gerlach H, Opal SM, et al. Surviving Sepsis Campaign: International Guidelines for Management of Severe Sepsis and Septic Shock: 2012. Crit Care Med. 2013;41:580–637. doi: 10.1097/CCM.0b013e31827e83af.
    1. Boomer JS, To K, Chang KC, Takasu O, Osborne DF, Walton AH, et al. Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA. 2011;306:2594–605. doi: 10.1001/jama.2011.1829.
    1. Gentile LF, Cuenca AG, Efron PA, Ang D, Bihorac A, McKinley BA, et al. Persistent inflammation and immunosuppression: a common syndrome and new horizon for surgical intensive care. J Trauma Acute Care Surg. 2012;72:1491–501. doi: 10.1097/TA.0b013e318256e000.
    1. 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;38:1276–83. doi: 10.1097/CCM.0b013e3181d8cc1d.
    1. 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;277:1058–63. doi: 10.1001/jama.1997.03540370048035.
    1. Yende S, Angus DC. Long-term outcomes from sepsis. Curr Infect Dis Rep. 2007;9:382–6. doi: 10.1007/s11908-007-0059-3.
    1. Karlsson S, Ruokonen E, Varpula T, Ala-Kokko TI, Pettilä V. Long-term outcome and quality-adjusted life years after severe sepsis. Crit Care Med. 2009;37:1268–74. doi: 10.1097/CCM.0b013e31819c13ac.
    1. Weycker D, Akhras KS, Edelsberg J, Angus DC, Oster G. Long-term mortality and medical care charges in patients with severe sepsis. Crit Care Med. 2003;31:2316–23. doi: 10.1097/01.CCM.0000085178.80226.0B.
    1. Carson WF, Cavassani KA, Dou Y, Kunkel SL. Epigenetic regulation of immune cell functions during post-septic immunosuppression. Epigenetics. 2011;6:273–83. doi: 10.4161/epi.6.3.14017.
    1. Markwart R, Condotta SA, Requardt RP, Borken F, Schubert K, Weigel C, et al. Immunosuppression after sepsis: systemic inflammation and sepsis induce a loss of naïve T-cells but no enduring cell-autonomous defects in T-cell function. PLoS One. 2014;9:e115094. doi: 10.1371/journal.pone.0115094.
    1. Condotta SA, Khan SH, Rai D, Griffith TS, Badovinac VP. Polymicrobial Sepsis Increases Susceptibility to Chronic Viral Infection and Exacerbates CD8+ T Cell Exhaustion. J Immunol. 2015;195:116–25. doi: 10.4049/jimmunol.1402473.
    1. Weiterer S, Uhle F, Lichtenstern C, Siegler BH, Bhuju S, Jarek M, et al. Sepsis induces specific changes in histone modification patterns in human monocytes. PLoS One. 2015;10:e0121748. doi: 10.1371/journal.pone.0121748.
    1. Anderson ST, O’Callaghan EK, Commins S, Coogan AN. Does prior sepsis alter subsequent circadian and sickness behaviour response to lipopolysaccharide treatment in mice? J Neural Transm. 2015;122(Suppl):S63–73. doi: 10.1007/s00702-013-1124-8.
    1. Anderson ST, Commins S, Moynagh PN, Coogan AN. Lipopolysaccharide-induced sepsis induces long-lasting affective changes in the mouse. Brain Behav Immun. 2015;43:98–109. doi: 10.1016/j.bbi.2014.07.007.
    1. Benjamim CF, Hogaboam CM, Lukacs NW, Kunkel SL. Septic mice are susceptible to pulmonary aspergillosis. Am J Pathol. 2003;163:2605–17. doi: 10.1016/S0002-9440(10)63615-2.
    1. Deng JC. Sepsis-induced suppression of lung innate immunity is mediated by IRAK-M. J Clin Invest. 2006;116:2532–42.
    1. Davis CG, Chang K, Osborne D, Walton AH, Dunne WM, Muenzer JT. Increased susceptibility to Candida infection following cecal ligation and puncture. Biochem Biophys Res Commun. 2011;414:37–43. doi: 10.1016/j.bbrc.2011.09.017.
    1. Boomer JS, Shuherk-Shaffer J, Hotchkiss RS, Green JM. A prospective analysis of lymphocyte phenotype and function over the course of acute sepsis. Crit Care. 2012;16:R112. doi: 10.1186/cc11404.
    1. Zhang Y, Li J, Lou J, Zhou Y, Bo L, Zhu J, et al. Upregulation of programmed death-1 on T cells and programmed death ligand-1 on monocytes in septic shock patients. Crit Care. 2011;15:R70. doi: 10.1186/cc10059.
    1. Chang KC, Burnham C-A, Compton SM, Rasche DP, Mazuski R, McDonough JS, et al. Blockade of the negative co-stimulatory molecules PD-1 and CTLA-4 improves survival in primary and secondary fungal sepsis. Crit Care. 2013;17:R85. doi: 10.1186/cc12711.
    1. Monneret G, Venet F, Pachot A, Lepape A. Monitoring immune dysfunctions in the septic patient: a new skin for the old ceremony. Mol Med. 2008;14:64–78. doi: 10.2119/2007-00102.Monneret.
    1. Monneret G, Lepape A, Voirin N, Bohé J, Venet F, Debard A-L, et al. Persisting low monocyte human leukocyte antigen-DR expression predicts mortality in septic shock. Intensive Care Med. 2006;32:1175–83. doi: 10.1007/s00134-006-0204-8.
    1. Cheron A, Floccard B, Allaouchiche B, Guignant C, Poitevin F, Malcus C, et al. Lack of recovery in monocyte human leukocyte antigen-DR expression is independently associated with the development of sepsis after major trauma. Crit Care. 2010;14:R208. doi: 10.1186/cc9331.
    1. Landelle C, Lepape A, Voirin N, Tognet E, Venet F, Bohé J, et al. Low monocyte human leukocyte antigen-DR is independently associated with nosocomial infections after septic shock. Intensive Care Med. 2010;36:1859–66. doi: 10.1007/s00134-010-1962-x.
    1. Lukaszewicz A-C, Grienay M, Resche-Rigon M, Pirracchio R, Faivre V, Boval B, et al. Monocytic HLA-DR expression in intensive care patients: interest for prognosis and secondary infection prediction. Crit Care Med. 2009;37:2746–52. doi: 10.1097/CCM.0b013e3181ab858a.
    1. Demaret J, Walencik A, Jacob M-C, Timsit J-F, Venet F, Lepape A, et al. Inter-laboratory assessment of flow cytometric monocyte HLA-DR expression in clinical samples. Cytometry B Clin Cytom. 2013;84:59–62. doi: 10.1002/cyto.b.21043.

Source: PubMed

Подписаться