Skip to main content
Log in

Genome-wide study identifies two loci associated with lung function decline in mild to moderate COPD

  • Original Investigation
  • Published:
Human Genetics Aims and scope Submit manuscript

Abstract

Accelerated lung function decline is a key COPD phenotype; however, its genetic control remains largely unknown. We performed a genome-wide association study using the Illumina Human660W-Quad v.1_A BeadChip. Generalized estimation equations were used to assess genetic contributions to lung function decline over a 5-year period in 4,048 European American Lung Health Study participants with largely mild COPD. Genotype imputation was performed using reference HapMap II data. To validate regions meeting genome-wide significance, replication of top SNPs was attempted in independent cohorts. Three genes (TMEM26, ANK3 and FOXA1) within the regions of interest were selected for tissue expression studies using immunohistochemistry. Two intergenic SNPs (rs10761570, rs7911302) on chromosome 10 and one SNP on chromosome 14 (rs177852) met genome-wide significance after Bonferroni. Further support for the chromosome 10 region was obtained by imputation, the most significantly associated imputed SNPs (rs10761571, rs7896712) being flanked by observed markers rs10761570 and rs7911302. Results were not replicated in four general population cohorts or a smaller cohort of subjects with moderate to severe COPD; however, we show novel expression of genes near regions of significantly associated SNPS, including TMEM26 and FOXA1 in airway epithelium and lung parenchyma, and ANK3 in alveolar macrophages. Levels of expression were associated with lung function and COPD status. We identified two novel regions associated with lung function decline in mild COPD. Genes within these regions were expressed in relevant lung cells and their expression related to airflow limitation suggesting they may represent novel candidate genes for COPD susceptibility.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  • Anthonisen NR, Connett JE, Kiley JP, Altose MD, Bailey WC, Buist AS, Conway WA Jr, Enright PL, Kanner RE, O’Hara P (1994) Effects of smoking intervention and the use of an inhaled anticholinergic bronchodilator on the rate of decline of FEV1. The Lung Health Study. JAMA 272:1497–1505

    CAS  Google Scholar 

  • Anthonisen NR, Lindgren PG, Tashkin DP, Kanner RE, Scanlon PD, Connett JE (2005) Bronchodilator response in the lung health study over 11 years. Eur Respir J 26:45–51

    Article  PubMed  CAS  Google Scholar 

  • Bakke PS, Zhu G, Gulsvik A, Kong X, Agusti AG, Calverley PM, Donner CF, Levy RD, Make BJ, Pare PD, Rennard SI, Vestbo J, Wouters EF, Anderson W, Lomas DA, Silverman EK, Pillai SG (2011) Candidate genes for COPD in two large data sets. Eur Respir J 37:255–263

    Article  PubMed  CAS  Google Scholar 

  • Besnard V, Wert SE, Kaestner KH, Whitsett JA (2005) Stage-specific regulation of respiratory epithelial cell differentiation by Foxa1. Am J Physiol Lung Cell Mol Physiol 289:L750–L759

    Article  PubMed  CAS  Google Scholar 

  • Celli BR, Thomas NE, Anderson JA, Ferguson GT, Jenkins CR, Jones PW, Vestbo J, Knobil K, Yates JC, Calverley PM (2008) Effect of pharmacotherapy on rate of decline of lung function in chronic obstructive pulmonary disease: results from the TORCH study. Am J Respir Crit Care Med 178:332–338

    Article  PubMed  Google Scholar 

  • Cho MH, Boutaoui N, Klanderman BJ, Sylvia JS, Ziniti JP, Hersh CP, Demeo DL, Hunninghake GM, Litonjua AA, Sparrow D, Lange C, Won S, Murphy JR, Beaty TH, Regan EA, Make BJ, Hokanson JE, Crapo JD, Kong X, Anderson WH, Tal-Singer R, Lomas DA, Bakke P, Gulsvik A, Pillai SG, Silverman EK (2010) Variants in FAM13A are associated with chronic obstructive pulmonary disease. Nat Genet 42:200–202

    Article  PubMed  CAS  Google Scholar 

  • Clarke DL, Clifford RL, Jindarat S, Proud D, Pang L, Belvisi M, Knox AJ (2010) TNFalpha and IFNgamma synergistically enhance transcriptional activation of CXCL10 in human airway smooth muscle cells via STAT-1, NF-kappaB, and the transcriptional coactivator CREB-binding protein. J Biol Chem 285:29101–29110

    Article  PubMed  CAS  Google Scholar 

  • Connett JE, Kusek JW, Bailey WC, O’Hara P, Wu M (1993) Design of the Lung Health Study: a randomized clinical trial of early intervention for chronic obstructive pulmonary disease. Control Clin Trials 14:3S–19S

    Article  PubMed  CAS  Google Scholar 

  • Gottlieb DJ, Wilk JB, Harmon M, Evans JC, Joost O, Levy D, O’Connor GT, Myers RH (2001) Heritability of longitudinal change in lung function. The Framingham study. Am J Respir Crit Care Med 164:1655–1659

    PubMed  CAS  Google Scholar 

  • Hansel NN, Gao L, Rafaels NM, Mathias RA, Neptune ER, Tankersley C, Grant AV, Connett J, Beaty TH, Wise RA, Barnes KC (2009) Leptin receptor polymorphisms and lung function decline in COPD. Eur Respir J 34:103–110

    Article  PubMed  CAS  Google Scholar 

  • Hansel NN, Sidhaye V, Rafaels NM, Gao L, Gao P, Williams R, Connett JE, Beaty TH, Mathias RA, Wise RA, King LS, Barnes, KC (2010) Aquaporin 5 polymorphisms and rate of lung function decline in chronic obstructive pulmonary disease. PLoS One 5:e14226

    Google Scholar 

  • He JQ, Ruan J, Connett JE, Anthonisen NR, Pare PD, Sandford AJ (2002) Antioxidant gene polymorphisms and susceptibility to a rapid decline in lung function in smokers. Am J Respir Crit Care Med 166:323–328

    Article  PubMed  Google Scholar 

  • He JQ, Connett JE, Anthonisen NR, Sandford AJ (2003) Polymorphisms in the IL13, IL13RA1, and IL4RA genes and rate of decline in lung function in smokers. Am J Respir Cell Mol Biol 28:379–385

    Article  PubMed  CAS  Google Scholar 

  • He JQ, Connett JE, Anthonisen NR, Pare PD, Sandford AJ (2004) Glutathione S-transferase variants and their interaction with smoking on lung function. Am J Respir Crit Care Med 170:388–394

    Article  PubMed  Google Scholar 

  • He JQ, Burkett K, Connett JE, Anthonisen NR, Pare PD, Sandford AJ (2006) Interferon gamma polymorphisms and their interaction with smoking are associated with lung function. Hum Genet 119:365–375

    Article  PubMed  CAS  Google Scholar 

  • Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, Pare PD (2004) The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med 350:2645–2653

    Article  PubMed  CAS  Google Scholar 

  • Hoock TC, Peters LL, Lux SE (1997) Isoforms of ankyrin-3 that lack the NH2-terminal repeats associate with mouse macrophage lysosomes. J Cell Biol 136:1059–1070

    Article  PubMed  CAS  Google Scholar 

  • Kanner RE (1996) Early intervention in chronic obstructive pulmonary disease. A review of the Lung Health Study results. Med Clin North Am 80:523–547

    PubMed  CAS  Google Scholar 

  • Kanner RE, Connett JE, Williams DE, Buist AS (1999) Effects of randomized assignment to a smoking cessation intervention and changes in smoking habits on respiratory symptoms in smokers with early chronic obstructive pulmonary disease: the Lung Health Study. Am J Med 106:410–416

    Article  PubMed  CAS  Google Scholar 

  • Khoury MJ, Beaty TH, Tockman MS, Self SG, Cohen BH (1985) Familial aggregation in chronic obstructive pulmonary disease: use of the loglinear model to analyze intermediate environmental and genetic risk factors. Genet Epidemiol 2:155–166

    Article  PubMed  CAS  Google Scholar 

  • Kizhatil K, Yoon W, Mohler PJ, Davis LH, Hoffman JA, Bennett V (2007) Ankyrin-G and beta2-spectrin collaborate in biogenesis of lateral membrane of human bronchial epithelial cells. J Biol Chem 282:2029–2037

    Article  PubMed  CAS  Google Scholar 

  • Mannino DM, Homa DM, Akinbami LJ, Ford ES, Redd SC (2002) Chronic obstructive pulmonary disease surveillance—United States, 1971–2000. Respir Care 47:1184–1199

    PubMed  Google Scholar 

  • Minino AM (2011) Death in the United States, 2009. NCHS Data Brief, pp 1–8

  • Molfino NA (2007) Genetic predisposition to accelerated decline of lung function in COPD. Int. J. Chron. Obstruct. Pulmon. Dis. 2:117–119

    PubMed  Google Scholar 

  • Murray CJ, Lopez AD (1997) Alternative projections of mortality and disability by cause 1990–2020: global Burden of Disease Study. Lancet 349:1498–1504

    Article  PubMed  CAS  Google Scholar 

  • Newton-Cheh C, Johnson T, Gateva V, Tobin MD, Bochud M, Coin L, Najjar SS, Zhao JH, Heath SC, Eyheramendy S, Papadakis K, Voight BF, Scott LJ, Zhang F, Farrall M, Tanaka T, Wallace C, Chambers JC, Khaw KT, Nilsson P, van der HP, Polidoro S, Grobbee DE, Onland-Moret NC, Bots ML, Wain LV, Elliott KS, Teumer A, Luan J, Lucas G, Kuusisto J, Burton PR, Hadley D, McArdle WL, Brown M, Dominiczak A, Newhouse SJ, Samani NJ, Webster J, Zeggini E, Beckmann JS, Bergmann S, Lim N, Song K, Vollenweider P, Waeber G, Waterworth DM, Yuan X, Groop L, Orho-Melander M, Allione A, Di GA, Guarrera S, Panico S, Ricceri F, Romanazzi V, Sacerdote C, Vineis P, Barroso I, Sandhu MS, Luben RN, Crawford GJ, Jousilahti P, Perola M, Boehnke M, Bonnycastle LL, Collins FS, Jackson AU, Mohlke KL, Stringham HM, Valle TT, Willer CJ, Bergman RN, Morken MA, Doring A, Gieger C, Illig T, Meitinger T, Org E, Pfeufer A, Wichmann HE, Kathiresan S, Marrugat J, O’Donnell CJ, Schwartz SM, Siscovick DS, Subirana I, Freimer NB, Hartikainen AL, McCarthy MI, O’Reilly PF, Peltonen L, Pouta A, de Jong PE, Snieder H, van Gilst WH, Clarke R, Goel A, Hamsten A, Peden JF, Seedorf U, Syvanen AC, Tognoni G, Lakatta EG, Sanna S, Scheet P, Schlessinger D, Scuteri A, Dorr M, Ernst F, Felix SB, Homuth G, Lorbeer R, Reffelmann T, Rettig R, Volker U, Galan P, Gut IG, Hercberg S, Lathrop GM, Zelenika D, Deloukas P, Soranzo N, Williams FM, Zhai G, Salomaa V, Laakso M, Elosua R, Forouhi NG, Volzke H, Uiterwaal CS, van der Schouw YT, Numans ME, Matullo G, Navis G, Berglund G, Bingham SA, Kooner JS, Connell JM, Bandinelli S, Ferrucci L, Watkins H, Spector TD, Tuomilehto J, Altshuler D, Strachan DP, Laan M, Meneton P, Wareham NJ, Uda M, Jarvelin MR, Mooser V, Melander O, Loos RJ, Elliott P, Abecasis GR, Caulfield M, Munroe PB (2009) Genome-wide association study identifies eight loci associated with blood pressure. Nat Genet 41:666–676

  • Pillai SG, Ge D, Zhu G, Kong X, Shianna KV, Need AC, Feng S, Hersh CP, Bakke P, Gulsvik A, Ruppert A, Lodrup Carlsen KC, Roses A, Anderson W, Rennard SI, Lomas DA, Silverman EK, Goldstein DB (2009) A genome-wide association study in chronic obstructive pulmonary disease (COPD): identification of two major susceptibility loci. PLoS Genet 5:e1000421

    Google Scholar 

  • Rennard SI, Vestbo J (2006) COPD: the dangerous underestimate of 15 %. Lancet 367:1216–1219

    Article  PubMed  Google Scholar 

  • Sandford AJ, Chagani T, Weir TD, Connett JE, Anthonisen NR, Pare PD (2001) Susceptibility genes for rapid decline of lung function in the lung health study. Am J Respir Crit Care Med 163:469–473

    PubMed  CAS  Google Scholar 

  • Siedlinski M, Boezen HM, Boer JM, Smit HA, Postma DS (2009a) ABCC1 polymorphisms contribute to level and decline of lung function in two population-based cohorts. Pharmacogenet Genomics 19:675–684

    Article  PubMed  CAS  Google Scholar 

  • Siedlinski M, Postma DS, Boer JM, van der SG, Schouten JP, Smit HA, Boezen HM (2009b) Level and course of FEV1 in relation to polymorphisms in NFE2L2 and KEAP1 in the general population. Respir Res 10:73

    Article  PubMed  Google Scholar 

  • Song L, Wei X, Zhang B, Luo X, Liu J, Feng Y, Xiao X (2009a) Role of Foxa1 in regulation of bcl2 expression during oxidative-stress-induced apoptosis in A549 type II pneumocytes. Cell Stress Chaperones 14:417–425

    Article  PubMed  CAS  Google Scholar 

  • Song L, Zhang B, Feng Y, Luo X, Wei X, Xiao X (2009b) A role for forkhead box A1 in acute lung injury. Inflammation 32:322–332

    Article  PubMed  CAS  Google Scholar 

  • Tuder RM, Yoshida T, Arap W, Pasqualini R, Petrache I (2006) State of the art. Cellular and molecular mechanisms of alveolar destruction in emphysema: an evolutionary perspective. Proc Am Thorac Soc 3:503–510

    Article  PubMed  CAS  Google Scholar 

  • van Diemen CC, Postma DS, Vonk JM, Bruinenberg M, Schouten JP, Boezen HM (2005) A disintegrin and metalloprotease 33 polymorphisms and lung function decline in the general population. Am.J Respir. Crit Care Med 172:329–333

    Article  Google Scholar 

  • van Diemen CC, Postma DS, Siedlinski M, Blokstra A, Smit HA, Boezen HM (2011) Genetic variation in TIMP1 but not MMPs predict excess FEV1 decline in two general population-based cohorts. Respir Res 12:57

    Article  PubMed  Google Scholar 

  • Vestbo J, Edwards LD, Scanlon PD, Yates JC, Agusti A, Bakke P, Calverley PM, Celli B, Coxson HO, Crim C, Lomas DA, Macnee W, Miller BE, Silverman EK, Tal-Singer R, Wouters E, Rennard SI (2011) Changes in forced expiratory volume in 1 second over time in COPD. N Engl J Med 365:1184–1192

    Article  PubMed  CAS  Google Scholar 

  • Zeger SL, Liang KY, Albert PS (1988) Models for longitudinal data: a generalized estimating equation approach. Biometrics 44:1049–1060

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

This project was part of the Gene, Environment Association Studies (GENEVA) Consortium funded by the National Human Genome Research Institute (NHGRI) to enhance communication and collaboration among researchers conducting genome-wide studies of complex diseases. Our group benefited greatly from the work and efforts of the entire consortium, especially the Coordinating Center (directed by B. Weir and C. Laurie of the University of Washington) in data cleaning and preparation for submission to the Database for Genotypes and Phenotypes (dbGaP). Special thanks also to David Levine for additional, technical support. We also acknowledge the leadership of T. Manolio of NHGRI. We would also like to thank Helen Voelker and Kathy Farnell of the LHS Data Coordinating Center, University of Minnesota for assistance with the LHS database. We would also like to thank Corinne Boehm and Jane Romm of the Center for Inherited Disease Research, Johns Hopkins University, for technical support. The principal investigators and senior staff of the clinical and coordinating centers, the NHLBI, and members of the Safety and Data Monitoring Board of the Lung Health Study are as follows: Case Western Reserve University, Cleveland, OH: M.D. Altose, M.D. (Principal Investigator), C.D. Deitz, Ph.D. (Project Coordinator); Henry Ford Hospital, Detroit, MI: M.S. Eichenhorn, M.D. (Principal Investigator), K.J. Braden, A.A.S. (Project Coordinator), R.L. Jentons, M.A.L.L.P. (Project Coordinator); Johns Hopkins University School of Medicine, Baltimore, MD: R.A. Wise, M.D. (Principal Investigator), C.S. Rand, Ph.D. (Co-Principal Investigator), K.A. Schiller (Project Coordinator); Mayo Clinic, Rochester, MN: P.D. Scanlon, M.D. (Principal Investigator), G.M. Caron (Project Coordinator), K.S. Mieras, L.C. Walters; Oregon Health Sciences University, Portland: A.S. Buist, M.D. (Principal Investigator), L.R. Johnson, Ph.D. (LHS Pulmonary Function Coordinator), V.J. Bortz (Project Coordinator); University of Alabama at Birmingham: W.C. Bailey, M.D. (Principal Investigator), L.B. Gerald, Ph.D., M.S.P.H. (Project Coordinator); University of California, Los Angeles: D.P. Tashkin, M.D. (Principal Investigator), I.P. Zuniga (Project Coordinator); University of Manitoba, Winnipeg: N.R. Anthonisen, M.D. (Principal Investigator, Steering Committee Chair), J. Manfreda, M.D. (Co-Principal Investigator), R.P. Murray, Ph.D. (Co-Principal Investigator), S.C. Rempel-Rossum (Project Coordinator); University of Minnesota Coordinating Center, Minneapolis: J.E. Connett, Ph.D. (Principal Investigator), P.L. Enright, M.D., P.G. Lindgren, M.S., P. O’Hara, Ph.D., (LHS Intervention Coordinator), M.A. Skeans, M.S., H.T. Voelker; University of Pittsburgh, Pittsburgh, PA: R.M. Rogers, M.D. (Principal Investigator), M.E. Pusateri (Project Coordinator); University of Utah, Salt Lake City: R.E. Kanner, M.D. (Principal Investigator), G.M. Villegas (Project Coordinator); Safety and Data Monitoring Board: M. Becklake, M.D., B. Burrows, M.D. (deceased), P. Cleary, Ph.D., P. Kimbel, M.D. (Chairperson; deceased), L. Nett, R.N., R.R.T. (former member), J.K. Ockene, Ph.D., R.M. Senior, M.D. (Chairperson), G.L. Snider, M.D., W. Spitzer, M.D. (former member), O.D. Williams, Ph.D.; Morbidity and Mortality Review Board: T.E. Cuddy, M.D., R.S. Fontana, M.D., R.E. Hyatt, M.D., C.T. Lambrew, M.D., B.A. Mason, M.D., D.M. Mintzer, M.D., R.B. Wray, M.D.; National Heart, Lung, and Blood Institute staff, Bethesda, MD: S.S. Hurd, Ph.D. (Former Director, Division of Lung Diseases), J.P. Kiley, Ph.D. (Former Project Officer and Director, Division of Lung Diseases), G. Weinmann, M.D. (Former Project Officer and Director, Airway Biology and Disease Program, DLD), M.C. Wu, Ph.D. (Division of Epidemiology and Clinical Applications). Principal investigators and centers participating in ECLIPSE include: Bulgaria: Y. Ivanov, Pleven; K. Kostov, Sofia. Canada: J. Bourbeau, Montreal; M. Fitzgerald, Vancouver; P. Hernández, Halifax; K. Killian, Hamilton; R. Levy, Vancouver; F. Maltais, Montreal; .D O’Donnell, Kingston. Czech Republic: J. Krepelka, Praha. Denmark: J. Vestbo, Hvidovre. The Netherlands: E. Wouters, Horn. New Zealand: D. Quinn, Wellington. Norway: P. Bakke, Bergen, Slovenia: M. Kosnik, Golnik. Spain: A. Agusti, Jaume Sauleda, Palma de Mallorca. Ukraine: Y. Feschenko, Kiev; V. Gavrisyuk, Kiev; L. Yashina, Kiev. UK: L. Yashina, W. MacNee, Edinburgh; D. Singh, Manchester; J. Wedzicha, London. USA: A. Anzueto, San Antonio, TX; S. Braman, Providence. RI; R. Casaburi, Torrance CA; B. Celli, Boston, MA; G. Giessel, Richmond, VA; M. Gotfried, Phoenix, AZ; G. Greenwald, Rancho Mirage, CA; N. Hanania, Houston, TX; D, Mahler, Lebanon, NH; B. Make, Denver, CO; S. Rennard, Omaha, NE; C. Rochester, New Haven, CT; P. Scanlon, Rochester, MN; D. Schuller, Omaha, NE; F. Sciurba, Pittsburg, PA; A. Sharafkhaneh, Houston, TX; T. Siler, St Charles, MO; E. Silverman, Boston, MA; A. Wanner, Miami, FL; R. Wise, Baltimore, MD; R. ZuWallack, Hartford, CT. Steering Committee: H. Coxson (Canada), C. Crim (GlaxoSmithKline, USA), L. Edwards (GlaxoSmithKline, USA), D. Lomas (UK), W. MacNee (UK), E. Silverman (USA), R. Tal-Singer (Co-chair, GlaxoSmithKline, USA), J. Vestbo (Co-chair, Denmark), J. Yates (GlaxoSmithKline, USA). Scientific Committee: A. Agusti (Spain), P. Calverley (UK), B. Celli (USA), C. Crim (GlaxoSmithKline, USA), B. Miller (GlaxoSmithKline, US), W. MacNee (Chair, UK), S. Rennard (USA), R. Tal-Singer (GlaxoSmithKline, USA), E. Wouters (The Netherlands), J. Yates (GlaxoSmithKline, USA). Data deposition: Data can be obtained from dbGaP at http://www.ncbi.nlm.nih.gov/projects/gap/cgi-bin/study.cgi?study_id=phs000335.v1.p1 through dbGaP accession number phs000335.v1.pl. This research was supported by GENEVA (U01HG004738). The Lung Health Study I was supported by contract NIH/N01-HR-46002. Lung tissue validation studies were supported by the NHLBI HL095406-01. KCB was supported in part by the Mary Beryl Patch Turnbull Scholar Program. This CHS research was supported by NHLBI contracts HHSN268201200036C, N01-HC-85239, N01-HC-85079 through N01-HC-85086; N01-HC-35129, N01 HC-15103, N01 HC-55222, N01-HC-75150, N01-HC-45133 and NHLBI grants HL080295, HL087652, HL105756 with additional contribution from NINDS. Additional support was provided through AG-023629, AG-15928, AG-20098, and AG-027058 from the NIA. See also http://www.chs-nhlbi.org/pi.htm. DNA handling and genotyping was supported in part by National Center of Advancing Translational Technologies CTSI grant UL 1TR000124 and National Institute of Diabetes and Digestive and Kidney Diseases grant DK063491 to the Southern California Diabetes Endocrinology Research Center. FHS was funded by N01 HC 25195 from NHLBI. ECLIPSE was supported by GlaxoSmithKline. The BLSA was supported in part by the Intramural Research Program of the NIH, National Institute on Aging. A portion of that support was through a R&D contract with MedStar Research Institute. COPACETIC (acronym of COPD Pathology: Addressing Critical gaps, Early Treatment & diagnosis and Innovative Concepts) is funded by the European Union FP7 program, grant agreement number: 201379. GWAS genotyping was performed at the Center for Inhreited Disease Research under the support of NIH GEI grant U01 HG004438.

Ethical standards

All experiments described here comply with the current laws of the country in which they were performed.

Conflict of interest

The authors declare that they have no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Kathleen C. Barnes.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 89 kb)

Supplementary material 2 (PDF 612 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hansel, N.N., Ruczinski, I., Rafaels, N. et al. Genome-wide study identifies two loci associated with lung function decline in mild to moderate COPD. Hum Genet 132, 79–90 (2013). https://doi.org/10.1007/s00439-012-1219-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00439-012-1219-6

Keywords

Navigation