Skip to main content
Top
Published in: Critical Care 1/2018

Open Access 01-12-2018 | Research

Postoperative cellular stress in the kidney is associated with an early systemic γδ T-cell immune cell response

Authors: Ivan Göcze, Katharina Ehehalt, Florian Zeman, Paloma Riquelme, Karin Pfister, Bernhard M. Graf, Thomas Bein, Edward K. Geissler, Piotr Kasprzak, Hans J. Schlitt, John A. Kellum, James A. Hutchinson, Elke Eggenhofer, Philipp Renner

Published in: Critical Care | Issue 1/2018

Login to get access

Abstract

Background

Basic science data suggest that acute kidney injury (AKI) induced by ischemia-reperfusion injury (IRI) is an inflammatory process involving the adaptive immune response. Little is known about the T-cell contribution in the very early phase, so we investigated if tubular cellular stress expressed by elevated cell cycle biomarkers is associated with early changes in circulating T-cell subsets, applying a bedside-to-bench approach.

Methods

Our observational pilot study included 20 consecutive patients undergoing endovascular aortic repair for aortic aneurysms affecting the renal arteries, thereby requiring brief kidney hypoperfusion and reperfusion. Clinical-grade flow cytometry-based immune monitoring of peripheral immune cell populations was conducted perioperatively and linked to tubular cell stress biomarkers ([TIMP-2]•[IGFBP7]) immediately after surgery. To confirm clinical results and prove T-cell infiltration in the kidney, we simulated tubular cellular injury in an established mouse model of mild renal IRI.

Results

A significant correlation between tubular cell injury and a peripheral decline of γδ T cells, but no other T-cell subpopulation, was discovered within the first 24 hours (r = 0.53; p = 0.022). Turning to a mouse model of kidney warm IRI, a similar decrease in circulating γδ T cells was found and concomitantly was associated with a 6.65-fold increase in γδ T cells (p = 0.002) in the kidney tissue without alterations in other T-cell subsets, consistent with our human data. In search of a mechanistic driver of IRI, we found that the damage-associated molecule high-mobility group box 1 protein HMGB1 was significantly elevated in the peripheral blood of clinical study subjects after tubular cell injury (p = 0.019). Correspondingly, HMGB1 RNA content was significantly elevated in the murine kidney.

Conclusions

Our investigation supports a hypothesis that γδ T cells are important in the very early phase of human AKI and should be considered when designing clinical trials aimed at preventing kidney damage.

Trial registration

ClinicalTrials.gov, NCT01915446. Registered on 5 Aug 2013.
Appendix
Available only for authorised users
Literature
1.
go back to reference Kashani K, Al-Khafaji A, Ardiles T, Artigas A, Bagshaw SM, Bell M, Bihorac A, Birkhahn R, Cely CM, Chawla LS, et al. Discovery and validation of cell cycle arrest biomarkers in human acute kidney injury. Crit Care. 2013;17(1):R25.CrossRefPubMedPubMedCentral Kashani K, Al-Khafaji A, Ardiles T, Artigas A, Bagshaw SM, Bell M, Bihorac A, Birkhahn R, Cely CM, Chawla LS, et al. Discovery and validation of cell cycle arrest biomarkers in human acute kidney injury. Crit Care. 2013;17(1):R25.CrossRefPubMedPubMedCentral
2.
go back to reference Bihorac A, Chawla LS, Shaw AD, Al-Khafaji A, Davison DL, Demuth GE, Fitzgerald R, Gong MN, Graham DD, Gunnerson K, et al. Validation of cell-cycle arrest biomarkers for acute kidney injury using clinical adjudication. Am J Respir Crit Care Med. 2014;189(8):932–9.CrossRefPubMed Bihorac A, Chawla LS, Shaw AD, Al-Khafaji A, Davison DL, Demuth GE, Fitzgerald R, Gong MN, Graham DD, Gunnerson K, et al. Validation of cell-cycle arrest biomarkers for acute kidney injury using clinical adjudication. Am J Respir Crit Care Med. 2014;189(8):932–9.CrossRefPubMed
3.
go back to reference Gocze I, Renner P, Graf BM, Schlitt HJ, Bein T, Pfister K. Simplified approach for the assessment of kidney perfusion and acute kidney injury at the bedside using contrast-enhanced ultrasound. Intensive Care Med. 2015;41(2):362–3.CrossRefPubMed Gocze I, Renner P, Graf BM, Schlitt HJ, Bein T, Pfister K. Simplified approach for the assessment of kidney perfusion and acute kidney injury at the bedside using contrast-enhanced ultrasound. Intensive Care Med. 2015;41(2):362–3.CrossRefPubMed
4.
go back to reference Meersch M, Schmidt C, Hoffmeier A, Van Aken H, Wempe C, Gerss J, Zarbock A. Prevention of cardiac surgery-associated AKI by implementing the KDIGO guidelines in high risk patients identified by biomarkers: the PrevAKI randomized controlled trial. Intensive Care Med. 2017;43(11):1551–61.CrossRefPubMedPubMedCentral Meersch M, Schmidt C, Hoffmeier A, Van Aken H, Wempe C, Gerss J, Zarbock A. Prevention of cardiac surgery-associated AKI by implementing the KDIGO guidelines in high risk patients identified by biomarkers: the PrevAKI randomized controlled trial. Intensive Care Med. 2017;43(11):1551–61.CrossRefPubMedPubMedCentral
5.
go back to reference Göcze I, Jauch D, Götz M, Kennedy P, Jung B, Zeman F, Gnewuch C, Graf BM, Gnann W, Banas B, et al. Biomarker-guided intervention to prevent acute kidney injury after major surgery: the prospective randomized BigpAK study. Ann Surg. 2018;267(6):1013–20.CrossRefPubMed Göcze I, Jauch D, Götz M, Kennedy P, Jung B, Zeman F, Gnewuch C, Graf BM, Gnann W, Banas B, et al. Biomarker-guided intervention to prevent acute kidney injury after major surgery: the prospective randomized BigpAK study. Ann Surg. 2018;267(6):1013–20.CrossRefPubMed
6.
7.
go back to reference Andres-Hernando A, Okamura K, Bhargava R, Kiekhaefer CM, Soranno D, Kirkbride-Romeo LA, Gil HW, Altmann C, Faubel S. Circulating IL-6 upregulates IL-10 production in splenic CD4+ T cells and limits acute kidney injury-induced lung inflammation. Kidney Int. 2017;91(5):1057–69.CrossRefPubMed Andres-Hernando A, Okamura K, Bhargava R, Kiekhaefer CM, Soranno D, Kirkbride-Romeo LA, Gil HW, Altmann C, Faubel S. Circulating IL-6 upregulates IL-10 production in splenic CD4+ T cells and limits acute kidney injury-induced lung inflammation. Kidney Int. 2017;91(5):1057–69.CrossRefPubMed
8.
go back to reference Weller S, Varrier M, Ostermann M. Lymphocyte function in human acute kidney injury. Nephron. 2017;137(4):287–93.CrossRefPubMed Weller S, Varrier M, Ostermann M. Lymphocyte function in human acute kidney injury. Nephron. 2017;137(4):287–93.CrossRefPubMed
9.
go back to reference Kellum JA, Lameire N, KDIGO AKI Guideline Work Group. Diagnosis, evaluation, and management of acute kidney injury: a KDIGO summary (Part 1). Crit Care. 2013;17(1):204.CrossRefPubMedPubMedCentral Kellum JA, Lameire N, KDIGO AKI Guideline Work Group. Diagnosis, evaluation, and management of acute kidney injury: a KDIGO summary (Part 1). Crit Care. 2013;17(1):204.CrossRefPubMedPubMedCentral
10.
go back to reference Hoste EA, McCullough PA, Kashani K, Chawla LS, Joannidis M, Shaw AD, Feldkamp T, Uettwiller-Geiger DL, McCarthy P, Shi J, et al. Derivation and validation of cutoffs for clinical use of cell cycle arrest biomarkers. Nephrol Dial Transplant. 2014;29(11):2054–61.CrossRefPubMedPubMedCentral Hoste EA, McCullough PA, Kashani K, Chawla LS, Joannidis M, Shaw AD, Feldkamp T, Uettwiller-Geiger DL, McCarthy P, Shi J, et al. Derivation and validation of cutoffs for clinical use of cell cycle arrest biomarkers. Nephrol Dial Transplant. 2014;29(11):2054–61.CrossRefPubMedPubMedCentral
11.
go back to reference Linkermann A, Brasen JH, Himmerkus N, Liu S, Huber TB, Kunzendorf U, Krautwald S. Rip1 (receptor-interacting protein kinase 1) mediates necroptosis and contributes to renal ischemia/reperfusion injury. Kidney Int. 2012;81(8):751–61.CrossRefPubMed Linkermann A, Brasen JH, Himmerkus N, Liu S, Huber TB, Kunzendorf U, Krautwald S. Rip1 (receptor-interacting protein kinase 1) mediates necroptosis and contributes to renal ischemia/reperfusion injury. Kidney Int. 2012;81(8):751–61.CrossRefPubMed
12.
go back to reference Chien YH, Meyer C, Bonneville M. γδ T cells: first line of defense and beyond. Annu Rev Immunol. 2014;32:121–55.CrossRefPubMed Chien YH, Meyer C, Bonneville M. γδ T cells: first line of defense and beyond. Annu Rev Immunol. 2014;32:121–55.CrossRefPubMed
13.
go back to reference Hayday AC. γδ T cells and the lymphoid stress-surveillance response. Immunity. 2009;31(2):184–96.CrossRefPubMed Hayday AC. γδ T cells and the lymphoid stress-surveillance response. Immunity. 2009;31(2):184–96.CrossRefPubMed
14.
go back to reference Gentles AJ, Newman AM, Liu CL, Bratman SV, Feng W, Kim D, Nair VS, Xu Y, Khuong A, Hoang CD, et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med. 2015;21(8):938–45.CrossRefPubMedPubMedCentral Gentles AJ, Newman AM, Liu CL, Bratman SV, Feng W, Kim D, Nair VS, Xu Y, Khuong A, Hoang CD, et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med. 2015;21(8):938–45.CrossRefPubMedPubMedCentral
15.
go back to reference Shichita T, Sugiyama Y, Ooboshi H, Sugimori H, Nakagawa R, Takada I, Iwaki T, Okada Y, Iida M, Cua DJ, et al. Pivotal role of cerebral interleukin-17-producing γδT cells in the delayed phase of ischemic brain injury. Nat Med. 2009;15(8):946–50.CrossRefPubMed Shichita T, Sugiyama Y, Ooboshi H, Sugimori H, Nakagawa R, Takada I, Iwaki T, Okada Y, Iida M, Cua DJ, et al. Pivotal role of cerebral interleukin-17-producing γδT cells in the delayed phase of ischemic brain injury. Nat Med. 2009;15(8):946–50.CrossRefPubMed
17.
go back to reference Eggenhofer E, Rovira J, Sabet-Baktach M, Groell A, Scherer MN, Dahlke MH, Farkas SA, Loss M, Koehl GE, Lang SA, et al. Unconventional RORγt+ T cells drive hepatic ischemia reperfusion injury. J Immunol. 2013;191(1):480–7.CrossRefPubMed Eggenhofer E, Rovira J, Sabet-Baktach M, Groell A, Scherer MN, Dahlke MH, Farkas SA, Loss M, Koehl GE, Lang SA, et al. Unconventional RORγt+ T cells drive hepatic ischemia reperfusion injury. J Immunol. 2013;191(1):480–7.CrossRefPubMed
18.
go back to reference Gelderblom M, Arunachalam P, Magnus T. γδ T cells as early sensors of tissue damage and mediators of secondary neurodegeneration. Front Cell Neurosci. 2014;8:368.CrossRefPubMedPubMedCentral Gelderblom M, Arunachalam P, Magnus T. γδ T cells as early sensors of tissue damage and mediators of secondary neurodegeneration. Front Cell Neurosci. 2014;8:368.CrossRefPubMedPubMedCentral
19.
go back to reference Rani M, Zhang Q, Oppeltz RF, Schwacha MG. Gamma delta T cells regulate inflammatory cell infiltration of the lung after trauma-hemorrhage. Shock. 2015;43(6):589–97.CrossRefPubMed Rani M, Zhang Q, Oppeltz RF, Schwacha MG. Gamma delta T cells regulate inflammatory cell infiltration of the lung after trauma-hemorrhage. Shock. 2015;43(6):589–97.CrossRefPubMed
20.
go back to reference Wang X, Sun R, Wei H, Tian Z. High-mobility group box 1 (HMGB1)-Toll-like receptor (TLR)4-interleukin (IL)-23-IL-17A axis in drug-induced damage-associated lethal hepatitis: interaction of γδ T cells with macrophages. Hepatology. 2013;57(1):373–84. Wang X, Sun R, Wei H, Tian Z. High-mobility group box 1 (HMGB1)-Toll-like receptor (TLR)4-interleukin (IL)-23-IL-17A axis in drug-induced damage-associated lethal hepatitis: interaction of γδ T cells with macrophages. Hepatology. 2013;57(1):373–84.
21.
go back to reference Savransky V, Molls RR, Burne-Taney M, Chien CC, Racusen L, Rabb H. Role of the T-cell receptor in kidney ischemia-reperfusion injury. Kidney Int. 2006;69(2):233–8.CrossRefPubMed Savransky V, Molls RR, Burne-Taney M, Chien CC, Racusen L, Rabb H. Role of the T-cell receptor in kidney ischemia-reperfusion injury. Kidney Int. 2006;69(2):233–8.CrossRefPubMed
22.
go back to reference Mulay SR, Linkermann A, Anders HJ. Necroinflammation in kidney disease. J Am Soc Nephrol. 2016;27(1):27–39.CrossRefPubMed Mulay SR, Linkermann A, Anders HJ. Necroinflammation in kidney disease. J Am Soc Nephrol. 2016;27(1):27–39.CrossRefPubMed
24.
go back to reference Marlin R, Pappalardo A, Kaminski H, Willcox CR, Pitard V, Netzer S, Khairallah C, Lomenech AM, Harly C, Bonneville M, et al. Sensing of cell stress by human γδ TCR-dependent recognition of annexin A2. Proc Natl Acad Sci U S A. 2017;114(12):3163–8.CrossRefPubMedPubMedCentral Marlin R, Pappalardo A, Kaminski H, Willcox CR, Pitard V, Netzer S, Khairallah C, Lomenech AM, Harly C, Bonneville M, et al. Sensing of cell stress by human γδ TCR-dependent recognition of annexin A2. Proc Natl Acad Sci U S A. 2017;114(12):3163–8.CrossRefPubMedPubMedCentral
25.
go back to reference Zou C, Zhao P, Xiao Z, Han X, Fu F, Fu L. γδ T cells in cancer immunotherapy. Oncotarget. 2017;8(5):8900–9.CrossRefPubMed Zou C, Zhao P, Xiao Z, Han X, Fu F, Fu L. γδ T cells in cancer immunotherapy. Oncotarget. 2017;8(5):8900–9.CrossRefPubMed
26.
go back to reference Zarbock A, Schmidt C, Van Aken H, Wempe C, Martens S, Zahn PK, Wolf B, Goebel U, Schwer CI, Rosenberger P, et al. Effect of remote ischemic preconditioning on kidney injury among high-risk patients undergoing cardiac surgery: a randomized clinical trial. JAMA. 2015;313(21):2133–41.CrossRefPubMed Zarbock A, Schmidt C, Van Aken H, Wempe C, Martens S, Zahn PK, Wolf B, Goebel U, Schwer CI, Rosenberger P, et al. Effect of remote ischemic preconditioning on kidney injury among high-risk patients undergoing cardiac surgery: a randomized clinical trial. JAMA. 2015;313(21):2133–41.CrossRefPubMed
27.
go back to reference Chen CB, Liu LS, Zhou J, Wang XP, Han M, Jiao XY, He XS, Yuan XP. Up-regulation of HMGB1 exacerbates renal ischemia-reperfusion injury by stimulating inflammatory and immune responses through the TLR4 signaling pathway in mice. Cell Physiol Biochem. 2017;41(6):2447–60.CrossRefPubMed Chen CB, Liu LS, Zhou J, Wang XP, Han M, Jiao XY, He XS, Yuan XP. Up-regulation of HMGB1 exacerbates renal ischemia-reperfusion injury by stimulating inflammatory and immune responses through the TLR4 signaling pathway in mice. Cell Physiol Biochem. 2017;41(6):2447–60.CrossRefPubMed
28.
go back to reference Rabb H, Griffin MD, McKay DB, Swaminathan S, Pickkers P, Rosner MH, Kellum JA, Ronco C, Acute Dialysis Quality Initiative Consensus XIII Work Group. Inflammation in AKI: current understanding, key questions, and knowledge gaps. J Am Soc Nephrol. 2016;27(2):371–9.CrossRefPubMed Rabb H, Griffin MD, McKay DB, Swaminathan S, Pickkers P, Rosner MH, Kellum JA, Ronco C, Acute Dialysis Quality Initiative Consensus XIII Work Group. Inflammation in AKI: current understanding, key questions, and knowledge gaps. J Am Soc Nephrol. 2016;27(2):371–9.CrossRefPubMed
29.
go back to reference Xia Q, Duan L, Shi L, Zheng F, Gong F, Fang M. High-mobility group box 1 accelerates early acute allograft rejection via enhancing IL-17+ γδ T-cell response. Transpl Int. 2014;27(4):399–407.CrossRefPubMed Xia Q, Duan L, Shi L, Zheng F, Gong F, Fang M. High-mobility group box 1 accelerates early acute allograft rejection via enhancing IL-17+ γδ T-cell response. Transpl Int. 2014;27(4):399–407.CrossRefPubMed
30.
go back to reference Schwacha MG, Rani M, Nicholson SE, Lewis AM, Holloway TL, Sordo S, Cap AP. Dermal γδ T-cells can be activated by mitochondrial damage-associated molecular patterns. PLoS One. 2016;11(7):e0158993.CrossRefPubMedPubMedCentral Schwacha MG, Rani M, Nicholson SE, Lewis AM, Holloway TL, Sordo S, Cap AP. Dermal γδ T-cells can be activated by mitochondrial damage-associated molecular patterns. PLoS One. 2016;11(7):e0158993.CrossRefPubMedPubMedCentral
31.
go back to reference Schwacha MG, Rani M, Zhang Q, Nunez-Cantu O, Cap AP. Mitochondrial damage-associated molecular patterns activate γδ T-cells. Innate Immun. 2014;20(3):261–8.CrossRefPubMed Schwacha MG, Rani M, Zhang Q, Nunez-Cantu O, Cap AP. Mitochondrial damage-associated molecular patterns activate γδ T-cells. Innate Immun. 2014;20(3):261–8.CrossRefPubMed
33.
go back to reference Shirali AC, Goldstein DR. Tracking the toll of kidney disease. J Am Soc Nephrol. 2008;19(8):1444–50.CrossRefPubMed Shirali AC, Goldstein DR. Tracking the toll of kidney disease. J Am Soc Nephrol. 2008;19(8):1444–50.CrossRefPubMed
34.
Metadata
Title
Postoperative cellular stress in the kidney is associated with an early systemic γδ T-cell immune cell response
Authors
Ivan Göcze
Katharina Ehehalt
Florian Zeman
Paloma Riquelme
Karin Pfister
Bernhard M. Graf
Thomas Bein
Edward K. Geissler
Piotr Kasprzak
Hans J. Schlitt
John A. Kellum
James A. Hutchinson
Elke Eggenhofer
Philipp Renner
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Critical Care / Issue 1/2018
Electronic ISSN: 1364-8535
DOI
https://doi.org/10.1186/s13054-018-2094-x

Other articles of this Issue 1/2018

Critical Care 1/2018 Go to the issue