Extremes of Interferon-Stimulated Gene Expression Associate with Worse Outcomes in the Acute Respiratory Distress Syndrome

Jerry A Nick, Silvia M Caceres, Jennifer E Kret, Katie R Poch, Matthew Strand, Anna V Faino, David P Nichols, Milene T Saavedra, Jennifer L Taylor-Cousar, Mark W Geraci, Ellen L Burnham, Michael B Fessler, Benjamin T Suratt, Edward Abraham, Marc Moss, Kenneth C Malcolm, Jerry A Nick, Silvia M Caceres, Jennifer E Kret, Katie R Poch, Matthew Strand, Anna V Faino, David P Nichols, Milene T Saavedra, Jennifer L Taylor-Cousar, Mark W Geraci, Ellen L Burnham, Michael B Fessler, Benjamin T Suratt, Edward Abraham, Marc Moss, Kenneth C Malcolm

Abstract

Acute Respiratory Distress Syndrome (ARDS) severity may be influenced by heterogeneity of neutrophil activation. Interferon-stimulated genes (ISG) are a broad gene family induced by Type I interferons, often as a response to viral infections, which evokes extensive immunomodulation. We tested the hypothesis that over- or under-expression of immunomodulatory ISG by neutrophils is associated with worse clinical outcomes in patients with ARDS. Genome-wide transcriptional profiles of circulating neutrophils isolated from patients with sepsis-induced ARDS (n = 31) and healthy controls (n = 19) were used to characterize ISG expression. Hierarchical clustering of expression identified 3 distinct subject groups with Low, Mid and High ISG expression. ISG accounting for the greatest variability in expression were identified (MX1, IFIT1, and ISG15) and used to analyze a prospective cohort at the Colorado ARDS Network site. One hundred twenty ARDS patients from four urban hospitals were enrolled within 72 hours of initiation of mechanical ventilation. Circulating neutrophils were isolated from patients and expression of ISG determined by PCR. Samples were stratified by standard deviation from the mean into High (n = 21), Mid, (n = 82) or Low (n = 17) ISG expression. Clinical outcomes were compared between patients with High or Low ISG expression to those with Mid-range expression. At enrollment, there were no differences in age, gender, co-existing medical conditions, or type of physiologic injury between cohorts. After adjusting for age, race, gender and BMI, patients with either High or Low ISG expression had significantly worse clinical outcomes than those in the Mid for number of 28-day ventilator- and ICU-free days (P = 0.0006 and 0.0004), as well as 90-day mortality and 90-day home with unassisted breathing (P = 0.02 and 0.004). These findings suggest extremes of ISG expression by circulating neutrophils from ARDS patients recovered early in the syndrome are associated with poorer clinical outcomes.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Transcriptome analysis to characterize ISG…
Fig 1. Transcriptome analysis to characterize ISG expression of ARDS patient and healthy control neutrophils.
(A) Log2-transformed expression of 31 unique transcripts within the Type I Interferon-mediated Signaling Pathway Cluster from neutrophils isolated from sepsis-induced ARDS patients (n = 31) and healthy volunteers (n = 19). ISG expression was ordered by hierarchical clustering (Euclidean distance with complete linkage), and contains only genes determined to significantly change between subjects. Three major clusters of subjects (columns) are broadly grouped as High ISG expression (left), Mid (middle) and Low (right). Subject groupings are represented by yellow (healthy) and blue (ARDS) blocks at the profile base. Only ARDS patients were contained within the Low ISG expression subject cluster. When these genes were ranked for extent of variance of expression using relative size of the standard deviation between subjects, the genes with the largest variance were identified as MX1, ISG15, IFIT1, and IFIT3 (identified by shading). (B) A Principal Component Analysis identified the same 4 genes that comprised nearly all of the variability between subjects in Panel A.
Fig 2. Classification of ARDS patients based…
Fig 2. Classification of ARDS patients based on neutrophil ISG expression.
The mean of the transformed expression values of neutrophil MX1, IFIT1, and ISG15 is plotted for ARDS patients (triangle, n = 120) or healthy subjects (circle, n = 40). Subjects whose ISG expression was greater or less than one standard deviation from the mean were designated as High (red) or Low (blue) ISG expressers, respectively. ISG expression in healthy subjects overlapped only with the High and Mid cohorts of ARDS patients.
Fig 3. Kaplan-Meier Analysis of 90-Day Discharge…
Fig 3. Kaplan-Meier Analysis of 90-Day Discharge to Home with Unassisted Breathing and Survival.
(A) Proportion of patients confirmed to be discharged to home with unassisted breathing within 90 days of study enrollment. Overall log-rank P-value comparing groups was 0.009. After adjusting for age, race, gender and BMI, rate of discharge to home was significantly lower for both the High ISG (red line, P = 0.006) and Low ISG (blue line, P = 0.004; p-values from Cox PH model) expressing cohorts when compared to the Mid (black line). (B) Proportion of patients confirmed to be dead within 90 days of study enrollment. Overall log-rank P-value comparing groups was 0.01. After adjusting for age, race, gender and BMI, rate of death was significantly worse for both the High ISG (red line, P = 0.02) and Low ISG (blue line, P = 0.02; p-values from Cox PH model) expressing cohorts when compared to the Mid (black line).
Fig 4. Association of ISG expression with…
Fig 4. Association of ISG expression with confirmed viral infections and circulating IFNα.
(A) The presence of a viral infection, as determined either through available medical history or within the course of clinical care, did not reach significance in the cohort with High ISG expression (5 of 21) compared to the Mid (8 of 82) and Low (1 of 17) cohorts (Fisher’s exact test P = 0.23). (B) Detection of circulating IFNα tended to occur more frequently within the High ISG cohort (11 of 21) compared to the Mid (20 of 76) and Low (5 of 15) cohorts, but this difference did not reach significance (Fisher’s exact test P = 0.08). (C) Levels of circulating IFNα trended higher within the High ISG cohort (n = 21) compared to the Mid (n = 76), (Kruskal-Wallis overall P-value = 0.03; High vs. Mid pairwise P = 0.009). n = 15 for the Low cohort. Plot depicts range (minimum≈0, maximum = upper whisker), 1st to 3rd quartile (box) and median (line).

References

    1. Iribarren C, Jacobs DR Jr., Sidney S, Gross MD, Eisner MD (2000) Cigarette smoking, alcohol consumption, and risk of ARDS: a 15-year cohort study in a managed care setting. Chest 117: 163–168.
    1. Calfee CS, Matthay MA, Eisner MD, Benowitz N, Call M, Pittet JF, et al. (2011) Active and passive cigarette smoking and acute lung injury after severe blunt trauma. Am J Respir Crit Care Med 183: 1660–1665. 10.1164/rccm.201011-1802OC
    1. Hudson L, Milberg J, Anardi D, Maunder R (1995) Clinical risk for development of the acute respiratory distress syndrome. Am J Respir Crit Care Med 151: 293–301.
    1. Moss M, Bucher B, Moore FA, Moore EE, Parsons PE (1996) The role of chronic alcohol abuse in the development of acute respiratory distress syndrome in adults. Jama 275: 50–54.
    1. Milberg JA, Davis DR, Steinberg KP, Hudson LD (1995) Improved survival of patients with acute respiratory distress syndrome (ARDS): 1983–1993. Jama 273: 306–309.
    1. Ely EW, Wheeler AP, Thompson BT, Ancukiewicz M, Steinberg KP, Bernard GR (2002) Recovery rate and prognosis in older persons who develop acute lung injury and the acute respiratory distress syndrome. Ann Intern Med 136: 25–36.
    1. Kangelaris KN, Sapru A, Calfee CS, Liu KD, Pawlikowska L, Witte JS, et al. (2012) The association between a Darc gene polymorphism and clinical outcomes in African American patients with acute lung injury. Chest 141: 1160–1169. 10.1378/chest.11-1766
    1. Moss M, Mannino DM (2002) Race and gender differences in acute respiratory distress syndrome deaths in the United States: an analysis of multiple-cause mortality data (1979–1996). Crit Care Med 30: 1679–1685.
    1. Rubenfeld GD, Caldwell E, Peabody E, Weaver J, Martin DP, Neff M, et al. (2005) Incidence and outcomes of acute lung injury. N Engl J Med 353: 1685–1693.
    1. Matthay MA, Ware LB, Zimmerman GA (2012) The acute respiratory distress syndrome. J Clin Invest 122: 2731–2740. 10.1172/JCI60331
    1. Gao L, Barnes KC (2009) Recent advances in genetic predisposition to clinical acute lung injury. Am J Physiol Lung Cell Mol Physiol 296: L713–725. 10.1152/ajplung.90269.2008
    1. Arcaroli J, Fessler MB, Abraham E (2005) Genetic Polymorphisms And Sepsis. Shock 24: 300–312.
    1. Ware LB, Matthay MA (2000) The acute respiratory distress syndrome. N Engl J Med 342: 1334–1349.
    1. Abraham E (2003) Neutrophils and acute lung injury. Crit Care Med 31: S195–199.
    1. Lamb NJ, Gutteridge JMC, Baker C, Evans TW, Quinlan GJ (1999) Oxidative damage to proteins of BAL fluid in patients with ARDS: Evidence for neutrophil-mediated hydroxylation, nitration, and chlorination. Critical Care Medicine 27: 1738–1744.
    1. Borregaard N, Cowland JB (1997) Granules of the Human Neutrophilic Polymorphonuclear Leukocyte. Blood 89: 3503–3521.
    1. Buckley JM, Wang JH, Redmond HP (2006) Cellular reprogramming by gram-positive bacterial components: a review. J Leukoc Biol 80: 731–741.
    1. Fan H, Cook JA (2004) Molecular mechanisms of endotoxin tolerance. J Endotoxin Res 10: 71–84.
    1. Lee KS, Choi YH, Kim YS, Baik SH, Oh YJ, Sheen SS, et al. (2008) Evaluation of bronchoalveolar lavage fluid from ARDS patients with regard to apoptosis. Respir Med 102: 464–469.
    1. Aggarwal A, Baker CS, Evans TW, Haslam PL (2000) G-CSF and IL-8 but not GM-CSF correlate with severity of pulmonary neutrophilia in acute respiratory distress syndrome. Eur Respir J 15: 895–901.
    1. Goodman RB, Strieter RM, Martin DP, Steinberg KP, Milberg JA, Maunder RJ, et al. (1996) Inflammatory cytokines in patients with persistence of the acute respiratory distress syndrome. Am J Respir Crit Care Med 154: 602–611.
    1. Lesur O, Kokis A, Hermans C, Fulop T, Bernard A, Lane D (2000) Interleukin-2 involvement in early acute respiratory distress syndrome: relationship with polymorphonuclear neutrophil apoptosis and patient survival. Crit Care Med 28: 3814–3822.
    1. Mascellino MT, Delogu G, Pelaia MR, Ponzo R, Parrinello R, Giardina A (2001) Reduced bactericidal activity against Staphylococcus aureus and Pseudomonas aeruginosa of blood neutrophils from patients with early adult respiratory distress syndrome. J Med Microbiol 50: 49–54.
    1. Martin TR, Pistorese BP, Hudson LD, Maunder RJ (1991) The function of lung and blood neutrophils in patients with the adult respiratory distress syndrome. Implications for the pathogenesis of lung infections. Am Rev Respir Dis 144: 254–262.
    1. Rivkind AI SJ, Littleton M, De Gaetano A, Mamantov T, Laghi F, Stoklosa JC. (1991) Neutrophil oxidative burst activation and the pattern of respiratory physiologic abnormalities in the fulminant post-traumatic adult respiratory distress syndrome. Circ Shock 33: 48–62.
    1. Fialkow L, Fochesatto Filho L, Bozzetti MC, Milani AR, Rodrigues Filho EM, Ladniuk RM, et al. (2006) Neutrophil apoptosis: a marker of disease severity in sepsis and sepsis-induced acute respiratory distress syndrome. Crit Care 10: R155
    1. McNamee LA, Harmsen AG (2006) Both influenza-induced neutrophil dysfunction and neutrophil-independent mechanisms contribute to increased susceptibility to a secondary Streptococcus pneumoniae infection. Infect Immun 74: 6707–6721.
    1. Brundage JF (2006) Interactions between influenza and bacterial respiratory pathogens: implications for pandemic preparedness. Lancet Infect Dis 6: 303–312.
    1. Brundage JF, Shanks GD (2008) Deaths from bacterial pneumonia during 1918–19 influenza pandemic. Emerg Infect Dis 14: 1193–1199. 10.3201/eid1408.071313
    1. Metzger DW, Sun K (2013) Immune dysfunction and bacterial coinfections following influenza. J Immunol 191: 2047–2052. 10.4049/jimmunol.1301152
    1. Sun K, Metzger DW (2008) Inhibition of pulmonary antibacterial defense by interferon-gamma during recovery from influenza infection. Nat Med 14: 558–564. 10.1038/nm1765
    1. Rothberg MB, Haessler SD, Brown RB (2008) Complications of viral influenza. Am J Med 121: 258–264. 10.1016/j.amjmed.2007.10.040
    1. Sellers TF Jr., Schulman J, Bouvier C, Mc CR, Kilbourne ED (1961) The influence of influenza virus infection on exogenous staphylococcal and endogenous murine bacterial infection of the bronchopulmonary tissues of mice. J Exp Med 114: 237–256.
    1. McCullers JA (2006) Insights into the interaction between influenza virus and pneumococcus. Clin Microbiol Rev 19: 571–582.
    1. Stetson DB, Medzhitov R (2006) Type 1 interferons in host defense. Immunity 25: 373–381.
    1. Roers A, Hochkeppel HK, Horisberger MA, Hovanessian A, Haller O (1994) MxA gene expression after live virus vaccination: a sensitive marker for endogenous type I interferon. J Infect Dis 169: 807–813.
    1. Halminen M, Ilonen J, Julkunen I, Ruuskanen O, Simell O, Makela MJ (1997) Expression of MxA protein in blood lymphocytes discriminates between viral and bacterial infections in febrile children. Pediatr Res 41: 647–650.
    1. Ramilo O, Allman W, Chung W, Mejias A, Ardura M, Glaser C, et al. (2007) Gene expression patterns in blood leukocytes discriminate patients with acute infections. Blood 109: 2066–2077.
    1. Zaas AK, Chen M, Varkey J, Veldman T, Hero AO 3rd, Lucas J, et al. (2009) Gene expression signatures diagnose influenza and other symptomatic respiratory viral infections in humans. Cell Host Microbe 6: 207–217. 10.1016/j.chom.2009.07.006
    1. Metz P, Reuter A, Bender S, Bartenschlager R (2013) Interferon-stimulated genes and their role in controlling hepatitis C virus. J Hepatol 59: 1331–1341. 10.1016/j.jhep.2013.07.033
    1. Schoggins JW, Wilson SJ, Panis M, Murphy MY, Jones CT, Bieniasz P, et al. (2011) A diverse range of gene products are effectors of the type I interferon antiviral response. Nature 472: 481–485. 10.1038/nature09907
    1. Pichlmair A, Reis e Sousa C (2007) Innate recognition of viruses. Immunity 27: 370–383.
    1. Parker D, Planet PJ, Soong G, Narechania A, Prince A (2014) Induction of type I interferon signaling determines the relative pathogenicity of Staphylococcus aureus strains. PLoS Pathog 10: e1003951 10.1371/journal.ppat.1003951
    1. Mancuso G, Midiri A, Biondo C, Beninati C, Zummo S, Galbo R, et al. (2007) Type I IFN signaling is crucial for host resistance against different species of pathogenic bacteria. J Immunol 178: 3126–3133.
    1. Carrero JA, Calderon B, Unanue ER (2004) Type I interferon sensitizes lymphocytes to apoptosis and reduces resistance to Listeria infection. J Exp Med 200: 535–540.
    1. O'Connell RM, Saha SK, Vaidya SA, Bruhn KW, Miranda GA, Zarnegar B, et al. (2004) Type I interferon production enhances susceptibility to Listeria monocytogenes infection. J Exp Med 200: 437–445.
    1. Kelly-Scumpia KM, Scumpia PO, Delano MJ, Weinstein JS, Cuenca AG, Wynn JL, et al. (2010) Type I interferon signaling in hematopoietic cells is required for survival in mouse polymicrobial sepsis by regulating CXCL10. J Exp Med 207: 319–326. 10.1084/jem.20091959
    1. Malcolm KC, Kret J, Young RL, Poch KR, Caceres SM, Douglas IS, et al. (2011) Bacteria-Specific Neutrophil Dysfunction Associated with Interferon-Stimulated Gene Expression in the Acute Respiratory Distress Syndrome. PLoS 6: e21958.
    1. Brukman A, Enquist LW (2006) Suppression of the interferon-mediated innate immune response by pseudorabies virus. J Virol 80: 6345–6356.
    1. Eidson KM, Hobbs WE, Manning BJ, Carlson P, DeLuca NA (2002) Expression of herpes simplex virus ICP0 inhibits the induction of interferon-stimulated genes by viral infection. J Virol 76: 2180–2191.
    1. Kotla S, Peng T, Bumgarner RE, Gustin KE (2008) Attenuation of the type I interferon response in cells infected with human rhinovirus. Virology 374: 399–410. 10.1016/j.virol.2008.01.022
    1. Kumthip K, Chusri P, Jilg N, Zhao L, Fusco DN, Zhao H, et al. (2012) Hepatitis C virus NS5A disrupts STAT1 phosphorylation and suppresses type I interferon signaling. J Virol 86: 8581–8591. 10.1128/JVI.00533-12
    1. Ivashkiv LB, Donlin LT (2014) Regulation of type I interferon responses. Nat Rev Immunol 14: 36–49. 10.1038/nri3581
    1. Bode JG, Brenndorfer ED, Haussinger D (2007) Subversion of innate host antiviral strategies by the hepatitis C virus. Arch Biochem Biophys 462: 254–265.
    1. Silva E, Arcaroli J, He Q, Svetkauskaite D, Coldren C, Nick JA, et al. (2007) HMGB1 and LPS induce distinct patterns of gene expression and activation in neutrophils from patients with sepsis-induced acute lung injury. Intensive Care Med.
    1. Golpon HA, Coldren CD, Zamora MR, Cosgrove GP, Moore MD, Tuder RM, et al. (2004) Emphysema lung tissue gene expression profiling. Am J Respir Cell Mol Biol 31: 595–600.
    1. Brazma A, Hingamp P, Quackenbush J, Sherlock G, Spellman P, Stoeckert C, et al. (2001) Minimum information about a microarray experiment (MIAME)-toward standards for microarray data. Nat Genet 29: 365–371.
    1. (2004) Expression profiling—best practices for data generation and interpretation in clinical trials. Nat Rev Genet 5: 229–237.
    1. Saeed AI, Sharov V, White J, Li J, Liang W, Bhagabati N, et al. (2003) TM4: a free, open-source system for microarray data management and analysis. Biotechniques 34: 374–378.
    1. Matthay MA, Brower RG, Carson S, Douglas IS, Eisner M, Hite D, et al. (2011) Randomized, placebo-controlled clinical trial of an aerosolized beta(2)-agonist for treatment of acute lung injury. Am J Respir Crit Care Med 184: 561–568. 10.1164/rccm.201012-2090OC
    1. Rice TW, Wheeler AP, Thompson BT, Steingrub J, Hite RD, Moss M, et al. (2012) Initial trophic vs full enteral feeding in patients with acute lung injury: the EDEN randomized trial. Jama 307: 795–803. 10.1001/jama.2012.137
    1. Rice TW, Wheeler AP, Thompson BT, deBoisblanc BP, Steingrub J, Rock P (2011) Enteral omega-3 fatty acid, gamma-linolenic acid, and antioxidant supplementation in acute lung injury. Jama 306: 1574–1581. 10.1001/jama.2011.1435
    1. National Heart L, Blood Institute ACTN, Truwit JD, Bernard GR, Steingrub J, Matthay MA, et al. (2014) Rosuvastatin for sepsis-associated acute respiratory distress syndrome. N Engl J Med 370: 2191–2200. 10.1056/NEJMoa1401520
    1. Levy MM, Fink MP, Marshall JC, Abraham E, Angus D, Cook D, et al. (2003) 2001 SCCM/ESICM/ACCP/ATS/SIS International Sepsis Definitions Conference. Intensive Care Med 29: 530–538.
    1. Haslett C, Guthrie LA, Kopaniak M, Johnston RB Jr., Henson PM (1985) Modulation of multiple neutrophil functions by trace amounts of bacterial LPS and by preparative methods. Am J Pathol 119: 101–110.
    1. Malcolm KC, Arndt PG, Manos EJ, Jones DA, Worthen GS (2003) Microarray analysis of lipopolysaccharide-treated human neutrophils. Am J Physiol Lung Cell Mol Physiol 284: L663–670.
    1. Schoenfeld DA, Bernard GR, Network A (2002) Statistical evaluation of ventilator-free days as an efficacy measure in clinical trials of treatments for acute respiratory distress syndrome. Crit Care Med 30: 1772–1777.
    1. Fessler MB, Malcolm KC, Duncan MW, Worthen GS (2002) A Genomic and Proteomic Analysis of Activation of the Human Neutrophil by Lipopolysaccharide and Its Mediation by p38 Mitogen-activated Protein Kinase. J Biol Chem 277: 31291–31302.
    1. Wright HJ, Matthews JB, Chapple IL, Ling-Mountford N, Cooper PR (2008) Periodontitis associates with a type 1 IFN signature in peripheral blood neutrophils. J Immunol 181: 5775–5784.
    1. Landolt-Marticorena C, Bonventi G, Lubovich A, Ferguson C, Unnithan T, Su J, et al. (2009) Lack of association between the interferon-alpha signature and longitudinal changes in disease activity in systemic lupus erythematosus. Ann Rheum Dis 68: 1440–1446. 10.1136/ard.2008.093146
    1. Ramilo O, Mejias A (2009) Shifting the paradigm: host gene signatures for diagnosis of infectious diseases. Cell Host Microbe 6: 199–200. 10.1016/j.chom.2009.08.007
    1. Malcolm KC, Worthen GS (2003) Lipopolysaccharide stimulates p38-dependent induction of antiviral genes in neutrophils independently of paracrine factors. J Biol Chem 278: 15693–15701.
    1. Morris AE, Stapleton RD, Rubenfeld GD, Hudson LD, Caldwell E, Steinberg KP (2007) The association between body mass index and clinical outcomes in acute lung injury. Chest 131: 342–348.
    1. Sadler AJ, Williams BR (2008) Interferon-inducible antiviral effectors. Nat Rev Immunol 8: 559–568. 10.1038/nri2314
    1. Bellingan G, Maksimow M, Howell DC, Stotz M, Beale R, Beatty M, et al. (2014) The effect of intravenous interferon-beta-1a (FP-1201) on lung CD73 expression and on acute respiratory distress syndrome mortality: an open-label study. Lancet Respir Med 2: 98–107. 10.1016/S2213-2600(13)70259-5
    1. Limaye AP, Kirby KA, Rubenfeld GD, Leisenring WM, Bulger EM, Neff MJ, et al. (2008) Cytomegalovirus reactivation in critically ill immunocompetent patients. JAMA 300: 413–422. 10.1001/jama.300.4.413

Source: PubMed

3
Iratkozz fel