期刊论文详细信息
BMC Medicine
Incorporating genetics into the identification and treatment of Idiopathic Pulmonary Fibrosis
David A. Schwartz1  Marvin I. Schwarz1  Ivana V. Yang1  Susan K. Mathai1 
[1] Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, 12631 East 17th Avenue, Aurora 80045, CO, USA
关键词: Telomere;    TERT;    TERC;    Pulmonary fibrosis;    Mucin;    MUC5B;    Idiopathic pulmonary fibrosis;    Familial pulmonary fibrosis;    DSP;   
Others  :  1227204
DOI  :  10.1186/s12916-015-0434-0
 received in 2015-05-02, accepted in 2015-07-24,  发布年份 2015
PDF
【 摘 要 】

Background

Idiopathic pulmonary fibrosis, the most common form of idiopathic interstitial pneumonia, is characterized by progressive, irreversible scarring of the lung parenchyma. Idiopathic pulmonary fibrosis has a poor prognosis, and there are no medical therapies available that have been shown to improve survival. It is usually sporadic, but there is evidence of familial clustering of pulmonary fibrosis, suggesting a genetic basis for this disease. More recently, studies have confirmed that specific genetic variants are associated with both familial and sporadic forms of pulmonary fibrosis.

Discussion

Although there are common and rare genetic variants that have been associated with the risk of developing pulmonary fibrosis, the genotyping of patients is not a generally accepted strategy. Better understanding of the interplay between genetic risk and environmental exposure is likely needed to inform both treatment and disease prevention. Several identified disease-associated genetic variants have implications for disease progression and survival, but systematic studies of known genetic variants and their influence on therapeutic efficacy are lacking. Future investigations should focus on understanding phenotypic differences between patients carrying different risk alleles, and clinical studies should be designed to control for the influence of different genetic risk variants on patient outcomes.

Summary

Inherited genetic factors play a significant role in the risk of developing pulmonary fibrosis. Future studies will be needed to characterize patient phenotypes and to understand how these genetic factors will influence clinical decision-making for both diagnosis and treatment of idiopathic pulmonary fibrosis.

【 授权许可】

   
2015 Mathai et al.

【 预 览 】
附件列表
Files Size Format View
20150928022728431.pdf 669KB PDF download
Fig. 1. 41KB Image download
【 图 表 】

Fig. 1.

【 参考文献 】
  • [1]Raghu G, Weycker D, Edelsberg J, Bradford WZ, Oster G. Incidence and prevalence of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2006; 174:810-6.
  • [2]Olson AL, Swigris JJ, Lezotte DC, Norris JM, Wilson CG, Brown KK. Mortality from pulmonary fibrosis increased in the United States from 1992 to 2003. Am J Respir Crit Care Med. 2007; 176:277-84.
  • [3]King TE, Pardo A, Selman M. Idiopathic pulmonary fibrosis. Lancet. 2011; 378:1949-61.
  • [4]Fingerlin TE, Murphy E, Zhang W, Peljto AL, Brown KK, Steele MP et al.. Genome-wide association study identifies multiple susceptibility loci for pulmonary fibrosis. Nat Genet. 2013; 45:613-20.
  • [5]García-Sancho C, Buendía-Roldán I, Fernández-Plata MR, Navarro C, Pérez-Padilla R, Vargas MH et al.. Familial pulmonary fibrosis is the strongest risk factor for idiopathic pulmonary fibrosis. Respir Med. 2011; 105:1902-7.
  • [6]Seibold MA, Wise A, Speer M, Steele M, Brown K, Lloyd JE et al.. A common MUC5B promoter polymorphism and pulmonary fibrosis. N Engl J Med. 2011; 364:1503-12.
  • [7]Bonanni PP, Frymoyer JW, Jacox RF. A family study of idiopathic pulmonary fibrosis. A possible dysproteinemic and genetically determined disease. Am J Med. 1965; 39:411-21.
  • [8]Bitterman PB, Rennard SI, Keogh BA, Wewers MD, Adelberg S, Crystal RG. Familial idiopathic pulmonary fibrosis. Evidence of lung inflammation in unaffected family members. N Engl J Med. 1986; 314:1343-7.
  • [9]Javaheri S, Lederer DH, Pella JA, Mark GJ, Levine BW. Idiopathic pulmonary fibrosis in monozygotic twins: the importance of genetic predisposition. Chest. 1980; 78:591-4.
  • [10]Solliday N, Williams J, Gaensler E, Coutu R, Carringon C. Familial chronic interstitial pneumonia. Am Rev Respir Dis. 1973; 108:193-204.
  • [11]Nogee LM, Dunbar AE, Wert SE, Askin F, Hamvas A, Whitsett JA. A mutation in the surfactant protein C gene associated with familial interstitial lung disease. N Engl J Med. 2001; 344:573-9.
  • [12]Lawson WE, Grant SW, Ambrosini V, Womble KE, Dawson EP, Lane KB et al.. Genetic mutations in surfactant protein C are a rare cause of sporadic cases of IPF. Thorax. 2004; 59:977-80.
  • [13]Fernandez BA, Fox G, Bhatia R, Sala E, Noble B, Denic N et al.. A Newfoundland cohort of familial and sporadic idiopathic pulmonary fibrosis patients: clinical and genetic features. Respir Res. 2012; 13:64. BioMed Central Full Text
  • [14]Maitra M, Wang Y, Gerard RD, Mendelson CR, Garcia CK. Surfactant protein A2 mutations associated with pulmonary fibrosis lead to protein instability and endoplasmic reticulum stress. J Biol Chem. 2010; 285:22103-13.
  • [15]Wang Y, Kuan PJ, Xing C, Cronkhite JT, Torres F, Rosenblatt RL et al.. Genetic defects in surfactant protein A2 are associated with pulmonary fibrosis and lung cancer. Am J Hum Genet. 2009; 84:52-9.
  • [16]Gochuico BR, Huizing M, Golas GA, Scher CD, Tsokos M, Denver SD et al.. Interstitial lung disease and pulmonary fibrosis in Hermansky-Pudlak syndrome type 2, an adaptor protein-3 complex disease. Mol Med. 2012; 18:56-64.
  • [17]Wei ML. Hermansky-Pudlak syndrome: a disease of protein trafficking and organelle function. Pigment Cell Res. 2006; 19:19-42.
  • [18]Vulliamy TJ, Marrone A, Knight SW, Walne A, Mason PJ, Dokal I. Mutations in dyskeratosis congenita: their impact on telomere length and the diversity of clinical presentation. Blood. 2011; 107:2680-5.
  • [19]Armanios M, Chen J-L, Chang Y-PC, Brodsky RA, Hawkins A, Griffin CA et al.. Haploinsufficiency of telomerase reverse transcriptase leads to anticipation in autosomal dominant dyskeratosis congenita. Proc Natl Acad Sci U S A. 2005; 102:15960-4.
  • [20]Armanios MY, Chen JJ, Cogan JD, Alder JK, Ingersoll RG, Markin C et al.. Telomerase mutations in families with idiopathic pulmonary fibrosis. N Engl J Med. 2007; 356:1317-26.
  • [21]Armanios M, Blackburn EH. The telomere syndromes. Nat Rev Genet. 2012; 13:693-704.
  • [22]Tsakiri KD, Cronkhite JT, Kuan PJ, Xing C, Raghu G, Weissler JC et al.. Adult-onset pulmonary fibrosis caused by mutations in telomerase. Proc Natl Acad Sci U S A. 2007; 104:7552-7.
  • [23]Cogan JD, Kropski JA, Zhao M, Mitchell DB, Rives L, Markin C et al.. Rare variants in RTEL1 are associated with familial interstitial pneumonia. Am J Respir Crit Care Med. 2015; 191:646-55.
  • [24]Stuart BD, Choi J, Zaidi S, Xing C, Holohan B, Chen R et al.. Exome sequencing links mutations in PARN and RTEL1 with familial pulmonary fibrosis and telomere shortening. Nat Genet. 2015; 47:512-7.
  • [25]Seibold MA, Smith RW, Urbanek C, Groshong SD, Cosgrove GP, Brown KK et al.. The idiopathic pulmonary fibrosis honeycomb cyst contains a mucocilary pseudostratified epithelium. PLoS One. 2013; 8:e58658.
  • [26]Zhang Y, Noth I, Garcia JGN, Kaminski N. A variant in the promoter of MUC5B and idiopathic pulmonary fibrosis NT5E mutations and arterial calcifications. N Engl J Med. 2011; 364:1576-7.
  • [27]Stock CJ, Sato H, Fonseca C, Banya WAS, Molyneaux PL, Adamali H et al.. Mucin 5B promoter polymorphism is associated with idiopathic pulmonary fibrosis but not with development of lung fibrosis in systemic sclerosis or sarcoidosis. Thorax. 2013; 68:436-41.
  • [28]Noth I, Zhang Y, Ma S-F, Flores C, Barber M, Huang Y et al.. Genetic variants associated with idiopathic pulmonary fibrosis susceptibility and mortality: a genome-wide association study. Lancet Respir Med. 2013; 1:309-17.
  • [29]Borie R, Crestani B, Dieude P, Nunes H, Allanore Y, Kannengiesser C et al.. The MUC5B variant is associated with idiopathic pulmonary fibrosis but not with systemic sclerosis interstitial lung disease in the European Caucasian population. PLoS One. 2013; 8:e70621.
  • [30]Wei R, Li C, Zhang M, Jones-Hall YL, Myers JL, Noth I et al.. Association between MUC5B and TERT polymorphisms and different interstitial lung disease phenotypes. Transl Res. 2014; 163:494-502.
  • [31]Horimasu Y, Ohshimo S, Bonella F, Tanaka S, Ishikawa N, Hattori N et al.. MUC 5 B promoter polymorphism in Japanese patients with idiopathic pulmonary fibrosis. Respirology. 2015; 20:439-44.
  • [32]Hunninghake GM, Hatabu H, Okajima Y, Gao W, Dupuis J, Latourelle JC et al.. MUC5B promoter polymorphism and interstitial lung abnormalities. N Engl J Med. 2013; 368:2192-200.
  • [33]Peljto AL, Zhang Y, Fingerlin TE, Ma S-F, Garcia JGN, Richards TJ et al.. Association between the MUC5B promoter polymorphism and survival in patients with idiopathic pulmonary fibrosis. JAMA. 2013; 309:2232-9.
  • [34]Yang I V, Fingerlin TE, Evans CM, Schwarz MI, Schwartz DA. Genetic variants and outcome of idiopathic pulmonary fibrosis. Ann Am Thorac Soc. 2015; in press.
  • [35]Peljto AL, Selman M, Kim DS, Murphy E, Tucker L, Pardo A et al.. The muc5b promoter polymorphism is associated with idiopathic pulmonary fibrosis in a Mexican cohort but is rare among Asian ancestries. Chest. 2015; 147:460-4.
  • [36]Wang C, Zhuang Y, Guo W, Cao L, Zhang H, Xu L et al.. Mucin 5B promoter polymorphism is associated with susceptibility to interstitial lung diseases in Chinese males. PLoS One. 2014; 9:e104919.
  • [37]Ley B, Collard HR. Epidemiology of idiopathic pulmonary fibrosis. Clin Epidemiol. 2013; 5:483-92.
  • [38]dbSNP, NCBI:. http://www. ncbi.nlm.nih.gov/SNP/snp_ref.cgi?rs=35705950 webcite
  • [39]Mathai SK, Schwartz DA, Warg LA. Genetic susceptibility and pulmonary fibrosis. Curr Opin Pulm Med. 2014; 20:429-35.
  • [40]Armanios M. Telomerase and idiopathic pulmonary fibrosis. Mutat Res. 2013; 730:52-8.
  • [41]De Leon AD, Cronkhite JT, Katzenstein ALA, Godwin JD, Raghu G, Glazer CS et al.. Telomere lengths, pulmonary fibrosis and telomerase (TERT) mutations. PLoS One. 2010; 5:e10680.
  • [42]Silhan LL, Shah PD, Chambers DC, Snyder LD, Riise GC, Wagner CL et al.. Lung transplantation in telomerase mutation carriers with pulmonary fibrosis. Eur Respir J. 2014; 44:178-87.
  • [43]O’Dwyer DN, Armstrong ME, Trujillo G, Cooke G, Keane MP, Fallon PG et al.. The Toll-like receptor 3 L412F polymorphism and disease progression in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2013; 188:1442-50.
  • [44]Stuart BD, Lee JS, Kozlitina J, Noth I, Devine MS, Glazer CS et al.. Effect of telomere length on survival in patients with idiopathic pulmonary fibrosis: an observational cohort study with independent validation. Lancet Respir Med. 2014; 2:557-65.
  • [45]King TE, Bradford WZ, Castro-Bernardini S, Fagan EA, Glaspole I, Glassberg MK et al.. A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med. 2014; 370:2083-92.
  • [46]Richeldi L, du Bois RM, Raghu G, Azuma A, Brown KK, Costabel U et al.. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med. 2014; 370:2071-82.
  • [47]Schaffer JM, Singh SK, Reitz BA, Zamanian RT, Mallidi HR. Single- vs double-lung transplantation in patients with chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis since the implementation of lung allocation based on medical need. JAMA. 2015; 313:936.
  • [48]King TE, Albera C, Bradford WZ, Costabel U, Du Bois RM, Leff JA et al.. All-cause mortality rate in patients with idiopathic pulmonary fibrosis: implications for the design and execution of clinical trials. Am J Respir Crit Care Med. 2014; 189:825-31.
  • [49]Nathan SD, Meyer KC. IPF clinical trial design and endpoints. Curr Opin Pulm Med. 2014; 20:463-71.
  • [50]El-Chemaly S, Ziegler SG, Calado RT, Wilson KA, Wu HP, Haughey M, Peterson NR, Young NS, Gahl WA, Moss J, Gochuico BR. Natural history of pulmonary fibrosis in two subjects with the same telomerase mutation. Chest. 2011; 139:1203-1209.
  • [51]Kropski JA, Pritchett JM, Zoz DF, Crossno PF, Markin C, Garnett ET et al.. Extensive phenotyping of individuals at risk for familial interstitial pneumonia reveals clues to the pathogenesis of interstitial lung disease. Am J Respir Crit Care Med. 2015; 191:417-26.
  • [52]Steele MP, Speer MC, Loyd JE, Brown KK, Herron A, Slifer SH et al.. Clinical and pathologic features of familial interstitial pneumonia. Am J Respir Crit Care Med. 2005; 172:1146-52.
  • [53]Antonarakis SE, Chakravarti A, Cohen JC, Hardy J. Mendelian disorders and multifactorial traits: the big divide or one for all? Nat Rev Genet. 2010; 11:380-4.
  文献评价指标  
  下载次数:19次 浏览次数:12次