Skip to main content
Top
Published in: BMC Medicine 1/2012

Open Access 01-12-2012 | Research article

Perinatal nicotine exposure induces asthma in second generation offspring

Authors: Virender K Rehan, Jie Liu, Erum Naeem, Jia Tian, Reiko Sakurai, Kenny Kwong, Omid Akbari, John S Torday

Published in: BMC Medicine | Issue 1/2012

Login to get access

Abstract

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.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference 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. 10.1016/S0140-6736(97)07302-9. 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. 10.1016/S0140-6736(97)07302-9.
3.
go back to reference 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. 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.
5.
go back to reference 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.CrossRefPubMed 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.CrossRefPubMed
6.
go back to reference Adams PF, Hendershot GE, Marano MA: Current estimates from the National Health Interview Survey, 1996. Vital Health Stat 10. 1999, 1-203. Adams PF, Hendershot GE, Marano MA: Current estimates from the National Health Interview Survey, 1996. Vital Health Stat 10. 1999, 1-203.
7.
go back to reference 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. 10.1111/j.1398-9995.2004.00526.x.CrossRefPubMed 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. 10.1111/j.1398-9995.2004.00526.x.CrossRefPubMed
8.
go back to reference 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. 10.1136/thorax.55.4.271.CrossRefPubMedPubMedCentral 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. 10.1136/thorax.55.4.271.CrossRefPubMedPubMedCentral
9.
go back to reference 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. 10.1093/aje/kwm134.CrossRefPubMed 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. 10.1093/aje/kwm134.CrossRefPubMed
10.
go back to reference 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. 10.1164/ajrccm/145.5.1129.CrossRefPubMed 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. 10.1164/ajrccm/145.5.1129.CrossRefPubMed
11.
go back to reference 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. 10.1136/adc.88.12.1086.CrossRefPubMedPubMedCentral 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. 10.1136/adc.88.12.1086.CrossRefPubMedPubMedCentral
12.
go back to reference 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. 10.1164/rccm.200510-1552OC.CrossRefPubMed 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. 10.1164/rccm.200510-1552OC.CrossRefPubMed
16.
go back to reference 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.PubMed 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.PubMed
17.
go back to reference Li YF, Langholz B, Salam MT, Gilliland FD: Maternal and grandmaternal smoking patterns are associated with early childhood asthma. Chest. 2005, 127: 1232-1241. 10.1378/chest.127.4.1232.PubMed Li YF, Langholz B, Salam MT, Gilliland FD: Maternal and grandmaternal smoking patterns are associated with early childhood asthma. Chest. 2005, 127: 1232-1241. 10.1378/chest.127.4.1232.PubMed
18.
go back to reference 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. 10.1007/s00408-009-9158-2.CrossRefPubMedPubMedCentral 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. 10.1007/s00408-009-9158-2.CrossRefPubMedPubMedCentral
19.
go back to reference 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. 10.3109/01902141003714023.CrossRefPubMedPubMedCentral 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. 10.3109/01902141003714023.CrossRefPubMedPubMedCentral
20.
go back to reference 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. 10.1152/ajplung.00337.2010.CrossRefPubMedPubMedCentral 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. 10.1152/ajplung.00337.2010.CrossRefPubMedPubMedCentral
21.
go back to reference 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. 10.1016/j.nut.2003.09.011.CrossRefPubMed 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. 10.1016/j.nut.2003.09.011.CrossRefPubMed
22.
go back to reference Jaenisch R, Bird A: Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 2003, 33 (Suppl): 245-54.CrossRefPubMed Jaenisch R, Bird A: Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 2003, 33 (Suppl): 245-54.CrossRefPubMed
23.
go back to reference 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. 10.1096/fj.07-8229com.CrossRefPubMed 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. 10.1096/fj.07-8229com.CrossRefPubMed
24.
go back to reference 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. 10.1073/pnas.0607090103.CrossRefPubMedPubMedCentral 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. 10.1073/pnas.0607090103.CrossRefPubMedPubMedCentral
25.
go back to reference 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. 10.1136/tc.2005.015065.CrossRefPubMedPubMedCentral 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. 10.1136/tc.2005.015065.CrossRefPubMedPubMedCentral
26.
go back to reference 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. 10.1097/01.ede.0000198148.02347.33.CrossRefPubMed 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. 10.1097/01.ede.0000198148.02347.33.CrossRefPubMed
27.
go back to reference Gabory A, Attig L, Junien C: Sexual dimorphism in environmental epigenetic programming. Mol Cell Endocrinol. 2009, 304: 8-18. 10.1016/j.mce.2009.02.015.CrossRefPubMed Gabory A, Attig L, Junien C: Sexual dimorphism in environmental epigenetic programming. Mol Cell Endocrinol. 2009, 304: 8-18. 10.1016/j.mce.2009.02.015.CrossRefPubMed
28.
go back to reference Zaren B, Lindmark G, Bakketeig L: Maternal smoking affects fetal growth more in the male fetus. Paediatr Perinat Epidemiol. 2000, 14: 118-126. 10.1046/j.1365-3016.2000.00247.x.CrossRefPubMed Zaren B, Lindmark G, Bakketeig L: Maternal smoking affects fetal growth more in the male fetus. Paediatr Perinat Epidemiol. 2000, 14: 118-126. 10.1046/j.1365-3016.2000.00247.x.CrossRefPubMed
29.
go back to reference 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. 10.1007/s00213-006-0441-0.CrossRef 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. 10.1007/s00213-006-0441-0.CrossRef
30.
go back to reference 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. 10.1002/ppul.20407.CrossRefPubMed 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. 10.1002/ppul.20407.CrossRefPubMed
31.
go back to reference 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. 10.3109/01902148.2010.490288.CrossRefPubMed 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. 10.3109/01902148.2010.490288.CrossRefPubMed
32.
go back to reference 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. 10.1152/ajplung.00358.2004.CrossRefPubMed 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. 10.1152/ajplung.00358.2004.CrossRefPubMed
33.
go back to reference Youngson NA, Whitelaw E: Transgenerational epigenetic effects. Annu Rev Genomics Hum Genet. 2008, 9: 233-257. 10.1146/annurev.genom.9.081307.164445.CrossRefPubMed Youngson NA, Whitelaw E: Transgenerational epigenetic effects. Annu Rev Genomics Hum Genet. 2008, 9: 233-257. 10.1146/annurev.genom.9.081307.164445.CrossRefPubMed
34.
go back to reference Kabesch M: Gene by environment interactions and the development of asthma and allergy. Toxicol Lett. 2006, 162: 43-48. 10.1016/j.toxlet.2005.10.009.CrossRefPubMed Kabesch M: Gene by environment interactions and the development of asthma and allergy. Toxicol Lett. 2006, 162: 43-48. 10.1016/j.toxlet.2005.10.009.CrossRefPubMed
35.
go back to reference 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. 10.1111/j.1365-3016.1992.tb00764.x.CrossRefPubMed 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. 10.1111/j.1365-3016.1992.tb00764.x.CrossRefPubMed
36.
go back to reference Ho DH, Burggren WW: Epigenetics and transgenerational transfer: a physiological perspective. J Exp Biol. 2010, 213: 3-16. 10.1242/jeb.019752.CrossRefPubMed Ho DH, Burggren WW: Epigenetics and transgenerational transfer: a physiological perspective. J Exp Biol. 2010, 213: 3-16. 10.1242/jeb.019752.CrossRefPubMed
37.
go back to reference Bygren LO, Kaati G, Edvinsson S: Longevity determined by paternal ancestors' nutrition during their slow growth period. Acta Biotheor. 2001, 49: 53-59. 10.1023/A:1010241825519.CrossRefPubMed Bygren LO, Kaati G, Edvinsson S: Longevity determined by paternal ancestors' nutrition during their slow growth period. Acta Biotheor. 2001, 49: 53-59. 10.1023/A:1010241825519.CrossRefPubMed
38.
go back to reference 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. 10.1038/sj.ejhg.5200859.CrossRefPubMed 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. 10.1038/sj.ejhg.5200859.CrossRefPubMed
39.
go back to reference 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. 10.1038/sj.ejhg.5201538.CrossRefPubMed 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. 10.1038/sj.ejhg.5201538.CrossRefPubMed
40.
go back to reference Skinner MK, Manikkam M, Guerrero-Bosagna C: Epigenetic transgenerational actions of environmental factors in disease etiology. Trends Endocrinol Metab. 2010, 21: 214-222. 10.1016/j.tem.2009.12.007.CrossRefPubMedPubMedCentral Skinner MK, Manikkam M, Guerrero-Bosagna C: Epigenetic transgenerational actions of environmental factors in disease etiology. Trends Endocrinol Metab. 2010, 21: 214-222. 10.1016/j.tem.2009.12.007.CrossRefPubMedPubMedCentral
42.
go back to reference 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. 10.1152/ajplung.00393.2010.CrossRefPubMedPubMedCentral 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. 10.1152/ajplung.00393.2010.CrossRefPubMedPubMedCentral
43.
go back to reference 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. 10.1080/10937400801921320.CrossRefPubMed 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. 10.1080/10937400801921320.CrossRefPubMed
44.
go back to reference Skinner MK: Environmental epigenetic transgenerational inheritance and somatic epigenetic mitotic stability. Epigenetics. 2011, 6: 838-842. 10.4161/epi.6.7.16537.CrossRefPubMed Skinner MK: Environmental epigenetic transgenerational inheritance and somatic epigenetic mitotic stability. Epigenetics. 2011, 6: 838-842. 10.4161/epi.6.7.16537.CrossRefPubMed
45.
go back to reference 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.PubMed 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.PubMed
46.
go back to reference Torday J: Cellular timing of fetal lung development. Semin Perinatol. 1992, 16: 130-139.PubMed Torday J: Cellular timing of fetal lung development. Semin Perinatol. 1992, 16: 130-139.PubMed
47.
go back to reference 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. 10.1210/en.2003-0741.CrossRefPubMed 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. 10.1210/en.2003-0741.CrossRefPubMed
48.
go back to reference 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. 10.1080/15227950307732.CrossRefPubMed 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. 10.1080/15227950307732.CrossRefPubMed
49.
go back to reference 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: 69-10.1186/1465-9921-11-69.CrossRefPubMedPubMedCentral 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: 69-10.1186/1465-9921-11-69.CrossRefPubMedPubMedCentral
50.
go back to reference 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. 10.1158/0008-5472.CAN-07-0478.CrossRefPubMed 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. 10.1158/0008-5472.CAN-07-0478.CrossRefPubMed
51.
go back to reference Luck W, Nau H: Nicotine and cotinine concentrations in serum and milk of nursing smokers. Br J Clin Pharmacol. 1984, 18: 9-15. 10.1111/j.1365-2125.1984.tb05014.x.CrossRefPubMedPubMedCentral Luck W, Nau H: Nicotine and cotinine concentrations in serum and milk of nursing smokers. Br J Clin Pharmacol. 1984, 18: 9-15. 10.1111/j.1365-2125.1984.tb05014.x.CrossRefPubMedPubMedCentral
52.
go back to reference Dempsey D, Jacob P, Benowitz NL: Accelerated metabolism of nicotine and cotinine in pregnant smokers. J Pharmacol Exp Ther. 2002, 301: 594-598. 10.1124/jpet.301.2.594.CrossRefPubMed Dempsey D, Jacob P, Benowitz NL: Accelerated metabolism of nicotine and cotinine in pregnant smokers. J Pharmacol Exp Ther. 2002, 301: 594-598. 10.1124/jpet.301.2.594.CrossRefPubMed
53.
go back to reference 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.PubMed 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.PubMed
54.
go back to reference 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.CrossRefPubMed 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.CrossRefPubMed
55.
go back to reference Maritz GS, Thomas RA: Maternal nicotine exposure: response of type II pneumocytes of neonatal rat pups. Cell Biol Int. 1995, 19: 323-331. 10.1006/cbir.1995.1075.CrossRefPubMed Maritz GS, Thomas RA: Maternal nicotine exposure: response of type II pneumocytes of neonatal rat pups. Cell Biol Int. 1995, 19: 323-331. 10.1006/cbir.1995.1075.CrossRefPubMed
56.
go back to reference 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.PubMed 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.PubMed
57.
go back to reference 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.CrossRefPubMed 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.CrossRefPubMed
58.
go back to reference 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. 10.1172/JCI5232.CrossRefPubMedPubMedCentral 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. 10.1172/JCI5232.CrossRefPubMedPubMedCentral
59.
go back to reference 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.CrossRefPubMed 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.CrossRefPubMed
60.
go back to reference 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. 10.1006/taap.1995.1087.CrossRefPubMed 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. 10.1006/taap.1995.1087.CrossRefPubMed
61.
go back to reference Tsukamoto H, Zhu NL, Asahina K, Mann DA, Mann J: Epigenetic cell fate regulation of hepatic stellate cells. Hepatol Res. 2011, 41: 675-682. 10.1111/j.1872-034X.2011.00804.x.CrossRefPubMedPubMedCentral Tsukamoto H, Zhu NL, Asahina K, Mann DA, Mann J: Epigenetic cell fate regulation of hepatic stellate cells. Hepatol Res. 2011, 41: 675-682. 10.1111/j.1872-034X.2011.00804.x.CrossRefPubMedPubMedCentral
Metadata
Title
Perinatal nicotine exposure induces asthma in second generation offspring
Authors
Virender K Rehan
Jie Liu
Erum Naeem
Jia Tian
Reiko Sakurai
Kenny Kwong
Omid Akbari
John S Torday
Publication date
01-12-2012
Publisher
BioMed Central
Published in
BMC Medicine / Issue 1/2012
Electronic ISSN: 1741-7015
DOI
https://doi.org/10.1186/1741-7015-10-129

Other articles of this Issue 1/2012

BMC Medicine 1/2012 Go to the issue