Semin Respir Crit Care Med 2019; 40(01): 012-018
DOI: 10.1055/s-0039-1683891
Review Article
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Deconstructing ARDS Variability: Platelet Count, an ARDS Intermediate Phenotype and Novel Mediator of Genetic Effects in ARDS

Paula Tejera
1   Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
,
David C. Christiani
1   Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
2   Massachusetts General Hospital (MGH), Boston, Massachusetts
› Author Affiliations
Further Information

Publication History

Publication Date:
06 May 2019 (online)

Abstract

Genome-wide association studies (GWASs) in acute respiratory distress syndrome (ARDS) have been hampered by the heterogeneity of the clinical phenotypes and the large sample size requirement. As the limitations of these studies to uncover the complex genetic architecture of ARDS are evident, new approaches intended to reduce data complexity need to be applied. Intermediate phenotypes are mechanism-related manifestations of the disease, located closer to the genetic substrate than to disease phenotype, and therefore able to reflect more directly and more strongly the effect of causal genes. The dissection of complex phenotypes into less complex intermediate phenotypes is a valuable strategy to facilitate the discovery of those genetic variants whose effect is not strong enough to be detected as markers of disease in traditional GWASs. Genetic causal inference methodologies can be then applied to estimate the implication of the intermediate trait in the causal circuit between genes and disease. By following this strategy, platelet count, a relevant intermediate quantitative trait in ARDS, has been recently identified as a novel mediator in the genetic contribution to ARDS risk and mortality. The use of intermediate phenotypes and causal inference are emerging methodological and statistical strategies that can help to overcome the limitations of traditional GWASs in ARDS. Moreover, these approaches can provide evidence for the mechanisms linking genes to ARDS and help to prioritize therapeutic targets for the treatment of this devastating syndrome.

 
  • References

  • 1 Guttmacher AE, Collins FS. Welcome to the genomic era. N Engl J Med 2003; 349 (10) 996-998
  • 2 Christie JD, Wurfel MM, Feng R. , et al; Trauma ALI SNP Consortium (TASC) investigators. Genome wide association identifies PPFIA1 as a candidate gene for acute lung injury risk following major trauma. PLoS One 2012; 7 (01) e28268
  • 3 Bime C, Pouladi N, Sammani S. , et al. Genome-wide association study in African Americans with acute respiratory distress syndrome identifies the selectin p ligand gene as a risk factor. Am J Respir Crit Care Med 2018; 197 (11) 1421-1432
  • 4 Ware LB. Pathophysiology of acute lung injury and the acute respiratory distress syndrome. Semin Respir Crit Care Med 2006; 27 (04) 337-349
  • 5 Park JH, Wacholder S, Gail MH. , et al. Estimation of effect size distribution from genome-wide association studies and implications for future discoveries. Nat Genet 2010; 42 (07) 570-575
  • 6 Eichler EE, Flint J, Gibson G. , et al. Missing heritability and strategies for finding the underlying causes of complex disease. Nat Rev Genet 2010; 11 (06) 446-450
  • 7 Blanco-Gómez A, Castillo-Lluva S, Del Mar Sáez-Freire M. , et al. Missing heritability of complex diseases: enlightenment by genetic variants from intermediate phenotypes. BioEssays 2016; 38 (07) 664-673
  • 8 Goldman D, Ducci F. Deconstruction of vulnerability to complex diseases: enhanced effect sizes and power of intermediate phenotypes. ScientificWorldJournal 2007; 7: 124-130
  • 9 Li Y, Huang J, Amos CI. Genetic association analysis of complex diseases incorporating intermediate phenotype information. PLoS One 2012; 7 (10) e46612
  • 10 Pingault JB, O'Reilly PF, Schoeler T, Ploubidis GB, Rijsdijk F, Dudbridge F. Using genetic data to strengthen causal inference in observational research. Nat Rev Genet 2018; 19 (09) 566-580
  • 11 Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med 2000; 342 (18) 1334-1349
  • 12 Bellingan GJ. The pulmonary physician in critical care * 6: the pathogenesis of ALI/ARDS. Thorax 2002; 57 (06) 540-546
  • 13 Matthay MA, Ware LB, Zimmerman GA. The acute respiratory distress syndrome. J Clin Invest 2012; 122 (08) 2731-2740
  • 14 Brun-Buisson C, Minelli C, Bertolini G. , et al; ALIVE Study Group. Epidemiology and outcome of acute lung injury in European intensive care units. Results from the ALIVE study. Intensive Care Med 2004; 30 (01) 51-61
  • 15 Rubenfeld GD, Caldwell E, Peabody E. , et al. Incidence and outcomes of acute lung injury. N Engl J Med 2005; 353 (16) 1685-1693
  • 16 Boyle AJ, Mac Sweeney R, McAuley DF. Pharmacological treatments in ARDS; a state-of-the-art update. BMC Med 2013; 11: 166
  • 17 Standiford TJ, Ward PA. Therapeutic targeting of acute lung injury and acute respiratory distress syndrome. Transl Res 2016; 167 (01) 183-191
  • 18 Fan E, Brodie D, Slutsky AS. Acute respiratory distress syndrome: advances in diagnosis and treatment. JAMA 2018; 319 (07) 698-710
  • 19 Hager DN. Recent advances in the management of the acute respiratory distress syndrome. Clin Chest Med 2015; 36 (03) 481-496
  • 20 Papazian L, Forel JM, Gacouin A. , et al; ACURASYS Study Investigators. Neuromuscular blockers in early acute respiratory distress syndrome. N Engl J Med 2010; 363 (12) 1107-1116
  • 21 Guérin C, Reignier J, Richard JC. , et al; PROSEVA Study Group. Prone positioning in severe acute respiratory distress syndrome. N Engl J Med 2013; 368 (23) 2159-2168
  • 22 Matthay MA, McAuley DF, Ware LB. Clinical trials in acute respiratory distress syndrome: challenges and opportunities. Lancet Respir Med 2017; 5 (06) 524-534
  • 23 Pelosi P, D'Onofrio D, Chiumello D. , et al. Pulmonary and extrapulmonary acute respiratory distress syndrome are different. Eur Respir J Suppl 2003; 42: 48s-56s
  • 24 Calfee CS, Eisner MD, Ware LB. , et al; Acute Respiratory Distress Syndrome Network, National Heart, Lung, and Blood Institute. Trauma-associated lung injury differs clinically and biologically from acute lung injury due to other clinical disorders. Crit Care Med 2007; 35 (10) 2243-2250
  • 25 Tejera P, Meyer NJ, Chen F. , et al. Distinct and replicable genetic risk factors for acute respiratory distress syndrome of pulmonary or extrapulmonary origin. J Med Genet 2012; 49 (11) 671-680
  • 26 Calfee CS, Janz DR, Bernard GR. , et al. Distinct molecular phenotypes of direct vs indirect ARDS in single-center and multicenter studies. Chest 2015; 147 (06) 1539-1548
  • 27 Reilly JP, Bellamy S, Shashaty MG. , et al. Heterogeneous phenotypes of acute respiratory distress syndrome after major trauma. Ann Am Thorac Soc 2014; 11 (05) 728-736
  • 28 Calfee CS, Delucchi K, Parsons PE, Thompson BT, Ware LB, Matthay MA. ; NHLBI ARDS Network. Subphenotypes in acute respiratory distress syndrome: latent class analysis of data from two randomised controlled trials. Lancet Respir Med 2014; 2 (08) 611-620
  • 29 Meyer NJ, Calfee CS. Novel translational approaches to the search for precision therapies for acute respiratory distress syndrome. Lancet Respir Med 2017; 5 (06) 512-523
  • 30 Jabaudon M, Blondonnet R, Audard J. , et al. Recent directions in personalised acute respiratory distress syndrome medicine. Anaesth Crit Care Pain Med 2018; 37 (03) 251-258
  • 31 Sinha P, Calfee CS. Phenotypes in acute respiratory distress syndrome: moving towards precision medicine. Curr Opin Crit Care 2019; 25 (01) 12-20
  • 32 Gong MN. Genetic epidemiology of acute respiratory distress syndrome: implications for future prevention and treatment. Clin Chest Med 2006; 27 (04) 705-724 , abstract x
  • 33 Gao L, Barnes KC. Recent advances in genetic predisposition to clinical acute lung injury. Am J Physiol Lung Cell Mol Physiol 2009; 296 (05) L713 –L725
  • 34 Flores C, Pino-Yanes MM, Casula M, Villar J. Genetics of acute lung injury: past, present and future. Minerva Anestesiol 2010; 76 (10) 860-864
  • 35 Meyer NJ, Christie JD. Genetic heterogeneity and risk of acute respiratory distress syndrome. Semin Respir Crit Care Med 2013; 34 (04) 459-474
  • 36 Reilly JP, Christie JD, Meyer NJ. Fifty years of research in ARDS. Genomic contributions and opportunities. Am J Respir Crit Care Med 2017; 196 (09) 1113-1121
  • 37 Barreiro LB, Quintana-Murci L. From evolutionary genetics to human immunology: how selection shapes host defence genes. Nat Rev Genet 2010; 11 (01) 17-30
  • 38 Wang Z, Beach D, Su L, Zhai R, Christiani DC. A genome-wide expression analysis in blood identifies pre-elafin as a biomarker in ARDS. Am J Respir Cell Mol Biol 2008; 38 (06) 724-732
  • 39 Juss J, Herre J, Begg M. , et al. Genome-wide transcription profiling in neutrophils in acute respiratory distress syndrome. Lancet 2015; 385 (Suppl 1): S55
  • 40 Kangelaris KN, Prakash A, Liu KD. , et al. Increased expression of neutrophil-related genes in patients with early sepsis-induced ARDS. Am J Physiol Lung Cell Mol Physiol 2015; 308 (11) L1102 –L1113
  • 41 Grigoryev DN, Cheranova DI, Chaudhary S, Heruth DP, Zhang LQ, Ye SQ. Identification of new biomarkers for acute respiratory distress syndrome by expression-based genome-wide association study. BMC Pulm Med 2015; 15: 95
  • 42 Simon BA, Easley RB, Grigoryev DN. , et al. Microarray analysis of regional cellular responses to local mechanical stress in acute lung injury. Am J Physiol Lung Cell Mol Physiol 2006; 291 (05) L851 –L861
  • 43 Wan QQ, Wu D, Ye QF. Candidate genes as biomarkers in lipopolysaccharide-induced acute respiratory distress syndrome based on mRNA expression profile by next-generation RNA-seq analysis. BioMed Res Int 2018; 2018: 4384797
  • 44 Grigoryev DN, Ma SF, Irizarry RA, Ye SQ, Quackenbush J, Garcia JG. Orthologous gene-expression profiling in multi-species models: search for candidate genes. Genome Biol 2004; 5 (05) R34
  • 45 Marshall RP, Webb S, Bellingan GJ. , et al. Angiotensin converting enzyme insertion/deletion polymorphism is associated with susceptibility and outcome in acute respiratory distress syndrome. Am J Respir Crit Care Med 2002; 166 (05) 646-650
  • 46 Jerng JS, Yu CJ, Wang HC, Chen KY, Cheng SL, Yang PC. Polymorphism of the angiotensin-converting enzyme gene affects the outcome of acute respiratory distress syndrome. Crit Care Med 2006; 34 (04) 1001-1006
  • 47 Su L, Zhai R, Sheu CC. , et al. Genetic variants in the angiopoietin-2 gene are associated with increased risk of ARDS. Intensive Care Med 2009; 35 (06) 1024-1030
  • 48 Meyer NJ, Li M, Feng R. , et al. ANGPT2 genetic variant is associated with trauma-associated acute lung injury and altered plasma angiopoietin-2 isoform ratio. Am J Respir Crit Care Med 2011; 183 (10) 1344-1353
  • 49 Lin Z, Pearson C, Chinchilli V. , et al. Polymorphisms of human SP-A, SP-B, and SP-D genes: association of SP-B Thr131Ile with ARDS. Clin Genet 2000; 58 (03) 181-191
  • 50 Gong MN, Wei Z, Xu LL, Miller DP, Thompson BT, Christiani DC. Polymorphism in the surfactant protein-B gene, gender, and the risk of direct pulmonary injury and ARDS. Chest 2004; 125 (01) 203-211
  • 51 O'Mahony DS, Glavan BJ, Holden TD. , et al. Inflammation and immune-related candidate gene associations with acute lung injury susceptibility and severity: a validation study. PLoS One 2012; 7 (12) e51104
  • 52 Meyer NJ, Feng R, Li M. , et al. IL1RN coding variant is associated with lower risk of acute respiratory distress syndrome and increased plasma IL-1 receptor antagonist. Am J Respir Crit Care Med 2013; 187 (09) 950-959
  • 53 Tejera P, Wang Z, Zhai R. , et al. Genetic polymorphisms of peptidase inhibitor 3 (elafin) are associated with acute respiratory distress syndrome. Am J Respir Cell Mol Biol 2009; 41 (06) 696-704
  • 54 Tejera P, O'Mahony DS, Owen CA. , et al. Functional characterization of polymorphisms in the peptidase inhibitor 3 (elafin) gene and validation of their contribution to risk of acute respiratory distress syndrome. Am J Respir Cell Mol Biol 2014; 51 (02) 262-272
  • 55 Meyer NJ. Beyond single-nucleotide polymorphisms: genetics, genomics, and other 'omic approaches to acute respiratory distress syndrome. Clin Chest Med 2014; 35 (04) 673-684
  • 56 Flores C, Pino-Yanes MdelM, Villar J. A quality assessment of genetic association studies supporting susceptibility and outcome in acute lung injury. Crit Care 2008; 12 (05) R130
  • 57 Acosta-Herrera M, Pino-Yanes M, Perez-Mendez L, Villar J, Flores C. Assessing the quality of studies supporting genetic susceptibility and outcomes of ARDS. Front Genet 2014; 5: 20
  • 58 Famous KR, Delucchi K, Ware LB. , et al; ARDS Network. Acute respiratory distress syndrome subphenotypes respond differently to randomized fluid management strategy. Am J Respir Crit Care Med 2017; 195 (03) 331-338
  • 59 Calfee CS, Delucchi KL, Sinha P. , et al; Irish Critical Care Trials Group. Acute respiratory distress syndrome subphenotypes and differential response to simvastatin: secondary analysis of a randomised controlled trial. Lancet Respir Med 2018; 6 (09) 691-698
  • 60 Jansen RC, Nap JP. Genetical genomics: the added value from segregation. Trends Genet 2001; 17 (07) 388-391
  • 61 Westra HJ, Peters MJ, Esko T. , et al. Systematic identification of trans eQTLs as putative drivers of known disease associations. Nat Genet 2013; 45 (10) 1238-1243
  • 62 Gibson G, Powell JE, Marigorta UM. Expression quantitative trait locus analysis for translational medicine. Genome Med 2015; 7 (01) 60
  • 63 Gottesman II, Gould TD. The endophenotype concept in psychiatry: etymology and strategic intentions. Am J Psychiatry 2003; 160 (04) 636-645
  • 64 Meyer-Lindenberg A, Weinberger DR. Intermediate phenotypes and genetic mechanisms of psychiatric disorders. Nat Rev Neurosci 2006; 7 (10) 818-827
  • 65 Leuchter AF, Hunter AM, Krantz DE, Cook IA. Intermediate phenotypes and biomarkers of treatment outcome in major depressive disorder. Dialogues Clin Neurosci 2014; 16 (04) 525-537
  • 66 Castellanos-Martín A, Castillo-Lluva S, Sáez-Freire Mdel M. , et al. Unraveling heterogeneous susceptibility and the evolution of breast cancer using a systems biology approach. Genome Biol 2015; 16: 40
  • 67 Nava-Gonzalez EJ, Gallegos-Cabriales EC, Leal-Berumen I, Bastarrachea RA. Mini-review: the contribution of intermediate phenotypes to gxe effects on disorders of body composition in the new OMICS era. Int J Environ Res Public Health 2017; 14 (09) E1079
  • 68 Yadav H, Kor DJ. Platelets in the pathogenesis of acute respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol 2015; 309 (09) L915-L923
  • 69 Zarbock A, Ley K. The role of platelets in acute lung injury (ALI). Front Biosci 2009; 14: 150-158
  • 70 Bone RC, Francis PB, Pierce AK. Intravascular coagulation associated with the adult respiratory distress syndrome. Am J Med 1976; 61 (05) 585-589
  • 71 Tomashefski Jr JF, Davies P, Boggis C, Greene R, Zapol WM, Reid LM. The pulmonary vascular lesions of the adult respiratory distress syndrome. Am J Pathol 1983; 112 (01) 112-126
  • 72 Bozza FA, Shah AM, Weyrich AS, Zimmerman GA. Amicus or adversary: platelets in lung biology, acute injury, and inflammation. Am J Respir Cell Mol Biol 2009; 40 (02) 123-134
  • 73 Zarbock A, Singbartl K, Ley K. Complete reversal of acid-induced acute lung injury by blocking of platelet-neutrophil aggregation. J Clin Invest 2006; 116 (12) 3211-3219
  • 74 Weyrich AS, Zimmerman GA. Platelets in lung biology. Annu Rev Physiol 2013; 75: 569-591
  • 75 Katz JN, Kolappa KP, Becker RC. Beyond thrombosis: the versatile platelet in critical illness. Chest 2011; 139 (03) 658-668
  • 76 Hill RN, Shibel EM, Spragg RG, Moser KM. Adult respiratory distress syndrome: early predictors of mortality. Trans Am Soc Artif Intern Organs 1975; 21: 199-205
  • 77 Wang T, Liu Z, Wang Z. , et al. Thrombocytopenia is associated with acute respiratory distress syndrome mortality: an international study. PLoS One 2014; 9 (04) e94124
  • 78 Wei Y, Wang Z, Su L. , et al. Platelet count mediates the contribution of a genetic variant in LRRC16A to ARDS risk. Chest 2015; 147 (03) 607-617
  • 79 Wei Y, Tejera P, Wang Z. , et al. A missense genetic variant in LRRC16A/CARMIL1 improves acute respiratory distress syndrome survival by attenuating platelet count decline. Am J Respir Crit Care Med 2017; 195 (10) 1353-1361
  • 80 Reilly JP, Wang F, Jones TK. , et al. Plasma angiopoietin-2 as a potential causal marker in sepsis-associated ARDS development: evidence from Mendelian randomization and mediation analysis. Intensive Care Med 2018; 44 (11) 1849-1858
  • 81 Schafer DA, Jennings PB, Cooper JA. Dynamics of capping protein and actin assembly in vitro: uncapping barbed ends by polyphosphoinositides. J Cell Biol 1996; 135 (01) 169-179
  • 82 Yang C, Pring M, Wear MA. , et al. Mammalian CARMIL inhibits actin filament capping by capping protein. Dev Cell 2005; 9 (02) 209-221
  • 83 Fox JE. Cytoskeletal proteins and platelet signaling. Thromb Haemost 2001; 86 (01) 198-213
  • 84 Hartwig JH, Barkalow K, Azim A, Italiano J. The elegant platelet: signals controlling actin assembly. Thromb Haemost 1999; 82 (02) 392-398
  • 85 Kor DJ, Carter RE, Park PK. , et al; US Critical Illness and Injury Trials Group: Lung Injury Prevention with Aspirin Study Group (USCIITG: LIPS-A). Effect of aspirin on development of ARDS in at-risk patients presenting to the emergency department: the LIPS-a randomized clinical trial. JAMA 2016; 315 (22) 2406-2414