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

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, 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

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.

Keywords: Aortic surgery; Immunosurveillance; Ischemia-reperfusion injury; Tubular cell stress; γδ T cells.

Conflict of interest statement

Ethics approval and consent to participate

The study protocol was approved by the local institutional review board (University of Regensburg Ethics Committee, no. 13-101-0027). Written consent was obtained from all patients prior to study entry. The study is registered with ClinicalTrials.gov (NCT01915446).

Consent for publication

Not applicable.

Competing interests

JAK has received grant support and consulting fees from Astute Medical unrelated to the current study. IG have received lecture fees from Astute Medical unrelated to the current study. PR was supported by a research grant from the German Interdisciplinary Association of Intensive Care and Emergency Medicine. All of the other 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 between human γδ T-cell subsets and biomarkers of kidney stress. a Gating strategy used for flow cytometric analysis. Following exclusion of doublets, CD45+ cells were analyzed and further specified according to size/granularity. γδ T cells were defined as CD3+Vδ2+ cells. b Perioperative changes in human peripheral γδ T-cell frequencies were analyzed and stratified by biomarker results. In patients with elevated biomarkers, greater changes could be observed. c Perioperative changes (postsurgery versus presurgery) of T-cell subsets in human peripheral blood, including γδ T cells, were correlated with the biomarker level ([TIMP-2]•[IGFBP7]). Here, only changes in the γδ T-cell subpopulation significantly correlated with biomarker testing (p = 0.022), whereas no significant correlation could be observed in the (global) CD3 population or CD4/CD8 T-cell subsets
Fig. 2
Fig. 2
Murine ischemia-reperfusion model. a Thirty minutes of warm ischemia induced a highly significant increase in creatinine (left) and urea (right) after 24 hours. b Accordingly, histology (H&E staining) showed acute tubular injury with immune cell infiltration. Original magnification 200 ×. c Characterization of T-cell subsets isolated from murine kidneys (n = 6 per group) showed that γδ T cells, in contrast to other T-cell subpopulations, were significantly increased after ischemia-reperfusion injury. Data are shown as mean values with SEM. The Mann-Whitney U test was used to determine statistical significance. n.s. Not significant. **p < 0.01; ****p < 0.0001
Fig. 3
Fig. 3
HMGB1 as a potential driver of γδ T-cell response. a In the murine ischemia-reperfusion injury (IRI) model, the pattern of peripheral γδ T-cell changes was similar to that in the human samples (see Fig. 1c). Here, peripheral γδ T-cell frequency was significantly reduced after IRI (left, p = 0.036). Interestingly, HMGB1 gene expression was significantly increased in murine kidneys (right, p < 0.0001). b In study participants, the HMGB1 protein was increased in the peripheral blood on day 1 after surgery. Strikingly, biomarker-positive study subjects had significantly higher HMGB1 levels on day 1 than biomarker-negative patients (p = 0.019). All data are shown as means with SEM and were evaluated for significance using the Mann-Whitney U test

References

    1. 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. doi: 10.1186/cc12503.
    1. 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):932–939. doi: 10.1164/rccm.201401-0077OC.
    1. 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):362–363. doi: 10.1007/s00134-014-3554-7.
    1. 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):1551–1561. doi: 10.1007/s00134-016-4670-3.
    1. 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):1013–1020. doi: 10.1097/SLA.0000000000002485.
    1. Raulet DH, Guerra N. Oncogenic stress sensed by the immune system: role of natural killer cell receptors. Nat Rev Immunol. 2009;9(8):568–580. doi: 10.1038/nri2604.
    1. 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):1057–1069. doi: 10.1016/j.kint.2016.12.014.
    1. Weller S, Varrier M, Ostermann M. Lymphocyte function in human acute kidney injury. Nephron. 2017;137(4):287–293. doi: 10.1159/000478538.
    1. 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. doi: 10.1186/cc11454.
    1. 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):2054–2061. doi: 10.1093/ndt/gfu292.
    1. 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):751–761. doi: 10.1038/ki.2011.450.
    1. Chien YH, Meyer C, Bonneville M. γδ T cells: first line of defense and beyond. Annu Rev Immunol. 2014;32:121–155. doi: 10.1146/annurev-immunol-032713-120216.
    1. Hayday AC. γδ T cells and the lymphoid stress-surveillance response. Immunity. 2009;31(2):184–196. doi: 10.1016/j.immuni.2009.08.006.
    1. 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):938–945. doi: 10.1038/nm.3909.
    1. 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):946–950. doi: 10.1038/nm.1999.
    1. Vantourout P, Hayday A. Six-of-the-best: unique contributions of gammadelta T cells to immunology. Nat Rev Immunol. 2013;13(2):88–100. doi: 10.1038/nri3384.
    1. 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):480–487. doi: 10.4049/jimmunol.1202975.
    1. 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. doi: 10.3389/fncel.2014.00368.
    1. 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):589–597. doi: 10.1097/SHK.0000000000000358.
    1. 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.
    1. 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):233–238. doi: 10.1038/sj.ki.5000038.
    1. Mulay SR, Linkermann A, Anders HJ. Necroinflammation in kidney disease. J Am Soc Nephrol. 2016;27(1):27–39. doi: 10.1681/ASN.2015040405.
    1. Kinsey GR, Okusa MD. Role of leukocytes in the pathogenesis of acute kidney injury. Crit Care. 2012;16(2):214. doi: 10.1186/cc11228.
    1. 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):3163–3168. doi: 10.1073/pnas.1621052114.
    1. Zou C, Zhao P, Xiao Z, Han X, Fu F, Fu L. γδ T cells in cancer immunotherapy. Oncotarget. 2017;8(5):8900–8909. doi: 10.18632/oncotarget.13051.
    1. 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):2133–2141. doi: 10.1001/jama.2015.4189.
    1. 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):2447–2460. doi: 10.1159/000475914.
    1. 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):371–379. doi: 10.1681/ASN.2015030261.
    1. 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. doi: 10.1111/tri.12264.
    1. 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. doi: 10.1371/journal.pone.0158993.
    1. 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):261–268. doi: 10.1177/1753425913488969.
    1. Goligorsky MS. TLR4 and HMGB1: partners in crime? Kidney Int. 2011;80(5):450–452. doi: 10.1038/ki.2011.170.
    1. Shirali AC, Goldstein DR. Tracking the toll of kidney disease. J Am Soc Nephrol. 2008;19(8):1444–1450. doi: 10.1681/ASN.2008010123.
    1. Tang D, Kang R, Zeh HJ, 3rd, Lotze MT. High-mobility group box 1, oxidative stress, and disease. Antioxid Redox Signal. 2011;14(7):1315–1335. doi: 10.1089/ars.2010.3356.

Source: PubMed

3
Se inscrever