Plasma Vanin-1 as a Novel Biomarker of Sepsis for Trauma Patients: A Prospective Multicenter Cohort Study

Hongxiang Lu, Anqiang Zhang, Dalin Wen, Juan Du, Jianhui Sun, Liang Qiao, Dingyuan Du, Wei Gu, Jianxin Jiang, Hongxiang Lu, Anqiang Zhang, Dalin Wen, Juan Du, Jianhui Sun, Liang Qiao, Dingyuan Du, Wei Gu, Jianxin Jiang

Abstract

Introduction: Vanin-1 plays a pivotal role in oxidative stress and the inflammatory response. However, its relationship with traumatic sepsis remains unknown. The aim of our study was to evaluate whether plasma vanin-1 could be used for the early prediction of traumatic sepsis.

Methods: In this three-stage prospective cohort study, severe trauma patients admitted from January 2015 to October 2018 at two hospitals were enrolled. Plasma vanin-1 levels were measured by enzyme-linked immunosorbent assay (ELISA). The associations among variables and traumatic sepsis were identified by logistic regression models and the receiver operating characteristic (ROC) curve was analyzed to evaluate the diagnostic efficiency.

Results: A total of 426 trauma patients (22 in the discovery cohort, 283 in the internal test cohort, and 121 in the external validation cohort) and 16 healthy volunteers were recruited. The plasma vanin-1 of trauma patients was significantly higher than that of healthy volunteers (P < 0.05). Patients with sepsis had higher plasma vanin-1 than patients without sepsis in the discovery trauma cohort (P < 0.05). In the internal test cohort, plasma vanin-1 at day 1 after trauma was significantly associated with the incidence of sepsis (OR = 3.92, 95% CI 2.68-5.72, P = 1.62 × 10-12). As a predictive biomarker, vanin-1 afforded a better area under the curve (AUC) (0.82, 95% CI 0.77-0.87) than C-reaction protein (CRP) (0.62, 95% CI 0.56-0.68, P < 0.0001), procalcitonin (PCT) (0.66, 95% CI 0.60-0.71, P < 0.0001), and Acute Physiology and Chronic Health Evaluation II (APACHE II) (0.71, 95% CI 0.65-0.76, P = 6.70 × 10-3). The relevance was further validated in the external validation cohort (OR = 4.26, 95% CI 2.22-8.17, P = 1.28 × 10-5), with an AUC of 0.83 (95% CI 0.75-0.89). Vanin-1 could also improve the diagnostic efficiency of APACHE II (AUC = 0.85).

Conclusions: Our study demonstrated that plasma vanin-1 increased among trauma patients and was independently associated with the risk of sepsis. Vanin-1 might be a potential biomarker for the early prediction of traumatic sepsis.

Trial registration: Clinicaltrials.gov Identifier, NCT01713205.

Keywords: Biomarker; Sepsis; Trauma; Vanin-1.

Figures

Fig. 1
Fig. 1
Kinetics of plasma vanin-1 levels in trauma patients on admission and at days 3, 5, 7, and 14 during hospitalization from the discovery cohort. a Plasma vanin-1 of trauma patients is significantly higher than that of healthy volunteers (n = 22 trauma patients vs. 16 healthy controls). b Trauma patients with sepsis have higher plasma vanin-1 than patients without sepsis at the early stage after injury (n = 11 patients with sepsis vs. 11 patients without sepsis, P = 0.03 for day 1 and P = 0.04 for day 3). Data are expressed as the mean and 95% CI. Statistical analysis comprised Student’s t test
Fig. 2
Fig. 2
Plasma vanin-1 in trauma patients with and without sepsis for the a internal test and b external validation cohorts. Patients who developed sepsis had significantly higher levels of plasma vanin-1 when compared with patients who did not develop sepsis at day 1 after trauma. All samples were collected on day 1 after injury. Black bars show the mean and 95% CI. Statistical analysis comprised Student’s t test
Fig. 3
Fig. 3
Receiver operating curve (ROC) analysis of VNN1, CRP, PCT, and APACHE II for sepsis after trauma. Plasma vanin-1 afforded the best predictive value compared to other biomarkers and scores in the internal test cohort (a, n = 283) and external validation cohort (b, n = 121)

References

    1. Bohmer AB, Just KS, Lefering R, et al. Factors influencing lengths of stay in the intensive care unit for surviving trauma patients: a retrospective analysis of 30,157 cases. Crit Care. 2014;18(4):R143.
    1. Zhang AQ, Gu W, Zeng L, et al. Genetic variants of microRNA sequences and susceptibility to sepsis in patients with major blunt trauma. Ann Surg. 2015;261(1):189–196.
    1. Lamparello AJ, Namas RA, Constantine G, et al. A conceptual time window-based model for the early stratification of trauma patients. J Intern Med. 2019;286(1):2–15.
    1. Haagsma JA, Graetz N, Bolliger I, et al. The global burden of injury: incidence, mortality, disability-adjusted life years and time trends from the Global Burden of Disease study 2013. Inj Prev. 2016;22(1):3–18.
    1. Vourc'h M, Roquilly A, Asehnoune K. Trauma-induced damage-associated molecular patterns-mediated remote organ injury and immunosuppression in the acutely Ill patient. Front Immunol. 2018;9:1330.
    1. Xiao W, Mindrinos MN, Seok J, et al. A genomic storm in critically injured humans. J Exp Med. 2011;208(13):2581–2590.
    1. Lindner HA, Balaban U, Sturm T, Weiss C, Thiel M, Schneider-Lindner V. An algorithm for systemic inflammatory response syndrome criteria-based prediction of sepsis in a polytrauma cohort. Crit Care Med. 2016;44(12):2199–2207.
    1. Asehnoune K, Balogh Z, Citerio G, et al. The research agenda for trauma critical care. Intensive Care Med. 2017;43(9):1340–1351.
    1. Naquet P, Pitari G, Dupre S, Galland F. Role of the Vnn1 pantetheinase in tissue tolerance to stress. Biochem Soc Trans. 2014;42(4):1094–1100.
    1. Nitto T, Onodera K. Linkage between coenzyme a metabolism and inflammation: roles of pantetheinase. J Pharmacol Sci. 2013;123(1):1–8.
    1. Berruyer C, Martin FM, Castellano R, et al. Vanin-1-/- mice exhibit a glutathione-mediated tissue resistance to oxidative stress. Mol Cell Biol. 2004;24(16):7214–7224.
    1. Martin F, Penet MF, Malergue F, et al. Vanin-1(-/-) mice show decreased NSAID- and Schistosoma-induced intestinal inflammation associated with higher glutathione stores. J Clin Invest. 2004;113(4):591–597.
    1. Meghari S, Berruyer C, Lepidi H, Galland F, Naquet P, Mege JL. Vanin-1 controls granuloma formation and macrophage polarization in Coxiella burnetii infection. Eur J Immunol. 2007;37(1):24–32.
    1. Sandesc M, Rogobete AF, Bedreag OH, et al. Analysis of oxidative stress-related markers in critically ill polytrauma patients: an observational prospective single-center study. Bosn J Basic Med Sci. 2018;18(2):191–197.
    1. Singer M, Deutschman CS, Seymour CW, et al. The third international consensus definitions for sepsis and septic shock (Sepsis-3) JAMA. 2016;315(8):801–810.
    1. Chung KP, Chen GY, Chuang TY, et al. Increased plasma acetylcarnitine in sepsis is associated with multiple organ dysfunction and mortality: a multicenter cohort study. Crit Care Med. 2019;47(2):210–218.
    1. Ling L, Lu HT, Wang HF, Shen MJ, Zhang HB. MicroRNA-203 acts as a potent suppressor in septic shock by alleviating lung injury via inhibition of VNN1. Kidney Blood Press Res. 2019;44(4):565–582.
    1. Yip KT, Das PK, Suria D, Lim CR, Ng GH, Liew CC. A case-controlled validation study of a blood-based seven-gene biomarker panel for colorectal cancer in Malaysia. J Exp Clin Cancer Res. 2010;29:128.
    1. Marshall KW, Mohr S, Khettabi FE, et al. A blood-based biomarker panel for stratifying current risk for colorectal cancer. Int J Cancer. 2010;126(5):1177–1186.
    1. Izzi V, Lakkala J, Devarajan R, et al. Vanin 1 (VNN1) levels predict poor outcome in acute myeloid leukemia. Am J Hematol. 2018;93(1):E4–E7.
    1. Washino S, Hosohata K, Oshima M, et al. A novel biomarker for acute kidney injury, vanin-1, for obstructive nephropathy: a prospective cohort pilot study. Int J Mol Sci. 2019;20(4):899.
    1. Hosohata K, Matsuoka H, Iwanaga K, Kumagai E. Urinary vanin-1 associated with chronic kidney disease in hypertensive patients: a pilot study. J Clin Hypertens (Greenwich) 2020;22(8):1458–1465.
    1. Hosohata K, Ando H, Fujiwara Y, Fujimura A. Vanin-1: a potential biomarker for nephrotoxicant-induced renal injury. Toxicology. 2011;290(1):82–88.
    1. Xiao C, Biagini Myers JM, Ji H, et al. Vanin-1 expression and methylation discriminate pediatric asthma corticosteroid treatment response. J Allergy Clin Immunol. 2015;136(4):923–31 e3.
    1. Sutherland A, Thomas M, Brandon RA, et al. Development and validation of a novel molecular biomarker diagnostic test for the early detection of sepsis. Crit Care. 2011;15(3):R149.
    1. Demaret J, Venet F, Friggeri A, et al. Marked alterations of neutrophil functions during sepsis-induced immunosuppression. J Leukoc Biol. 2015;98(6):1081–1090.
    1. Shalova IN, Lim JY, Chittezhath M, et al. Human monocytes undergo functional re-programming during sepsis mediated by hypoxia-inducible factor-1alpha. Immunity. 2015;42(3):484–498.
    1. Ciriello V, Gudipati S, Stavrou PZ, Kanakaris NK, Bellamy MC, Giannoudis PV. Biomarkers predicting sepsis in polytrauma patients: current evidence. Injury. 2013;44(12):1680–1692.
    1. Berruyer C, Pouyet L, Millet V, et al. Vanin-1 licenses inflammatory mediator production by gut epithelial cells and controls colitis by antagonizing peroxisome proliferator-activated receptor gamma activity. J Exp Med. 2006;203(13):2817–2827.
    1. Pouyet L, Roisin-Bouffay C, Clement A, et al. Epithelial vanin-1 controls inflammation-driven carcinogenesis in the colitis-associated colon cancer model. Inflamm Bowel Dis. 2010;16(1):96–104.
    1. Yamashita N, Yashiro M, Ogawa H, et al. Metabolic pathway catalyzed by vanin-1 pantetheinase plays a suppressive role in influenza virus replication in human alveolar epithelial A549 cells. Biochem Biophys Res Commun. 2017;489(4):466–471.
    1. Jansen PA, Kamsteeg M, Rodijk-Olthuis D, et al. Expression of the vanin gene family in normal and inflamed human skin: induction by proinflammatory cytokines. J Invest Dermatol. 2009;129(9):2167–2174.
    1. Zhang B, Lo C, Shen L, et al. The role of vanin-1 and oxidative stress-related pathways in distinguishing acute and chronic pediatric ITP. Blood. 2011;117(17):4569–4579.
    1. Ricote M, Glass CK. PPARs and molecular mechanisms of transrepression. Biochim Biophys Acta. 2007;1771(8):926–935.
    1. Szeles L, Torocsik D, Nagy L. PPARgamma in immunity and inflammation: cell types and diseases. Biochim Biophys Acta. 2007;1771(8):1014–1030.

Source: PubMed

3
Sottoscrivi