Immune suppression is associated with enhanced systemic inflammatory, endothelial and procoagulant responses in critically ill patients

Xanthe Brands, Fabrice Uhel, Lonneke A van Vught, Maryse A Wiewel, Arie J Hoogendijk, René Lutter, Marcus J Schultz, Brendon P Scicluna, Tom van der Poll, Xanthe Brands, Fabrice Uhel, Lonneke A van Vught, Maryse A Wiewel, Arie J Hoogendijk, René Lutter, Marcus J Schultz, Brendon P Scicluna, Tom van der Poll

Abstract

Objective: Patients admitted to the Intensive Care Unit (ICU) oftentimes show immunological signs of immune suppression. Consequently, immune stimulatory agents have been proposed as an adjunctive therapy approach in the ICU. The objective of this study was to determine the relationship between the degree of immune suppression and systemic inflammation in patients shortly after admission to the ICU. Design: An observational study in two ICUs in the Netherlands.

Methods: The capacity of blood leukocytes to produce cytokines upon stimulation with lipopolysaccharide (LPS) was measured in 77 patients on the first morning after ICU admission. Patients were divided in four groups based on quartiles of LPS stimulated tumor necrosis factor (TNF)-α release, reflecting increasing extents of immune suppression. 15 host response biomarkers indicative of aberrations in inflammatory pathways implicated in sepsis pathogenesis were measured in plasma.

Results: A diminished capacity of blood leukocytes to produce TNF-α upon stimulation with LPS was accompanied by a correspondingly reduced ability to release of IL-1β and IL-6. Concurrently measured plasma concentrations of host response biomarkers demonstrated that the degree of reduction in TNF-α release by blood leukocytes was associated with increasing systemic inflammation, stronger endothelial cell activation, loss of endothelial barrier integrity and enhanced procoagulant responses.

Conclusions: In patients admitted to the ICU the strongest immune suppression occurs in those who simultaneously display signs of stronger systemic inflammation. These findings may have relevance for the selection of patients eligible for administration of immune enhancing agents.

Trial registration: ClinicalTrials.gov identifier NCT01905033.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Whole-blood leukocyte responsiveness to LPS…
Fig 1. Whole-blood leukocyte responsiveness to LPS in critically ill patients.
(A) LPS-induced whole blood leukocyte cytokine production in critically ill patients on the first day after admission (n = 77) stratified according to quartiles of TNF-α production capacity (normal, slightly reduced, moderately reduced, and strongly reduced), and in age and sex-matched healthy controls (n = 19). Dotted lines indicate median cytokine concentrations in unstimulated control samples. Data are expressed as box and whisker diagrams as specified by Tukey. HV, healthy volunteers; ICU, critically ill patients. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. Dot plots depicting the relationship between LPS-induced TNF-α and (B) IL-1β, and (C) IL-6 whole blood production capacity in critically ill patients. Rho, Spearman’s correlation coefficient.
Fig 2. Biomarkers of systemic inflammatory responses…
Fig 2. Biomarkers of systemic inflammatory responses in critically ill patients stratified according to whole blood TNF-α production capacity.
Data are presented as box and whiskers, as specified by Tukey. Dotted lines represent median values obtained in age-matched healthy subjects. Comparisons between groups were performed using the Kruskall-Wallis test followed by Dunn’s post-tests adjusted for multiple comparisons (Bonferroni). * P

Fig 3. Endothelial cell activation biomarkers in…

Fig 3. Endothelial cell activation biomarkers in critically ill patients stratified according to whole blood…

Fig 3. Endothelial cell activation biomarkers in critically ill patients stratified according to whole blood TNF-α production capacity.
Data are presented as box and whiskers, as specified by Tukey. Dotted lines represent median values obtained in age-matched healthy subjects. Comparisons between groups were performed using the Kruskall-Wallis test followed by Dunn’s post-tests adjusted for multiple comparisons (Bonferroni). ** P

Fig 4. Coagulation activation biomarkers in critically…

Fig 4. Coagulation activation biomarkers in critically ill patients stratified according to whole blood TNF-α…

Fig 4. Coagulation activation biomarkers in critically ill patients stratified according to whole blood TNF-α production capacity.
Dotted lines represent median values obtained in age-matched healthy subjects. Comparisons between groups were performed using the Kruskall-Wallis test followed by Dunn’s post-tests adjusted for multiple comparisons (Bonferroni). * P
Similar articles
References
    1. Deutschman CS, Tracey KJ. Sepsis: Current Dogma and New Perspectives. Immunity. 2014;40:463–75. doi: 10.1016/j.immuni.2014.04.001 - DOI - PubMed
    1. van der Poll T, van de Veerdonk FL, Scicluna BP, Netea MG. The immunopathology of sepsis and potential therapeutic targets. Nat Rev Immunol. 2017;17:407–20. doi: 10.1038/nri.2017.36 - DOI - PubMed
    1. Huber-Lang M, Lambris JD, Ward PA. Innate immune responses to trauma. Nat Immunol. 2018;19:327–41. doi: 10.1038/s41590-018-0064-8 - DOI - PMC - PubMed
    1. Biswas SK, Lopez-Collazo E. Endotoxin tolerance: new mechanisms, molecules and clinical significance. Trends Immunol. 2009;30:475–87. doi: 10.1016/j.it.2009.07.009 - DOI - PubMed
    1. Ward NS, Casserly B, Ayala A. The Compensatory Anti-inflammatory Response Syndrome (CARS) in Critically Ill Patients. Clin Chest Med. 2008;29:617–25. doi: 10.1016/j.ccm.2008.06.010 - DOI - PMC - PubMed
Show all 36 references
Publication types
Associated data
Grant support
This research was performed within the framework of the Center for Translational Molecular Medicine (CTMM) (www.ctmm.nl), project Molecular Diagnosis and Risk Stratification of Sepsis (grant 04I-201). The sponsor CTMM was not involved in the design and conduction of the study; nor was the sponsor involved in collection, management, analysis, and interpretation of the data or preparation, review or approval of the article. Decision to submit the article was not dependent on the sponsor. X.B. was supported by a grant from the Netherlands Organization for Health Research and Development (ZonMW #50-53000-98-139).
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Fig 3. Endothelial cell activation biomarkers in…
Fig 3. Endothelial cell activation biomarkers in critically ill patients stratified according to whole blood TNF-α production capacity.
Data are presented as box and whiskers, as specified by Tukey. Dotted lines represent median values obtained in age-matched healthy subjects. Comparisons between groups were performed using the Kruskall-Wallis test followed by Dunn’s post-tests adjusted for multiple comparisons (Bonferroni). ** P

Fig 4. Coagulation activation biomarkers in critically…

Fig 4. Coagulation activation biomarkers in critically ill patients stratified according to whole blood TNF-α…

Fig 4. Coagulation activation biomarkers in critically ill patients stratified according to whole blood TNF-α production capacity.
Dotted lines represent median values obtained in age-matched healthy subjects. Comparisons between groups were performed using the Kruskall-Wallis test followed by Dunn’s post-tests adjusted for multiple comparisons (Bonferroni). * P
Similar articles
References
    1. Deutschman CS, Tracey KJ. Sepsis: Current Dogma and New Perspectives. Immunity. 2014;40:463–75. doi: 10.1016/j.immuni.2014.04.001 - DOI - PubMed
    1. van der Poll T, van de Veerdonk FL, Scicluna BP, Netea MG. The immunopathology of sepsis and potential therapeutic targets. Nat Rev Immunol. 2017;17:407–20. doi: 10.1038/nri.2017.36 - DOI - PubMed
    1. Huber-Lang M, Lambris JD, Ward PA. Innate immune responses to trauma. Nat Immunol. 2018;19:327–41. doi: 10.1038/s41590-018-0064-8 - DOI - PMC - PubMed
    1. Biswas SK, Lopez-Collazo E. Endotoxin tolerance: new mechanisms, molecules and clinical significance. Trends Immunol. 2009;30:475–87. doi: 10.1016/j.it.2009.07.009 - DOI - PubMed
    1. Ward NS, Casserly B, Ayala A. The Compensatory Anti-inflammatory Response Syndrome (CARS) in Critically Ill Patients. Clin Chest Med. 2008;29:617–25. doi: 10.1016/j.ccm.2008.06.010 - DOI - PMC - PubMed
Show all 36 references
Publication types
Associated data
Grant support
This research was performed within the framework of the Center for Translational Molecular Medicine (CTMM) (www.ctmm.nl), project Molecular Diagnosis and Risk Stratification of Sepsis (grant 04I-201). The sponsor CTMM was not involved in the design and conduction of the study; nor was the sponsor involved in collection, management, analysis, and interpretation of the data or preparation, review or approval of the article. Decision to submit the article was not dependent on the sponsor. X.B. was supported by a grant from the Netherlands Organization for Health Research and Development (ZonMW #50-53000-98-139).
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Fig 4. Coagulation activation biomarkers in critically…
Fig 4. Coagulation activation biomarkers in critically ill patients stratified according to whole blood TNF-α production capacity.
Dotted lines represent median values obtained in age-matched healthy subjects. Comparisons between groups were performed using the Kruskall-Wallis test followed by Dunn’s post-tests adjusted for multiple comparisons (Bonferroni). * P

References

    1. Deutschman CS, Tracey KJ. Sepsis: Current Dogma and New Perspectives. Immunity. 2014;40:463–75. doi: 10.1016/j.immuni.2014.04.001
    1. van der Poll T, van de Veerdonk FL, Scicluna BP, Netea MG. The immunopathology of sepsis and potential therapeutic targets. Nat Rev Immunol. 2017;17:407–20. doi: 10.1038/nri.2017.36
    1. Huber-Lang M, Lambris JD, Ward PA. Innate immune responses to trauma. Nat Immunol. 2018;19:327–41. doi: 10.1038/s41590-018-0064-8
    1. Biswas SK, Lopez-Collazo E. Endotoxin tolerance: new mechanisms, molecules and clinical significance. Trends Immunol. 2009;30:475–87. doi: 10.1016/j.it.2009.07.009
    1. Ward NS, Casserly B, Ayala A. The Compensatory Anti-inflammatory Response Syndrome (CARS) in Critically Ill Patients. Clin Chest Med. 2008;29:617–25. doi: 10.1016/j.ccm.2008.06.010
    1. Adib-Conquy M, Cavaillon J-M. Compensatory anti-inflammatory response syndrome. Thromb Haemost. 2009;101:36–47.
    1. Marshall JC. Why have clinical trials in sepsis failed? Trends Mol Med. 2014;20:195–203. doi: 10.1016/j.molmed.2014.01.007
    1. Opal SM, Dellinger RP, Vincent J-L, Masur H, Angus DC. The Next Generation of Sepsis Clinical Trial Designs. Crit Care Med. 2014;42:1714–21. doi: 10.1097/CCM.0000000000000325
    1. Peters van Ton AM, Kox M, Abdo WF, Pickkers P. Precision Immunotherapy for Sepsis. Front Immunol. 2018;9. doi: 10.3389/fimmu.2018.01926
    1. Döcke W-D, Randow F, Syrbe U, Krausch D, Asadullah K, Reinke P, et al.. Monocyte deactivation in septic patients: Restoration by IFN-γ treatment. Nat Med. 1997;3:678–81. doi: 10.1038/nm0697-678
    1. Meisel C, Schefold JC, Pschowski R, Baumann T, Hetzger K, Gregor J, et al.. Granulocyte–Macrophage Colony-stimulating Factor to Reverse Sepsis-associated Immunosuppression. Am J Respir Crit Care Med. 2009;180:640–8. doi: 10.1164/rccm.200903-0363OC
    1. Delsing CE, Gresnigt MS, Leentjens J, Preijers F, Frager FA, Kox M, et al.. Interferon-gamma as adjunctive immunotherapy for invasive fungal infections: a case series. BMC Infect Dis. 2014;14:166. doi: 10.1186/1471-2334-14-166
    1. Proceedings of Reanimation 2021, the French Intensive Care Society International Congress. Ann Intensive Care. 2021;11:97. Available from: doi: 10.1186/s13613-021-00862-0
    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. Chest. 1992;101:1644–55. doi: 10.1378/chest.101.6.1644
    1. Klouwenberg PMCK Ong DSY, Bos LDJ, de Beer FM, van Hooijdonk RTM, Huson M A, et al.. Interobserver Agreement of Centers for Disease Control and Prevention Criteria for Classifying Infections in Critically Ill Patients*. Crit Care Med. 2013;41:2373–8. doi: 10.1097/CCM.0b013e3182923712
    1. Levy MM, Fink MP, Marshall JC, Abraham E, Angus D, Cook D, et al.. 2001 SCCM/ESICM/ACCP/ATS/SIS International Sepsis Definitions Conference. Crit Care Med. 2003;31:1250–6. doi: 10.1097/01.CCM.0000050454.01978.3B
    1. Vincent J-L, Moreno R, Takala J, Willatts S, De Mendonça A, Bruining H, et al.. The SOFA (Sepsis-related Organ Failure Assessment) score to describe organ dysfunction/failure. Intensive Care Med. 1996;22:707–10. doi: 10.1007/BF01709751
    1. Zimmerman JE, Kramer AA, McNair DS, Malila FM. Acute Physiology and Chronic Health Evaluation (APACHE) IV: Hospital mortality assessment for today’s critically ill patients*. Crit Care Med. 2006;34:1297–310. doi: 10.1097/01.CCM.0000215112.84523.F0
    1. van Vught LA, Klein Klouwenberg PMC, Spitoni C, Scicluna BP, Wiewel MA, Horn J, et al.. Incidence, Risk Factors, and Attributable Mortality of Secondary Infections in the Intensive Care Unit After Admission for Sepsis. JAMA. 2016;315:1469. doi: 10.1001/jama.2016.2691
    1. Charlson ME, Pompei P, Ales KL, MacKenzie CR. A new method of classifying prognostic comorbidity in longitudinal studies: Development and validation. J Chronic Dis. 1987;40:373–83. doi: 10.1016/0021-9681(87)90171-8
    1. Bellomo R, Ronco C, Kellum JA, Mehta RL, Palevsky P, Acute Dialysis Quality Initiative workgroup. Acute renal failure—definition, outcome measures, animal models, fluid therapy and information technology needs: the Second International Consensus Conference of the Acute Dialysis Quality Initiative (ADQI) Group. Crit Care. 2004;8:R204–12. doi: 10.1186/cc2872
    1. Ranieri V, Rubenfeld GD, Thompson B, Ferguson ND, Caldwell E, Fan E, et al.. Acute Respiratory Distress Syndrome. JAMA. 2012;307.
    1. van Vught LA, Wiewel MA, Hoogendijk AJ, Scicluna BP, Belkasim-Bohoudi H, Horn J, et al.. Reduced Responsiveness of Blood Leukocytes to Lipopolysaccharide Does not Predict Nosocomial Infections in Critically Ill Patients. Shock. 2015;44:110–4. doi: 10.1097/SHK.0000000000000391
    1. Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol. 2013;13:862–74. doi: 10.1038/nri3552
    1. Hawkins RB, Raymond SL, Stortz JA, Horiguchi H, Brakenridge SC, Gardner A, et al.. Chronic Critical Illness and the Persistent Inflammation, Immunosuppression, and Catabolism Syndrome. Front Immunol. 2018;9.
    1. Winkler MS, Rissiek A, Priefler M, Schwedhelm E, Robbe L, Bauer A, et al.. Human leucocyte antigen (HLA-DR) gene expression is reduced in sepsis and correlates with impaired TNFα response: A diagnostic tool for immunosuppression? Infante-Duarte C, editor. PLoS One. 2017;12:e0182427. doi: 10.1371/journal.pone.0182427
    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. Flohé S, Lendemans S, Selbach C, Waydhas C, Ackermann M, Schade FU, et al.. Effect of granulocyte-macrophage colony-stimulating factor on the immune response of circulating monocytes after severe trauma. Crit Care Med. 2003;31:2462–9. doi: 10.1097/01.CCM.0000089640.17523.57
    1. Galbraith NJ, Gardner SA, Walker SP, Trainor P, Carter J V., Bishop C, et al.. The role and function of IκKα/β in monocyte impairment. Sci Rep. 2020;10:12222. doi: 10.1038/s41598-020-68018-x
    1. Hoogendijk AJ, Garcia-Laorden MI, van Vught LA, Wiewel MA, Belkasim-Bohoudi H, Duitman J, et al.. Sepsis Patients Display a Reduced Capacity to Activate Nuclear Factor-κB in Multiple Cell Types*. Crit Care Med. 2017;45:e524–31. doi: 10.1097/CCM.0000000000002294
    1. Munoz C, Carlet J, Fitting C, Misset B, Blériot JP, Cavaillon JM. Dysregulation of in vitro cytokine production by monocytes during sepsis. J Clin Invest. 1991;88:1747–54. doi: 10.1172/JCI115493
    1. Santos SS, Carmo AM, Brunialti MKC, Machado FR, Azevedo LC, Assunção M, et al.. Modulation of monocytes in septic patients: preserved phagocytic activity, increased ROS and NO generation, and decreased production of inflammatory cytokines. Intensive Care Med Exp. 2016;4:5. doi: 10.1186/s40635-016-0078-1
    1. van Vught LA, Scicluna BP, Wiewel MA, Hoogendijk AJ, Klein Klouwenberg PMC, Franitza M, et al.. Comparative Analysis of the Host Response to Community-acquired and Hospital-acquired Pneumonia in Critically Ill Patients. Am J Respir Crit Care Med. 2016;194:1366–74. doi: 10.1164/rccm.201602-0368OC
    1. Opal SM, van der Poll T. Endothelial barrier dysfunction in septic shock. J Intern Med. 2015;277:277–93. doi: 10.1111/joim.12331
    1. Levi M, van der Poll T. Coagulation and sepsis. Thromb Res. 2017;149:38–44. doi: 10.1016/j.thromres.2016.11.007
    1. Brands X, Haak BW, Klarenbeek AM, Otto NA, Faber DR, Lutter R, et al.. Concurrent Immune Suppression and Hyperinflammation in Patients With Community-Acquired Pneumonia. Front Immunol. 2020;11.

Source: PubMed

3
Předplatit