Imbalance of Circulating Innate Lymphoid Cell Subpopulations in Patients With Septic Shock

Julien Carvelli, Christelle Piperoglou, Jeremy Bourenne, Catherine Farnarier, Nathalie Banzet, Clemence Demerlé, Marc Gainnier, Frédéric Vély, Julien Carvelli, Christelle Piperoglou, Jeremy Bourenne, Catherine Farnarier, Nathalie Banzet, Clemence Demerlé, Marc Gainnier, Frédéric Vély

Abstract

Background: Septic shock, a major cause of death in critical care, is the clinical translation of a cytokine storm in response to infection. It can be complicated by sepsis-induced immunosuppression, exemplified by blood lymphopenia, an excess of circulating Treg lymphocytes, and decreased HLA-DR expression on circulating monocytes. Such immunosuppression is associated with secondary infections, and higher mortality. The effect of these biological modifications on circulating innate lymphoid cells (ILCs) has been little studied. Methods: We prospectively enrolled patients with septic shock (Sepsis-3 definition) in the intensive care unit (ICU) of Timone CHU Hospital. ICU controls (trauma, cardiac arrest, neurological dysfunction) were recruited at the same time (NCT03297203). We performed immunophenotyping of adaptive lymphocytes (CD3+ T cells, CD19+ B cells, CD4+CD25+FoxP3+ Treg lymphocytes), ILCs (CD3-CD56+ NK cells and helper ILCs - ILC1, ILC2, and ILC3), and monocytes by flow cytometry on fresh blood samples collected between 24 and 72 h after admission. Results: We investigated adaptive and innate circulating lymphoid cells in the peripheral blood of 18 patients in septic shock, 15 ICU controls, and 30 healthy subjects. As expected, the peripheral blood lymphocytes of all ICU patients showed lymphopenia, which was not specific to sepsis, whereas those of the healthy volunteers did not. Circulating CD3+ T cells and CD3-CD56+ NK cells were mainly concerned. There was a tendency toward fewer Treg lymphocytes and lower HLA-DR expression on monocytes in ICU patients with sepsis. Although the ILC1 count was higher in septic patients than healthy subjects, ILC2, and ILC3 counts were lower in both ICU groups. However, ILC3s within the total ILCs were overrepresented in patients with septic shock. The depression of immune responses has been correlated with the occurrence of secondary infections. We did not find any differences in ILC distribution according to this criterion. Conclusion: All ICU patients exhibit lymphopenia, regardless of the nature (septic or sterile) of the initial medical condition. Specific distribution of circulating ILCs, with an excess of ILC1, and a lack of ILC3, may characterize septic shock during the first 3 days of the disease.

Keywords: NK cells; immunosuppression; innate lymphoid cells (ILC); lymphopenia; sepsis; sepsis-induced immunosuppression; septic shock.

Copyright © 2019 Carvelli, Piperoglou, Bourenne, Farnarier, Banzet, Demerlé, Gainnier and Vély.

Figures

Figure 1
Figure 1
Phenotypic analysis of circulating lymphocyte subsets in ICU patients. (A) Comparison of helper T-cell, cytotoxic T-cell, B-cell, and NK-cell counts (cells/μl) of septic patients (red circles) with those of healthy controls (dark circles), and ICU patients without sepsis (blue circles). (B) Comparison of helper T-cell, cytotoxic T-cell, B-cell, and NK cell counts (cells/μl) of patients with severe tissue injuries (green circles) with those of healthy controls (dark circles), and patients with less severe lesions (orange circles). The bars show the median. Statistical analyses were performed using the Mann Whitney U-test. Differences were considered significant when P < 0.05: *P < 0.05, ***P < 0.001, ****P < 0.0001. ns: not significant.
Figure 2
Figure 2
Phenotypic analysis of circulating Tregs and monocytes in ICU patients. (A, upper) Comparison of Treg cell counts (cells/μl) of septic patients (red circles) with those of ICU patients without sepsis (blue circles). (A, lower) Comparison of Treg cell counts (cells/μl) of patients with severe tissue injuries (green circles) with those of patients with less severe lesions (orange circles). (B, upper) Comparison of the MFI of HLA-DR expression and the number of HLA-DR molecules on monocytes of septic patients (red circles) with those of ICU patients without sepsis (blue circles). (B, lower) Comparison of the MFI of HLA-DR expression and the number of HLA-DR molecules on monocytes of patients with severe tissue injuries (green circles) with those of patients with less severe lesions (orange circles). The bars show the median. Statistical analyses were performed using the Mann Whitney U-test. Differences were considered significant when P < 0.05: *P < 0.05. ns: not significant.
Figure 3
Figure 3
Flow cytometry gating strategy for the identification of human peripheral blood ILCs. ILCs are defined as Lin−CD127+ cells with a lineage cocktail containing antibodies directed against CD3, CD19, CD14, TCRαβ, TCRγδ, CD94, CD16, FcεRI, CD34, CD123, and CD303 (Left). Within the ILC gate, ILC2s are CD294+ cells, whereas ILC1s are CD294−CD117− cells and ILC3s are CD294−CD117+ cells (Right).
Figure 4
Figure 4
Phenotypic analysis of circulating ILCs in ICU patients. (A, upper) Comparison of total ILC, ILC1, ILC2, and ILC3 counts (cells/ml) of septic patients (red circles) with those of healthy controls (dark circles) and ICU patients without sepsis (blue circles). (A, lower) Distribution of each ILC subset among total ILCs. (B, upper) Comparison of total ILC, ILC1, ILC2, and ILC3 counts (cells/ml) of patients with severe tissue injuries (green circles) with those of healthy controls (dark circles) and patients with less severe lesions (orange circles). (B, lower) Distribution of each ILC subset among total ILCs. The bars show the median. Statistical analyses were performed using the Mann Whitney U-test. Differences were considered significant when P < 0.05: *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. ns: not significant.

References

    1. Besen BA, Romano TG, Nassar AP, Jr, Taniguchi LU, Azevedo LC, Mendes PV, et al. . Sepsis-3 definitions predict ICU mortality in a low-middle-income country. Ann Intens Care. (2016) 6:107. 10.1186/s13613-016-0204-y
    1. Mira JC, Gentile LF, Mathias BJ, Efron PA, Brakenridge SC, Mohr AM, et al. . Sepsis pathophysiology, chronic critical illness, and persistent inflammation-immunosuppression and catabolism syndrome. Crit Care Med. (2017) 45:253–62. 10.1097/CCM.0000000000002074
    1. Pelekanou A, Tsangaris I, Kotsaki A, Karagianni V, Giamarellou H, Armaganidis A, et al. . Decrease of CD4-lymphocytes and apoptosis of CD14-monocytes are characteristic alterations in sepsis caused by ventilator-associated pneumonia: results from an observational study. Crit Care. (2009) 13:R172. 10.1186/cc8148
    1. Conway Morris A, Kefala K, Wilkinson TS, Dhaliwal K, Farrell L, Walsh T, et al. . C5a mediates peripheral blood neutrophil dysfunction in critically ill patients. Am J Respir Crit Care Med. (2009)180:19–28. 10.1164/rccm.200812-1928OC
    1. Chiche L, Forel JM, Thomas G, Farnarier C, Cognet C, Guervilly C, et al. . Interferon-γ production by natural killer cells and cytomegalovirus in critically ill patients. Crit Care Med. (2012) 40:3162–9. 10.1097/CCM.0b013e318260c90e
    1. Drewry AM, Ablordeppey EA, Murray ET, Beiter ER, Walton AH, Hall MW, 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. 10.1186/s13054-016-1505-0
    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. Intens Care Med. (2010) 36:1859–66. 10.1007/s00134-010-1962-x
    1. Hotchkiss RS, Tinsley KW, Swanson PE, Schmieg RE, Jr, Hui JJ, Chang KC, et al. . Sepsis-induced apoptosis causes progressive profound depletion of B and CD4+ T lymphocytes in humans. J Immunol. (2001) 166:6952–63. 10.4049/jimmunol.166.11.6952
    1. Venet F, Chung CS, Kherouf H, Geeraert A, Malcus C, Poitevin F, et al. . Increased circulating regulatory T cells (CD4(+)CD25 (+)CD127 (-)) contribute to lymphocyte anergy in septic shock patients. Intensive Care Med. (2009) 35:678–86. 10.1007/s00134-008-1337-8
    1. Faivre V, Lukaszewicz AC, Alves A, Charron D, Payen D, Haziot A. Human monocytes differentiate into dendritic cells subsets that induce anergic and regulatory T cells in sepsis. PLoS ONE. (2012) 7:e47209. 10.1371/journal.pone.0047209
    1. Spits H, Di Santo JP. The expanding family of innate lymphoid cells: regulators and effectors of immunity and tissue remodeling. Nat Immunol. (2011) 12:21–7. 10.1038/ni.1962
    1. Vivier E, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, et al. . Innate lymphoid cells: 10 years on. Cell. (2018) 174:1054–66. 10.1016/j.cell.2018.07.017
    1. Vély F, Barlogis V, Vallentin B, Neven B, Piperoglou C, Perchet T, et al. Evidence of innate lymphoid cell redundancy in humans. Nat Immunol. (2016) 17:1291–9. 10.1038/ni.3553
    1. Gasteiger G, Fan X, Dikiy S, Lee SY, Rudensky AY. Tissue residency of innate lymphoid cells in lymphoid and nonlymphoid organs. Science. (2015) 350:981–5. 10.1126/science.aac9593
    1. Ebbo M, Crinier A, Vély F, Vivier E. Innate lymphoid cells: major players in inflammatory diseases. Nat Rev Immunol. (2017) 17:665–78. 10.1038/nri.2017.86
    1. Silver JS, Kearley J, Copenhaver AM, Sanden C, Mori M, Yu L, et al. Inflammatory triggers associated with exacerbations of COPD orchestrate plasticity of group 2 innate lymphoid cells in the lungs. Nat Immunol. (2016) 17:626–35. 10.1038/ni.3443
    1. Bernink JH, Peters CP, Munneke M, te Velde AA, Meijer SL, Weijer K, et al. . Human type 1 innate lymphoid cells accumulate in inflamed mucosal tissues. Nat Immunol. (2013) 14:221–9. 10.1038/ni.2534
    1. Smith SG, Chen R, Kjarsgaard M, Huang C, Oliveria JP, O'Byrne PM, et al. . Increased numbers of activated group 2 innate lymphoid cells in the airways of patients with severe asthma and persistent airway eosinophilia. J Allergy Clin Immunol. (2016) 137:75–86.e8. 10.1016/j.jaci.2015.05.037
    1. Wohlfahrt T, Usherenko S, Englbrecht M, Dees C, Weber S, Beyer C, et al. . Type 2 innate lymphoid cell counts are increased in patients with systemic sclerosis and correlate with the extent of fibrosis. Ann Rheum Dis. (2016) 75:623–6. 10.1136/annrheumdis-2015-207388
    1. Teunissen MBM, Munneke JM, Bernink JH, Spuls PI, Res PCM, Te Velde A, et al. . Composition of innate lymphoid cell subsets in the human skin: enrichment of NCR(+) ILC3 in lesional skin and blood of psoriasis patients. J Invest Dermatol. (2014) 134:2351–60. 10.1038/jid.2014.146
    1. Shankar-Hari M, Phillips GS, Levy ML, Seymour CW, Liu VX, Deutschman CS, et al. . Developing a new definition and assessing new clinical criteria for septic shock: for the third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA. (2016) 315:775–87. 10.1001/jama.2016.0289
    1. Annane D, Renault A, Brun-Buisson C, Megarbane B, Quenot JP, Siami S, et al. . Hydrocortisone plus fludrocortisone for adults with septic shock. N Engl J Med. (2018) 378:809–18. 10.1056/NEJMoa1705716
    1. Venkatesh B, Finfer S, Cohen J, Rajbhandari D, Arabi Y, Bellomo R, et al. . Adjunctive glucocorticoid therapy in patients with septic shock. N Engl J Med. (2018) 378:797–808. 10.1056/NEJMoa1705835
    1. Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med. (2003). 348:138–50. 10.1056/NEJMra021333
    1. Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol. (2013) 13:862–74. 10.1038/nri3552
    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. 10.1001/jama.2011.1829
    1. Nascimento DC, Melo PH, Piñeros AR, Ferreira RG, Colón DF, Donate PB, et al. . IL-33 contributes to sepsis-induced long-term immunosuppression by expanding the regulatory T cell population. Nat Commun. (2017) 8:14919. 10.1038/ncomms14919
    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: a double-blind, randomized, placebo-controlled multicenter trial. Am J Respir Crit Care Med. (2009) 180:640–8. 10.1164/rccm.200903-0363OC
    1. Cazalis MA, Friggeri A, Cavé L, Demaret J, Barbalat V, Cerrato E, et al. . Decreased HLA-DR antigen-associated invariant chain (CD74) mRNA expression predicts mortality after septic shock. Crit Care. (2013) 17:R287. 10.1186/cc13150
    1. Monneret G, Venet F. Monocyte HLA-DR in sepsis: shall we stop following the flow? Crit Care. (2014) 18:102. 10.1186/cc13179
    1. Zorio V, Venet F, Delwarde B, Floccard B, Marcotte G, Textoris J, et al. . Assessment of sepsis-induced immunosuppression at ICU discharge and 6 months after ICU discharge. Ann Intens Care. (2017) 7:80. 10.1186/s13613-017-0304-3
    1. Satoh-Takayama N, Vosshenrich CA, Lesjean-Pottier S, Sawa S, Lochner M, Rattis F, et al. . Microbial flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal immune defense. Immunity. (2008) 29:958–70. 10.1016/j.immuni.2008.11.001
    1. Satoh-Takayama N, Dumoutier L, Lesjean-Pottier S, Ribeiro VS, Mandelboim O, Renauld JC, et al. . The natural cytotoxicity receptor NKp46 is dispensable for IL-22-mediated innate intestinal immunedefense against citrobacter rodentium. J Immunol. (2009) 183:6579–87. 10.4049/jimmunol.0901935
    1. Klose CS, Flach M, Möhle L, Rogell L, Hoyler T, Ebert K, et al. . Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell. (2014) 157:340–56. 10.1016/j.cell.2014.03.030
    1. Abt MC, Lewis BB, Caballero S, Xiong H, Carter RA, Sušac B, et al. . Innate immune defenses mediated by two ILC subsets are critical for protection against acute clostridium difficile infection. Cell Host Microbe. (2015) 18:27–37. 10.1016/j.chom.2015.06.011
    1. Sonnenberg GF, Artis D. Innate lymphoid cells in the initiation, regulation and resolution of inflammation. Nat Med. (2015) 21:698–708. 10.1038/nm.3892
    1. Quatrini L, Wieduwild E, Guia S, Bernat C, Glaichenhaus N, Vivier E, et al. . Host resistance to endotoxic shock requires the neuroendocrine regulation of group 1 innate lymphoid cells. J Exp Med. (2017) 214:3531–41. 10.1084/jem.20171048
    1. Chun TT, Chung CS, Fallon EA, Hutchins NA, Clarke E, Rossi AL, et al. . Group 2 innate lymphoid cells (ILC2s) are key mediators of the inflammatory response in polymicrobial sepsis. Am J Pathol. (2018) 188:2097–108. 10.1016/j.ajpath.2018.05.009
    1. Monticelli LA, Sonnenberg GF, Abt MC, Alenghat T, Ziegler CG, Doering TA, et al. . Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus. Nat Immunol. (2011) 12:1045–54. 10.1038/ni.2131
    1. Hotchkiss RS, Schmieg RE, Jr, Swanson PE, Freeman BD, Tinsley KW, Cobb JP, et al. . Rapid onset of intestinal epithelial and lymphocyte apoptotic cell death in patients with trauma and shock. Crit Care Med. (2000) 28:3207–17. 10.1097/00003246-200009000-00016
    1. Zhang F, Qiu XC, Wang JJ, Hong XD, Wang GY, Xia ZF. Burn-related dysregulation of inflammation and immunity in experimental and clinical studies. J Burn Care Res. (2017) 38:e892–9. 10.1097/BCR.0000000000000511
    1. Sasajima K, Inokuchi K, Onda M, Miyashita M, Okawa KI, Matsutani T, et al. . Detection of T cell apoptosis after major operations. Eur J Surg. (1999) 165:1020–3. 10.1080/110241599750007829
    1. Nishiguchi K, Okuda J, Toyoda M, Tanaka K, Tanigawa N. Comparative evaluation of surgical stress of laparoscopic and open surgeries for colorectal carcinoma. Dis Colon Rectum. (2001) 44:223–30. 10.1007/BF02234297
    1. Edwards MR, Sultan P, del Arroyo AG, Whittle J, Karmali SN, Moonesinghe SR, et al. . Metabolic dysfunction in lymphocytes promotes postoperative morbidity. Clin Sci. (2015) 129:423–37. 10.1042/CS20150024
    1. Chousterman BG, Swirski FK, Weber GF. Cytokine storm and sepsis disease pathogenesis. Semin Immunopathol. (2017) 39:517–28. 10.1007/s00281-017-0639-8
    1. Chen GY, Nuñez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol. (2010) 10:826–37. 10.1038/nri2873
    1. Seeley JJ, Baker RG, Mohamed G, Bruns T, Hayden MS, Deshmukh SD, et al. . Induction of innate immune memory via microRNA targeting of chromatin remodelling factors. Nature. (2018) 559:114–9. 10.1038/s41586-018-0253-5
    1. Hotchkiss RS, Nicholson DW. Apoptosis and caspases regulate death and inflammation in sepsis. Nat Rev Immunol. (2006) 6:813–22. 10.1038/nri1943
    1. Cruz-Zárate D, Cabrera-Rivera GL, Ruiz-Sánchez BP, Serafín-López J, Chacón-Salinas R, López-Macías C, et al. . Innate lymphoid cells have decreased HLA-DR expression but retain their responsiveness to TLR ligands during sepsis. J Immunol. (2018) 201:3401–10. 10.4049/jimmunol.1800735
    1. Monneret G, Lepape A, Voirin N, Bohé J, Venet F, Debard AL, et al. . Persisting low monocyte human leukocyte antigen-DR expression predicts mortality in septic shock. Intensive Care Med. (2006) 32:1175–83. 10.1007/s00134-006-0204-8
    1. Grimaldi D, Louis S, Pène F, Sirgo G, Rousseau C, Claessens YE, et al. . Profound and persistent decrease of circulating dendritic cells is associated with ICU-acquired infection in patients with septic shock. Intensive Care Med. (2011) 37:1438–46. 10.1007/s00134-011-2306-1
    1. van Vught LA, Wiewel MA, Hoogendijk AJ, Frencken JF, Scicluna BP, Klein Klouwenberg PMC, et al. . The host response in patients with sepsis developing intensive care unit-acquired secondary infections. Am J Respir Crit Care Med. (2017) 196:458–70. 10.1164/rccm.201606-1225OC
    1. van Vught LA, Klein Klouwenberg PM, 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) 315:1469–79. 10.1001/jama.2016.2691
    1. Hazenberg MD, Spits H. Human innate lymphoid cells. Blood. (2014) 124:700–9. 10.1182/blood-2013-11-427781
    1. Lim AI, Li Y, Lopez-Lastra S, Stadhouders R, Paul F, Casrouge A, et al. . Systemic human ILC precursors provide a substrate for tissue ILC differentiation. Cell. (2017) 168:1086–100.e10. 10.1016/j.cell.2017.02.021
    1. Lim AI, Di Santo JP. ILC-poiesis: ensuring tissue ILC differentiation at the right place and time. Eur J Immunol. (2019) 49:11–8. 10.1002/eji.201747294
    1. Bonelli M, Shih HY, Hirahara K, Singelton K, Laurence A, Poholek A, et al. . Helper T cell plasticity: impact of extrinsic and intrinsic signals on transcriptomes and epigenomes. Curr Top Microbiol Immunol. (2014) 381:279–326. 10.1007/82_2014_371
    1. Lim AI, Verrier T, Vosshenrich CA, Di Santo JP. Developmental options and functional plasticity of innate lymphoid cells. Curr Opin Immunol. (2017) 44:61–8. 10.1016/j.coi.2017.03.010
    1. Vonarbourg C, Mortha A, Bui VL, Hernandez PP, Kiss EA, Hoyler T, et al. . Regulated expression of nuclear receptor RORγt confers distinct functional fates to NK cell receptor-expressing RORγt(+) innate lymphocytes. Immunity. (2010) 33:736–51. 10.1016/j.immuni.2010.10.017
    1. Huang Q, Niu Z, Tan J, Yang J, Liu Y, Ma H, et al. . IL-25 Elicits innate lymphoid cells and multipotent progenitor type 2 cells that reduce renal ischemic/reperfusion injury. J Am Soc Nephrol. (2015) 26:2199–211. 10.1681/ASN.2014050479
    1. Russi AE, Walker-Caulfield ME, Ebel ME, Brown MA. Cutting edge: c-Kit signaling differentially regulates type 2 innate lymphoid cell accumulation and susceptibility to central nervous system demyelination in male and female SJL mice. J Immunol. (2015)194:5609–13. 10.4049/jimmunol.1500068
    1. Ito I, Bhopale KK, Kobayashi M, Finnerty CC, Herndon DN, Suzuki F. Lamina propria group 2 innate lymphoid cells impair the antibacterial defense of burned mice to enterococcal translocation. J Leukoc Biol. (2017) 102:1451–60. 10.1189/jlb.4A0517-195R
    1. Lai D, Tang J, Chen L, Fan EK, Scott MJ, Li Y, et al. . Group 2 innate lymphoid cells protect lung endothelial cells from pyroptosis in sepsis. Cell Death Dis. (2018) 9:369. 10.1038/s41419-018-0412-5
    1. Artis D, Spits H. The biology of innate lymphoid cells. Nature. (2015) 517:293–301. 10.1038/nature14189

Source: PubMed

3
구독하다