期刊论文详细信息
BMC Medicine
Perinatal nicotine exposure induces asthma in second generation offspring
John S Torday1  Omid Akbari2  Kenny Kwong1  Reiko Sakurai1  Jia Tian1  Erum Naeem1  Jie Liu1  Virender K Rehan1 
[1]Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center at David Geffen School of Medicine, 1124 West Carson Street, Torrance, 90502, USA
[2]Department of Molecular Microbiology and Immunology, University of Southern California, Keck School of Medicine, 1975 Zonal Avenue, Los Angeles, 90033, USA
关键词: gender difference;    multigenerational;    asthma;    epigenetic;    lung;    nicotine;   
Others  :  857322
DOI  :  10.1186/1741-7015-10-129
 received in 2012-05-09, accepted in 2012-10-30,  发布年份 2012
PDF
【 摘 要 】

Background

By altering specific developmental signaling pathways that are necessary for fetal lung development, perinatal nicotine exposure affects lung growth and differentiation, resulting in the offsprings' predisposition to childhood asthma; peroxisome proliferator-activated receptor gamma (PPARγ) agonists can inhibit this effect. However, whether the perinatal nicotine-induced asthma risk is restricted to nicotine-exposed offspring only; whether it can be transmitted to the next generation; and whether PPARγ agonists would have any effect on this process are not known.

Methods

Time-mated Sprague Dawley rat dams received either placebo or nicotine (1 mg/kg, s.c.), once daily from day 6 of gestation to postnatal day (PND) 21. Following delivery, at PND21, generation 1 (F1) pups were either subjected to pulmonary function tests, or killed to obtain their lungs, tracheas, and gonads to determine the relevant protein markers (mesenchymal contractile proteins), global DNA methylation, histone 3 and 4 acetylation, and for tracheal tension studies. Some F1 animals were used as breeders to generate F2 pups, but without any exposure to nicotine in the F1 pregnancy. At PND21, F2 pups underwent studies similar to those performed on F1 pups.

Results

Consistent with the asthma phenotype, nicotine affected lung function in both male and female F1 and F2 offspring (maximal 250% increase in total respiratory system resistance, and 84% maximal decrease in dynamic compliance following methacholine challenge; P < 0.01, nicotine versus control; P < 0.05, males versus females; and P > 0.05, F1 versus F2), but only affected tracheal constriction in males (51% maximal increase in tracheal constriction following acetylcholine challenge, P < 0.01, nicotine versus control; P < 0.0001, males versus females; P > 0.05, F1 versus F2); nicotine also increased the contractile protein content of whole lung (180% increase in fibronectin protein levels, P < 0.01, nicotine versus control, and P < 0.05, males versus females) and isolated lung fibroblasts (for example, 45% increase in fibronectin protein levels, P < 0.05, nicotine versus control), along with decreased PPARγ expression (30% decrease, P < 0.05, nicotine versus control), but only affected contractile proteins in the male trachea (P < 0.05, nicotine versus control, and P < 0.0001, males versus females). All of the nicotine-induced changes in the lung and gonad DNA methylation and histone 3 and 4 acetylation were normalized by the PPARγ agonist rosiglitazone except for the histone 4 acetylation in the lung.

Conclusions

Germline epigenetic marks imposed by exposure to nicotine during pregnancy can become permanently programmed and transferred through the germline to subsequent generations, a ground-breaking finding that shifts the current asthma paradigm, opening up many new avenues to explore.

【 授权许可】

   
2012 Rehan et al; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20140723074946961.pdf 643KB PDF download
45KB Image download
38KB Image download
28KB Image download
45KB Image download
43KB Image download
44KB Image download
42KB Image download
66KB Image download
27KB Image download
37KB Image download
【 图 表 】

【 参考文献 】
  • [1]Braman SS: The global burden of asthma. Chest 2006, 130:4S-12S.
  • [2]Worldwide variation in prevalence of symptoms of asthma, allergic rhinoconjunctivitis, and atopic eczema: ISAAC. The International Study of Asthma and Allergies in Childhood (ISAAC) Steering Committee Lancet 1998, 351:1225-1232.
  • [3]Mannino DM, Homa DM, Akinbami LJ, Moorman JE, Gwynn C, Redd SC: Surveillance for asthma--United States, 1980-1999. MMWR Surveill Summ 2002, 51:1-13.
  • [4]Akinbami LJ, Moorman JE, Liu X: Asthma prevalence, health care use, and mortality: United States, 2005-2009. [http://www.cdc.gov/nchs/data/nhsr/nhsr032.pdf] webcite 2011, 1-12.
  • [5]Smith DH, Malone DC, Lawson KA, Okamoto LJ, Battista C, Saunders WB: A national estimate of the economic costs of asthma. Am J Respir Crit Care Med 1997, 156:787-793.
  • [6]Adams PF, Hendershot GE, Marano MA: Current estimates from the National Health Interview Survey, 1996. Vital Health Stat 10 1999, 1-203.
  • [7]Masoli M, Fabian D, Holt S, Beasley R: The global burden of asthma: executive summary of the GINA Dissemination Committee report. Allergy 2004, 59:469-478.
  • [8]Gilliland FD, Berhane K, McConnell R, Gauderman WJ, Vora H, Rappaport EB, et al.: Maternal smoking during pregnancy, environmental tobacco smoke exposure and childhood lung function. Thorax 2000, 55:271-276.
  • [9]Haberg SE, Stigum H, Nystad W, Nafstad P: Effects of pre- and postnatal exposure to parental smoking on early childhood respiratory health. Am J Epidemiol 2007, 166:679-686.
  • [10]Hanrahan JP, Tager IB, Segal MR, Tosteson TD, Castile RG, Van VH, et al.: The effect of maternal smoking during pregnancy on early infant lung function. Am Rev Respir Dis 1992, 145:1129-1135.
  • [11]Hofhuis W, de Jongste JC, Merkus PJ: Adverse health effects of prenatal and postnatal tobacco smoke exposure on children. Arch Dis Child 2003, 88:1086-1090.
  • [12]Moshammer H, Hoek G, Luttmann-Gibson H, Neuberger MA, Antova T, Gehring U, et al.: Parental smoking and lung function in children: an international study. Am J Respir Crit Care Med 2006, 173:1255-1263.
  • [13]March of Dimes. Alcohol and drugs [http://www.marchofdimes.com/pregnancy/alcohol_smoking.html] webcite 2010.
  • [14]Federal Trade Commission. Federal Trade Commission Cigarette Report For 2002 [http://ftc.gov/reports/cigarette/041022cigaretterpt.pdf] webcite 2004.
  • [15]Centers For Disease Control and Prevention. Smoking & Tobacco Use [http://www.cdc.gov/tobacco/] webcite 5-8-2012
  • [16]Martin JA, Hamilton BE, Sutton PD, Ventura SJ, Menacker F, Munson ML: Births: final data for 2002. Natl Vital Stat Rep 2003, 52:1-113.
  • [17]Li YF, Langholz B, Salam MT, Gilliland FD: Maternal and grandmaternal smoking patterns are associated with early childhood asthma. Chest 2005, 127:1232-1241.
  • [18]Rehan VK, Asotra K, Torday JS: The effects of smoking on the developing lung: insights from a biologic model for lung development, homeostasis, and repair. Lung 2009, 187:281-289.
  • [19]Krebs M, Sakurai R, Torday JS, Rehan VK: Evidence for in vivo nicotine-induced alveolar interstitial fibroblast-to-myofibroblast transdifferentiation. Exp Lung Res 2010, 36:390-398.
  • [20]Liu J, Sakurai R, O'Roark EM, Kenyon NJ, Torday JS, Rehan VK: PPARgamma agonist rosiglitazone prevents perinatal nicotine exposure-induced asthma in rat offspring. Am J Physiol Lung Cell Mol Physiol 2011, 300:L710-L717.
  • [21]Waterland RA, Jirtle RL: Early nutrition, epigenetic changes at transposons and imprinted genes, and enhanced susceptibility to adult chronic diseases. Nutrition 2004, 20:63-8.
  • [22]Jaenisch R, Bird A: Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet 2003, 33(Suppl):245-54.
  • [23]Waterland RA, Travisano M, Tahiliani KG: Diet-induced hypermethylation at agouti viable yellow is not inherited transgenerationally through the female. FASEB J 2007, 21:3380-3385.
  • [24]Cropley JE, Suter CM, Beckman KB, Martin DI: Germ-line epigenetic modification of the murine A vy allele by nutritional supplementation. Proc Natl Acad Sci USA 2006, 103:17308-17312.
  • [25]Pattenden S, Antova T, Neuberger M, Nikiforov B, De SM, Grize L, et al.: Parental smoking and children's respiratory health: independent effects of prenatal and postnatal exposure. Tob Control 2006, 15:294-301.
  • [26]Alati R, Al MA, O'Callaghan M, Najman JM, Williams GM: In utero and postnatal maternal smoking and asthma in adolescence. Epidemiology 2006, 17:138-144.
  • [27]Gabory A, Attig L, Junien C: Sexual dimorphism in environmental epigenetic programming. Mol Cell Endocrinol 2009, 304:8-18.
  • [28]Zaren B, Lindmark G, Bakketeig L: Maternal smoking affects fetal growth more in the male fetus. Paediatr Perinat Epidemiol 2000, 14:118-126.
  • [29]Matta SG, Balfour DJ, Benowitz NL, Boyd RT, Buccafusco JJ, Caggiula AR, et al.: Guidelines on nicotine dose selection for in vivo research. Psychopharmacology (Berl) 2007, 190:269-319.
  • [30]Rehan VK, Wang Y, Patel S, Santos J, Torday JS: Rosiglitazone, a peroxisome proliferators-activated receptor γ agonist, prevents hyperoxia-induced neonatal rat lung injury in vivo. Pediatr Pulmonol 2006, 41:558-569.
  • [31]Sakurai R, Cerny LM, Torday JS, Rehan VK: Mechanism for nicotine-induced up-regulation of Wnt signaling in human alveolar interstitial fibroblasts. Exp Lung Res 2011, 37:144-154.
  • [32]Rehan VK, Wang Y, Sugano S, Romero S, Chen X, Santos J, et al.: Mechanism of nicotine-induced pulmonary fibroblast transdifferentiation. Am J Physiol Lung Cell Mol Physiol 2005, 289:L667-L676.
  • [33]Youngson NA, Whitelaw E: Transgenerational epigenetic effects. Annu Rev Genomics Hum Genet 2008, 9:233-257.
  • [34]Kabesch M: Gene by environment interactions and the development of asthma and allergy. Toxicol Lett 2006, 162:43-48.
  • [35]Lumey LH: Decreased birthweights in infants after maternal in utero exposure to the Dutch famine of 1944-1945. Paediatr Perinat Epidemiol 1992, 6:240-253.
  • [36]Ho DH, Burggren WW: Epigenetics and transgenerational transfer: a physiological perspective. J Exp Biol 2010, 213:3-16.
  • [37]Bygren LO, Kaati G, Edvinsson S: Longevity determined by paternal ancestors' nutrition during their slow growth period. Acta Biotheor 2001, 49:53-59.
  • [38]Kaati G, Bygren LO, Edvinsson S: Cardiovascular and diabetes mortality determined by nutrition during parents' and grandparents' slow growth period. Eur J Hum Genet 2002, 10:682-688.
  • [39]Pembrey ME, Bygren LO, Kaati G, Edvinsson S, Northstone K, Sjostrom M, et al.: Sex-specific, male-line transgenerational responses in humans. Eur J Hum Genet 2006, 14:159-166.
  • [40]Skinner MK, Manikkam M, Guerrero-Bosagna C: Epigenetic transgenerational actions of environmental factors in disease etiology. Trends Endocrinol Metab 2010, 21:214-222.
  • [41]Christensen BC, Marsit CJ: Epigenomics in environmental health. Front Genet 2011, 2:84.
  • [42]Rehan VK, Sakurai R, Torday JS: Thirdhand smoke: a new dimension to the effects of cigarette smoke on the developing lung. Am J Physiol Lung Cell Mol Physiol 2011, 301:L1-L8.
  • [43]Wigle DT, Arbuckle TE, Turner MC, Berube A, Yang Q, Liu S, et al.: Epidemiologic evidence of relationships between reproductive and child health outcomes and environmental chemical contaminants. J Toxicol Environ Health B Crit Rev 2008, 11:373-517.
  • [44]Skinner MK: Environmental epigenetic transgenerational inheritance and somatic epigenetic mitotic stability. Epigenetics 2011, 6:838-842.
  • [45]Mead J: Dysanapsis in normal lungs assessed by the relationship between maximal flow, static recoil, and vital capacity. Am Rev Respir Dis 1980, 121:339-342.
  • [46]Torday J: Cellular timing of fetal lung development. Semin Perinatol 1992, 16:130-139.
  • [47]Singh R, Artaza JN, Taylor WE, Gonzalez-Cadavid NF, Bhasin S: Androgens stimulate myogenic differentiation and inhibit adipogenesis in C3H 10T1/2 pluripotent cells through an androgen receptor-mediated pathway. Endocrinology 2003, 144:5081-5088.
  • [48]Torday JS, Torres E, Rehan VK: The role of fibroblast transdifferentiation in lung epithelial cell proliferation, differentiation, and repair in vitro. Pediatr Pathol Mol Med 2003, 22:189-207.
  • [49]Simon DM, Tsai LW, Ingenito EP, Starcher BC, Mariani TJ: PPARgamma deficiency results in reduced lung elastic recoil and abnormalities in airspace distribution. Respir Res 2010, 11:69. BioMed Central Full Text
  • [50]Liu XH, Kirschenbaum A, Yao S, Liu G, Aaronson SA, Levine AC: Androgen-induced Wnt signaling in preosteoblasts promotes the growth of MDA-PCa-2b human prostate cancer cells. Cancer Res 2007, 67:5747-5753.
  • [51]Luck W, Nau H: Nicotine and cotinine concentrations in serum and milk of nursing smokers. Br J Clin Pharmacol 1984, 18:9-15.
  • [52]Dempsey D, Jacob P III, Benowitz NL: Accelerated metabolism of nicotine and cotinine in pregnant smokers. J Pharmacol Exp Ther 2002, 301:594-598.
  • [53]Luck W, Nau H, Hansen R, Steldinger R: Extent of nicotine and cotinine transfer to the human fetus, placenta and amniotic fluid of smoking mothers. Dev Pharmacol Ther 1985, 8:384-395.
  • [54]Szuts T, Olsson S, Lindquist NG, Ullberg S, Pilotti A, Enzell C: Long-term fate of [14C]nicotine in the mouse: retention in the bronchi, melanin-containing tissues and urinary bladder wall. Toxicology 1978, 10:207-220.
  • [55]Maritz GS, Thomas RA: Maternal nicotine exposure: response of type II pneumocytes of neonatal rat pups. Cell Biol Int 1995, 19:323-331.
  • [56]Roman J, Ritzenthaler JD, Gil-Acosta A, Rivera HN, Roser-Page S: Nicotine and fibronectin expression in lung fibroblasts: implications for tobacco-related lung tissue remodeling. FASEB J 2004, 18:1436-1438.
  • [57]Rehan VK, Wang Y, Sugano S, Santos J, Patel S, Sakurai R, et al.: In utero nicotine exposure alters fetal rat lung alveolar type II cell proliferation, differentiation, and metabolism. Am J Physiol Lung Cell Mol Physiol 2007, 292:L323-L333.
  • [58]Sekhon HS, Jia Y, Raab R, Kuryatov A, Pankow JF, Whitsett JA, et al.: Prenatal nicotine increases pulmonary alpha7 nicotinic receptor expression and alters fetal lung development in monkeys. J Clin Invest 1999, 103:637-647.
  • [59]Sekhon HS, Keller JA, Proskocil BJ, Martin EL, Spindel ER: Maternal nicotine exposure upregulates collagen gene expression in fetal monkey lung. Association with alpha7 nicotinic acetylcholine receptors. Am J Respir Cell Mol Biol 2002, 26:31-41.
  • [60]Joad JP, Ji C, Kott KS, Bric JM, Pinkerton KE: In utero and postnatal effects of sidestream cigarette smoke exposure on lung function, hyperresponsiveness, and neuroendocrine cells in rats. Toxicol Appl Pharmacol 1995, 132:63-71.
  • [61]Tsukamoto H, Zhu NL, Asahina K, Mann DA, Mann J: Epigenetic cell fate regulation of hepatic stellate cells. Hepatol Res 2011, 41:675-682.
  文献评价指标  
  下载次数:9次 浏览次数:8次