Skip to main content
Top
Published in: Fluids and Barriers of the CNS 1/2023

Open Access 01-12-2023 | Research

Maternal e-cigarette use can disrupt postnatal blood-brain barrier (BBB) integrity and deteriorates motor, learning and memory function: influence of sex and age

Authors: Sabrina Rahman Archie, Ali Ehsan Sifat, Yong Zhang, Heidi Villalba, Sejal Sharma, Saeideh Nozohouri, Thomas J. Abbruscato

Published in: Fluids and Barriers of the CNS | Issue 1/2023

Login to get access

Abstract

Electronic nicotine delivery systems (ENDS), also commonly known as electronic cigarettes (e-cigs) are considered in most cases as a safer alternative to tobacco smoking and therefore have become extremely popular among all age groups and sex. It is estimated that up to 15% of pregnant women are now using e-cigs in the US which keeps increasing at an alarming rate. Harmful effects of tobacco smoking during pregnancy are well documented for both pregnancy and postnatal health, however limited preclinical and clinical studies exist to evaluate the long-term effects of prenatal e-cig exposure on postnatal health. Therefore, the aim of our study is to evaluate the effect of maternal e-cig use on postnatal blood-brain barrier (BBB) integrity and behavioral outcomes of mice of varying age and sex. In this study, pregnant CD1 mice (E5) were exposed to e‐Cig vapor (2.4% nicotine) until postnatal day (PD) 7. Weight of the offspring was measured at PD0, PD7, PD15, PD30, PD45, PD60 and PD90. The expression of structural elements of the BBB, tight junction proteins (ZO-1, claudin-5, occludin), astrocytes (GFAP), pericytes (PDGFRβ) and the basement membrane (laminin α1, laminin α4), neuron specific marker (NeuN), water channel protein (AQP4) and glucose transporter (GLUT1) were analyzed in both male and female offspring using western blot and immunofluorescence. Estrous cycle was recorded by vaginal cytology method. Long‐term motor and cognitive functions were evaluated using open field test (OFT), novel object recognition test (NORT) and morris water maze test (MWMT) at adolescence (PD 40–45) and adult (PD 90–95) age. In our study, significantly reduced expression of tight junction proteins and astrocyte marker were observed in male and female offspring until PD 90 (P < 0.05). Additionally, prenatally e-cig exposed adolescent and adult offspring showed impaired locomotor, learning, and memory function compared to control offspring (P < 0.05). Our findings suggest that prenatal e-cig exposure induces long-term neurovascular changes of neonates by disrupting postnatal BBB integrity and worsening behavioral outcomes.
Appendix
Available only for authorised users
Literature
1.
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(12):1086–90.PubMedPubMedCentralCrossRef Hofhuis W, de Jongste JC, Merkus PJ. Adverse health effects of prenatal and postnatal tobacco smoke exposure on children. Arch Dis Child. 2003;88(12):1086–90.PubMedPubMedCentralCrossRef
2.
go back to reference Rauschert S, et al. Maternal smoking during pregnancy induces persistent epigenetic changes into adolescence, independent of postnatal smoke exposure and is associated with cardiometabolic risk. Front Genet. 2019;10:770.PubMedPubMedCentralCrossRef Rauschert S, et al. Maternal smoking during pregnancy induces persistent epigenetic changes into adolescence, independent of postnatal smoke exposure and is associated with cardiometabolic risk. Front Genet. 2019;10:770.PubMedPubMedCentralCrossRef
3.
go back to reference Lange S, et al. National, regional, and global prevalence of smoking during pregnancy in the general population: a systematic review and meta-analysis. Lancet Glob Health. 2018;6(7):e769–76.PubMedCrossRef Lange S, et al. National, regional, and global prevalence of smoking during pregnancy in the general population: a systematic review and meta-analysis. Lancet Glob Health. 2018;6(7):e769–76.PubMedCrossRef
4.
go back to reference U.S. Department of Health and Human Services, C.f.D.C.a.P., National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health. The health consequences of smoking: 50 years of progress. A Report of the Surgeon General. 2014. U.S. Department of Health and Human Services, C.f.D.C.a.P., National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health. The health consequences of smoking: 50 years of progress. A Report of the Surgeon General. 2014.
5.
go back to reference Gould GS, et al. Exposure to tobacco, environmental tobacco smoke and nicotine in pregnancy: a pragmatic overview of reviews of maternal and child outcomes, effectiveness of interventions and barriers and facilitators to quitting. Int J Environ Res Public Health. 2020;17:6.CrossRef Gould GS, et al. Exposure to tobacco, environmental tobacco smoke and nicotine in pregnancy: a pragmatic overview of reviews of maternal and child outcomes, effectiveness of interventions and barriers and facilitators to quitting. Int J Environ Res Public Health. 2020;17:6.CrossRef
6.
go back to reference Sifat AE, et al. Prenatal electronic cigarette exposure decreases brain glucose utilization and worsens outcome in offspring hypoxic–ischemic brain injury. J Neurochem. 2020;153(1):63–79.PubMedCrossRef Sifat AE, et al. Prenatal electronic cigarette exposure decreases brain glucose utilization and worsens outcome in offspring hypoxic–ischemic brain injury. J Neurochem. 2020;153(1):63–79.PubMedCrossRef
7.
go back to reference Alkam T, et al. Evaluation of cognitive behaviors in young offspring of C57BL/6J mice after gestational nicotine exposure during different time-windows. Psychopharmacology. 2013;230(3):451–63.PubMedCrossRef Alkam T, et al. Evaluation of cognitive behaviors in young offspring of C57BL/6J mice after gestational nicotine exposure during different time-windows. Psychopharmacology. 2013;230(3):451–63.PubMedCrossRef
8.
go back to reference Li Y, et al. Perinatal nicotine exposure increases vulnerability of hypoxic-ischemic brain injury in neonatal rats: role of angiotensin II receptors. Stroke. 2012;43(9):2483–90.PubMedPubMedCentralCrossRef Li Y, et al. Perinatal nicotine exposure increases vulnerability of hypoxic-ischemic brain injury in neonatal rats: role of angiotensin II receptors. Stroke. 2012;43(9):2483–90.PubMedPubMedCentralCrossRef
9.
go back to reference Lacy RT, et al. Intravenous prenatal nicotine exposure alters METH-induced hyperactivity, conditioned hyperactivity, and BDNF in adult rat offspring. Dev Neurosci. 2016;38(3):171–85.PubMedCrossRef Lacy RT, et al. Intravenous prenatal nicotine exposure alters METH-induced hyperactivity, conditioned hyperactivity, and BDNF in adult rat offspring. Dev Neurosci. 2016;38(3):171–85.PubMedCrossRef
10.
go back to reference Lee H, Chung S, Noh J. Maternal nicotine exposure during late gestation and lactation increases anxiety-like and impulsive decision-making behavior in adolescent offspring of rat. Toxicol Res. 2016;32(4):275–80.PubMedPubMedCentralCrossRef Lee H, Chung S, Noh J. Maternal nicotine exposure during late gestation and lactation increases anxiety-like and impulsive decision-making behavior in adolescent offspring of rat. Toxicol Res. 2016;32(4):275–80.PubMedPubMedCentralCrossRef
11.
go back to reference Newman MB, Shytle RD, Sanberg PR. Locomotor behavioral effects of prenatal and postnatal nicotine exposure in rat offspring. Behav Pharmacol. 1999;10(6–7):699–706.PubMedCrossRef Newman MB, Shytle RD, Sanberg PR. Locomotor behavioral effects of prenatal and postnatal nicotine exposure in rat offspring. Behav Pharmacol. 1999;10(6–7):699–706.PubMedCrossRef
12.
go back to reference Pinheiro CR, et al. Maternal nicotine exposure during lactation alters food preference, anxiety-like behavior and the brain dopaminergic reward system in the adult rat offspring. Physiol Behav. 2015;149:131–41.PubMedCrossRef Pinheiro CR, et al. Maternal nicotine exposure during lactation alters food preference, anxiety-like behavior and the brain dopaminergic reward system in the adult rat offspring. Physiol Behav. 2015;149:131–41.PubMedCrossRef
15.
17.
go back to reference Whittington JR, et al. The use of electronic cigarettes in pregnancy: a review of the literature. Obstet Gynecol Surv. 2018;73(9):544–9.PubMedCrossRef Whittington JR, et al. The use of electronic cigarettes in pregnancy: a review of the literature. Obstet Gynecol Surv. 2018;73(9):544–9.PubMedCrossRef
18.
go back to reference Liu B, et al. Prevalence and distribution of electronic cigarette use before and during pregnancy among women in 38 states of the United States. Nicotine Tob Res. 2021;23(9):1459–67.PubMedPubMedCentralCrossRef Liu B, et al. Prevalence and distribution of electronic cigarette use before and during pregnancy among women in 38 states of the United States. Nicotine Tob Res. 2021;23(9):1459–67.PubMedPubMedCentralCrossRef
19.
go back to reference Obisesan OH, et al. E-cigarette use patterns and high-risk behaviors in pregnancy: behavioral risk factor surveillance system, 2016–2018. Am J Prev Med. 2020;59(2):187–95.PubMedPubMedCentralCrossRef Obisesan OH, et al. E-cigarette use patterns and high-risk behaviors in pregnancy: behavioral risk factor surveillance system, 2016–2018. Am J Prev Med. 2020;59(2):187–95.PubMedPubMedCentralCrossRef
20.
go back to reference Kaisar MA, et al. A decade of e-cigarettes: limited research & unresolved safety concerns. Toxicology. 2016;365:67–75.PubMedCrossRef Kaisar MA, et al. A decade of e-cigarettes: limited research & unresolved safety concerns. Toxicology. 2016;365:67–75.PubMedCrossRef
21.
go back to reference Zhang Y, et al. Maternal electronic cigarette exposure in relation to offspring development: a comprehensive review. Am J Obstet Gynecol MFM. 2022;4(5):100659.PubMedCrossRef Zhang Y, et al. Maternal electronic cigarette exposure in relation to offspring development: a comprehensive review. Am J Obstet Gynecol MFM. 2022;4(5):100659.PubMedCrossRef
22.
go back to reference Chen H, et al. Maternal e-cigarette exposure in mice alters dna methylation and lung cytokine expression in offspring. Am J Respir Cell Mol Biol. 2018;58(3):366–77.PubMedCrossRef Chen H, et al. Maternal e-cigarette exposure in mice alters dna methylation and lung cytokine expression in offspring. Am J Respir Cell Mol Biol. 2018;58(3):366–77.PubMedCrossRef
23.
go back to reference Chen H, et al. Modulation of neural regulators of energy homeostasis, and of inflammation, in the pups of mice exposed to e-cigarettes. Neurosci Lett. 2018;684:61–6.PubMedCrossRef Chen H, et al. Modulation of neural regulators of energy homeostasis, and of inflammation, in the pups of mice exposed to e-cigarettes. Neurosci Lett. 2018;684:61–6.PubMedCrossRef
25.
go back to reference Chen Z, et al. Single-nucleus chromatin accessibility and RNA sequencing reveal impaired brain development in prenatally e-cigarette exposed neonatal rats. iScience. 2022;25(8):104686.PubMedPubMedCentralCrossRef Chen Z, et al. Single-nucleus chromatin accessibility and RNA sequencing reveal impaired brain development in prenatally e-cigarette exposed neonatal rats. iScience. 2022;25(8):104686.PubMedPubMedCentralCrossRef
26.
go back to reference Orzabal MR, et al. Chronic exposure to e-cig aerosols during early development causes vascular dysfunction and offspring growth deficits. Transl Res. 2019;207:70–82.PubMedPubMedCentralCrossRef Orzabal MR, et al. Chronic exposure to e-cig aerosols during early development causes vascular dysfunction and offspring growth deficits. Transl Res. 2019;207:70–82.PubMedPubMedCentralCrossRef
27.
go back to reference Lauterstein DE, et al. Frontal cortex transcriptome analysis of mice exposed to electronic cigarettes during early life stages. Int J Environ Res Public Health. 2016;13:4.CrossRef Lauterstein DE, et al. Frontal cortex transcriptome analysis of mice exposed to electronic cigarettes during early life stages. Int J Environ Res Public Health. 2016;13:4.CrossRef
28.
go back to reference Zelikoff JT, et al. Microglia activation and gene expression alteration of neurotrophins in the hippocampus following early-life exposure to e-cigarette aerosols in a murine model. Toxicol Sci. 2018;162(1):276–86.PubMedCrossRef Zelikoff JT, et al. Microglia activation and gene expression alteration of neurotrophins in the hippocampus following early-life exposure to e-cigarette aerosols in a murine model. Toxicol Sci. 2018;162(1):276–86.PubMedCrossRef
30.
go back to reference Tahajjodi SS, et al. The effect of maternal nicotine on basement membrane collagen IV of brain microvessels changes in neonatal Balb/C mice. Iran J Reprod Med. 2014;12(4):275–80.PubMedPubMedCentral Tahajjodi SS, et al. The effect of maternal nicotine on basement membrane collagen IV of brain microvessels changes in neonatal Balb/C mice. Iran J Reprod Med. 2014;12(4):275–80.PubMedPubMedCentral
31.
go back to reference Archie SR, Al Shoyaib A, Cucullo L. Blood-brain barrier dysfunction in CNS disorders and putative therapeutic targets: an overview. Pharmaceutics. 2021;13(11):1779.PubMedPubMedCentralCrossRef Archie SR, Al Shoyaib A, Cucullo L. Blood-brain barrier dysfunction in CNS disorders and putative therapeutic targets: an overview. Pharmaceutics. 2021;13(11):1779.PubMedPubMedCentralCrossRef
32.
go back to reference Abbruscato TJ, et al. Nicotine and cotinine modulate cerebral microvascular permeability and protein expression of ZO-1 through nicotinic acetylcholine receptors expressed on brain endothelial cells. J Pharm Sci. 2002;91(12):2525–38.PubMedCrossRef Abbruscato TJ, et al. Nicotine and cotinine modulate cerebral microvascular permeability and protein expression of ZO-1 through nicotinic acetylcholine receptors expressed on brain endothelial cells. J Pharm Sci. 2002;91(12):2525–38.PubMedCrossRef
33.
go back to reference Kadry H, et al. Comparative assessment of in vitro BBB tight junction integrity following exposure to cigarette smoke and e-cigarette vapor: a quantitative evaluation of the protective effects of metformin using small-molecular-weight paracellular markers. Fluids and Barriers of the CNS. 2021;18(1):28.PubMedPubMedCentralCrossRef Kadry H, et al. Comparative assessment of in vitro BBB tight junction integrity following exposure to cigarette smoke and e-cigarette vapor: a quantitative evaluation of the protective effects of metformin using small-molecular-weight paracellular markers. Fluids and Barriers of the CNS. 2021;18(1):28.PubMedPubMedCentralCrossRef
34.
go back to reference Kaisar MA, et al. Offsetting the impact of smoking and e-cigarette vaping on the cerebrovascular system and stroke injury: is metformin a viable countermeasure? Redox Biol. 2017;13:353–62.PubMedPubMedCentralCrossRef Kaisar MA, et al. Offsetting the impact of smoking and e-cigarette vaping on the cerebrovascular system and stroke injury: is metformin a viable countermeasure? Redox Biol. 2017;13:353–62.PubMedPubMedCentralCrossRef
35.
go back to reference Abbruscato TJ, et al. Regulation of blood-brain barrier Na, K,2Cl-cotransporter through phosphorylation during in vitro stroke conditions and nicotine exposure. J Pharmacol Exp Ther. 2004;310(2):459–68.PubMedCrossRef Abbruscato TJ, et al. Regulation of blood-brain barrier Na, K,2Cl-cotransporter through phosphorylation during in vitro stroke conditions and nicotine exposure. J Pharmacol Exp Ther. 2004;310(2):459–68.PubMedCrossRef
36.
go back to reference Paulson JR, et al. Tobacco smoke chemicals attenuate brain-to-blood potassium transport mediated by the Na, K,2Cl-cotransporter during hypoxia-reoxygenation. J Pharmacol Exp Ther. 2006;316(1):248–54.PubMedCrossRef Paulson JR, et al. Tobacco smoke chemicals attenuate brain-to-blood potassium transport mediated by the Na, K,2Cl-cotransporter during hypoxia-reoxygenation. J Pharmacol Exp Ther. 2006;316(1):248–54.PubMedCrossRef
37.
go back to reference Sifat AE, et al. Nicotine and electronic cigarette (E-Cig) exposure decreases brain glucose utilization in ischemic stroke. J Neurochem. 2018;147(2):204–21.PubMedPubMedCentralCrossRef Sifat AE, et al. Nicotine and electronic cigarette (E-Cig) exposure decreases brain glucose utilization in ischemic stroke. J Neurochem. 2018;147(2):204–21.PubMedPubMedCentralCrossRef
40.
42.
go back to reference McGrath-Morrow SA, et al. The effects of electronic cigarette emissions on systemic cotinine levels, weight and postnatal lung growth in neonatal mice. PLoS ONE. 2015;10(2):e0118344.PubMedPubMedCentralCrossRef McGrath-Morrow SA, et al. The effects of electronic cigarette emissions on systemic cotinine levels, weight and postnatal lung growth in neonatal mice. PLoS ONE. 2015;10(2):e0118344.PubMedPubMedCentralCrossRef
44.
go back to reference Kaisar MA, et al. A convenient UHPLC-MS/MS method for routine monitoring of plasma and brain levels of nicotine and cotinine as a tool to validate newly developed preclinical smoking model in mouse. BMC Neurosci. 2017;18(1):71.PubMedPubMedCentralCrossRef Kaisar MA, et al. A convenient UHPLC-MS/MS method for routine monitoring of plasma and brain levels of nicotine and cotinine as a tool to validate newly developed preclinical smoking model in mouse. BMC Neurosci. 2017;18(1):71.PubMedPubMedCentralCrossRef
45.
go back to reference Zhang Y, et al. Role of P53-senescence induction in suppression of LNCaP prostate cancer growth by cardiotonic compound bufalin. Mol Cancer Ther. 2018;17(11):2341–52.PubMedPubMedCentralCrossRef Zhang Y, et al. Role of P53-senescence induction in suppression of LNCaP prostate cancer growth by cardiotonic compound bufalin. Mol Cancer Ther. 2018;17(11):2341–52.PubMedPubMedCentralCrossRef
46.
go back to reference Zhang Y, et al. Potential role of astrocyte angiotensin converting enzyme 2 in the neural transmission of COVID-19 and a neuroinflammatory state induced by smoking and vaping. Fluids Barriers CNS. 2022;19(1):46.PubMedPubMedCentralCrossRef Zhang Y, et al. Potential role of astrocyte angiotensin converting enzyme 2 in the neural transmission of COVID-19 and a neuroinflammatory state induced by smoking and vaping. Fluids Barriers CNS. 2022;19(1):46.PubMedPubMedCentralCrossRef
47.
go back to reference Sifat AE, et al. Prenatal electronic cigarette exposure decreases brain glucose utilization and worsens outcome in offspring hypoxic-ischemic brain injury. J Neurochem. 2020;153(1):63–79.PubMedCrossRef Sifat AE, et al. Prenatal electronic cigarette exposure decreases brain glucose utilization and worsens outcome in offspring hypoxic-ischemic brain injury. J Neurochem. 2020;153(1):63–79.PubMedCrossRef
48.
go back to reference Villalba H, et al. Potential role of myo-inositol to improve ischemic stroke outcome in diabetic mouse. Brain Res. 2018;1699:166–76.PubMedCrossRef Villalba H, et al. Potential role of myo-inositol to improve ischemic stroke outcome in diabetic mouse. Brain Res. 2018;1699:166–76.PubMedCrossRef
49.
go back to reference Swiergiel AH, Dunn AJ. Feeding, exploratory, anxiety- and depression-related behaviors are not altered in interleukin-6-deficient male mice. Behav Brain Res. 2006;171(1):94–108.PubMedPubMedCentralCrossRef Swiergiel AH, Dunn AJ. Feeding, exploratory, anxiety- and depression-related behaviors are not altered in interleukin-6-deficient male mice. Behav Brain Res. 2006;171(1):94–108.PubMedPubMedCentralCrossRef
51.
go back to reference Chan YL, et al. Maternal cigarette smoke exposure worsens neurological outcomes in adolescent offspring with hypoxic-ischemic injury. Front Mol Neurosci. 2017;10:306.PubMedPubMedCentralCrossRef Chan YL, et al. Maternal cigarette smoke exposure worsens neurological outcomes in adolescent offspring with hypoxic-ischemic injury. Front Mol Neurosci. 2017;10:306.PubMedPubMedCentralCrossRef
52.
54.
go back to reference Cora MC, Kooistra L, Travlos G. Vaginal cytology of the laboratory rat and mouse: review and criteria for the staging of the estrous cycle using stained vaginal smears. Toxicol Pathol. 2015;43(6):776–93.PubMedCrossRef Cora MC, Kooistra L, Travlos G. Vaginal cytology of the laboratory rat and mouse: review and criteria for the staging of the estrous cycle using stained vaginal smears. Toxicol Pathol. 2015;43(6):776–93.PubMedCrossRef
55.
go back to reference Roy TS, Seidler FJ, Slotkin TA. Prenatal nicotine exposure evokes alterations of cell structure in hippocampus and somatosensory cortex. J Pharmacol Exp Ther. 2002;300(1):124–33.PubMedCrossRef Roy TS, Seidler FJ, Slotkin TA. Prenatal nicotine exposure evokes alterations of cell structure in hippocampus and somatosensory cortex. J Pharmacol Exp Ther. 2002;300(1):124–33.PubMedCrossRef
56.
go back to reference El Marroun H, et al. Prenatal tobacco exposure and brain morphology: a prospective study in young children. Neuropsychopharmacology. 2014;39(4):792–800.PubMedCrossRef El Marroun H, et al. Prenatal tobacco exposure and brain morphology: a prospective study in young children. Neuropsychopharmacology. 2014;39(4):792–800.PubMedCrossRef
57.
58.
go back to reference Nguyen T, et al. Maternal e-cigarette exposure results in cognitive and epigenetic alterations in offspring in a mouse model. Chem Res Toxicol. 2018;31(7):601–11.PubMedCrossRef Nguyen T, et al. Maternal e-cigarette exposure results in cognitive and epigenetic alterations in offspring in a mouse model. Chem Res Toxicol. 2018;31(7):601–11.PubMedCrossRef
59.
go back to reference Smith D, et al. Adult behavior in male mice exposed to e-cigarette nicotine vapors during late prenatal and early postnatal life. PLoS ONE. 2015;10(9):e0137953.PubMedPubMedCentralCrossRef Smith D, et al. Adult behavior in male mice exposed to e-cigarette nicotine vapors during late prenatal and early postnatal life. PLoS ONE. 2015;10(9):e0137953.PubMedPubMedCentralCrossRef
60.
go back to reference Hawkins BT, et al. Nicotine increases in vivo blood-brain barrier permeability and alters cerebral microvascular tight junction protein distribution. Brain Res. 2004;1027(1–2):48–58.PubMedCrossRef Hawkins BT, et al. Nicotine increases in vivo blood-brain barrier permeability and alters cerebral microvascular tight junction protein distribution. Brain Res. 2004;1027(1–2):48–58.PubMedCrossRef
61.
go back to reference Kadry H, et al. Comparative assessment of in vitro BBB tight junction integrity following exposure to cigarette smoke and e-cigarette vapor: a quantitative evaluation of the protective effects of metformin using small-molecular-weight paracellular markers. Fluids Barriers CNS. 2021;18(1):28.PubMedPubMedCentralCrossRef Kadry H, et al. Comparative assessment of in vitro BBB tight junction integrity following exposure to cigarette smoke and e-cigarette vapor: a quantitative evaluation of the protective effects of metformin using small-molecular-weight paracellular markers. Fluids Barriers CNS. 2021;18(1):28.PubMedPubMedCentralCrossRef
63.
go back to reference Prasad S, et al. Impact of cigarette smoke extract and hyperglycemic conditions on blood–brain barrier endothelial cells. Fluids Barriers CNS. 2015;12(1):18.PubMedPubMedCentralCrossRef Prasad S, et al. Impact of cigarette smoke extract and hyperglycemic conditions on blood–brain barrier endothelial cells. Fluids Barriers CNS. 2015;12(1):18.PubMedPubMedCentralCrossRef
64.
go back to reference Jia W, et al. The role of claudin-5 in blood-brain barrier (BBB) and brain metastases (review). Mol Med Rep. 2014;9(3):779–85.PubMedCrossRef Jia W, et al. The role of claudin-5 in blood-brain barrier (BBB) and brain metastases (review). Mol Med Rep. 2014;9(3):779–85.PubMedCrossRef
65.
go back to reference Liu C, Wu J, Zou MH. Activation of AMP-activated protein kinase alleviates high-glucose-induced dysfunction of brain microvascular endothelial cell tight-junction dynamics. Free Radic Biol Med. 2012;53(6):1213–21.PubMedPubMedCentralCrossRef Liu C, Wu J, Zou MH. Activation of AMP-activated protein kinase alleviates high-glucose-induced dysfunction of brain microvascular endothelial cell tight-junction dynamics. Free Radic Biol Med. 2012;53(6):1213–21.PubMedPubMedCentralCrossRef
66.
go back to reference Balbuena P, Li W, Ehrich M. Assessments of tight junction proteins occludin, claudin 5 and scaffold proteins ZO1 and ZO2 in endothelial cells of the rat blood–brain barrier: cellular responses to neurotoxicants malathion and lead acetate. Neurotoxicology. 2011;32(1):58–67.PubMedCrossRef Balbuena P, Li W, Ehrich M. Assessments of tight junction proteins occludin, claudin 5 and scaffold proteins ZO1 and ZO2 in endothelial cells of the rat blood–brain barrier: cellular responses to neurotoxicants malathion and lead acetate. Neurotoxicology. 2011;32(1):58–67.PubMedCrossRef
67.
go back to reference Zlokovic BV. The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron. 2008;57(2):178–201.PubMedCrossRef Zlokovic BV. The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron. 2008;57(2):178–201.PubMedCrossRef
68.
go back to reference Hartz AM, Bauer B. Regulation of ABC transporters at the blood-brain barrier: new targets for CNS therapy. Mol Interv. 2010;10(5):293–304.PubMedCrossRef Hartz AM, Bauer B. Regulation of ABC transporters at the blood-brain barrier: new targets for CNS therapy. Mol Interv. 2010;10(5):293–304.PubMedCrossRef
69.
go back to reference Zhang Q, et al. Increase in blood-brain barrier (BBB) permeability is regulated by MMP3 via the ERK signaling pathway. Oxid Med Cell Longev. 2021;2021:6655122.PubMedPubMedCentral Zhang Q, et al. Increase in blood-brain barrier (BBB) permeability is regulated by MMP3 via the ERK signaling pathway. Oxid Med Cell Longev. 2021;2021:6655122.PubMedPubMedCentral
70.
71.
go back to reference Kamm DR. The Effects of diet and sex differences on cortical tight-junction protein expression in senescence-accelerated mouse-prone 8 (SAMP8) mice. Ann Arbor: Southern Illinois University at Edwardsville; 2019. Kamm DR. The Effects of diet and sex differences on cortical tight-junction protein expression in senescence-accelerated mouse-prone 8 (SAMP8) mice. Ann Arbor: Southern Illinois University at Edwardsville; 2019.
72.
go back to reference Archie SR, et al. Biological determinants impact the neurovascular toxicity of nicotine and tobacco smoke: a pharmacokinetic and pharmacodynamics perspective. Neurotoxicology. 2022;89:140–60.PubMedCrossRef Archie SR, et al. Biological determinants impact the neurovascular toxicity of nicotine and tobacco smoke: a pharmacokinetic and pharmacodynamics perspective. Neurotoxicology. 2022;89:140–60.PubMedCrossRef
73.
go back to reference Bosworth AP, Allen NJ. The diverse actions of astrocytes during synaptic development. Curr Opin Neurobiol. 2017;47:38–43.PubMedCrossRef Bosworth AP, Allen NJ. The diverse actions of astrocytes during synaptic development. Curr Opin Neurobiol. 2017;47:38–43.PubMedCrossRef
74.
go back to reference Kumar P, et al. Mechanisms involved in epigenetic down-regulation of Gfap under maternal hypothyroidism. Biochem Biophys Res Commun. 2018;502(3):375–81.PubMedCrossRef Kumar P, et al. Mechanisms involved in epigenetic down-regulation of Gfap under maternal hypothyroidism. Biochem Biophys Res Commun. 2018;502(3):375–81.PubMedCrossRef
75.
go back to reference Hughes EG, et al. Loss of glial fibrillary acidic protein results in decreased glutamate transport and inhibition of PKA-induced EAAT2 cell surface trafficking. Brain Res Mol Brain Res. 2004;124(2):114–23.PubMedCrossRef Hughes EG, et al. Loss of glial fibrillary acidic protein results in decreased glutamate transport and inhibition of PKA-induced EAAT2 cell surface trafficking. Brain Res Mol Brain Res. 2004;124(2):114–23.PubMedCrossRef
76.
go back to reference Li Y-K, et al. Aquaporin-4 deficiency impairs synaptic plasticity and associative fear memory in the lateral amygdala: involvement of downregulation of glutamate transporter-1 expression. Neuropsychopharmacology. 2012;37(8):1867–78.PubMedPubMedCentralCrossRef Li Y-K, et al. Aquaporin-4 deficiency impairs synaptic plasticity and associative fear memory in the lateral amygdala: involvement of downregulation of glutamate transporter-1 expression. Neuropsychopharmacology. 2012;37(8):1867–78.PubMedPubMedCentralCrossRef
77.
go back to reference Rozovsky I, et al. Estradiol (E2) enhances neurite outgrowth by repressing glial fibrillary acidic protein expression and reorganizing laminin. Endocrinology. 2002;143(2):636–46.PubMedCrossRef Rozovsky I, et al. Estradiol (E2) enhances neurite outgrowth by repressing glial fibrillary acidic protein expression and reorganizing laminin. Endocrinology. 2002;143(2):636–46.PubMedCrossRef
78.
go back to reference Day JR, et al. Gonadal steroids regulate the expression of glial fibrillary acidic protein in the adult male rat hippocampus. Neuroscience. 1993;55(2):435–43.PubMedCrossRef Day JR, et al. Gonadal steroids regulate the expression of glial fibrillary acidic protein in the adult male rat hippocampus. Neuroscience. 1993;55(2):435–43.PubMedCrossRef
79.
go back to reference Stone DJ, et al. Bidirectional transcription regulation of glial fibrillary acidic protein by estradiol in vivo and in vitro*. Endocrinology. 1998;139(7):3202–9.PubMedCrossRef Stone DJ, et al. Bidirectional transcription regulation of glial fibrillary acidic protein by estradiol in vivo and in vitro*. Endocrinology. 1998;139(7):3202–9.PubMedCrossRef
80.
go back to reference Mader S, Brimberg L. Aquaporin-4 water channel in the brain and its implication for health and disease. Cells. 2019;8:2.CrossRef Mader S, Brimberg L. Aquaporin-4 water channel in the brain and its implication for health and disease. Cells. 2019;8:2.CrossRef
82.
go back to reference Huang L, et al. Maternal smoking and attention-deficit/hyperactivity disorder in offspring: a meta-analysis. Pediatrics. 2018;141:1.CrossRef Huang L, et al. Maternal smoking and attention-deficit/hyperactivity disorder in offspring: a meta-analysis. Pediatrics. 2018;141:1.CrossRef
83.
go back to reference He Y, et al. Maternal smoking during pregnancy and ADHD: results from a systematic review and meta-analysis of prospective cohort studies. J Atten Disord. 2020;24(12):1637–47.PubMedCrossRef He Y, et al. Maternal smoking during pregnancy and ADHD: results from a systematic review and meta-analysis of prospective cohort studies. J Atten Disord. 2020;24(12):1637–47.PubMedCrossRef
84.
go back to reference Gustavson K, et al. Smoking in pregnancy and child ADHD. Pediatrics. 2017;139:2.CrossRef Gustavson K, et al. Smoking in pregnancy and child ADHD. Pediatrics. 2017;139:2.CrossRef
85.
go back to reference Elliott BM, et al. Effects of nicotine on elevated plus maze and locomotor activity in male and female adolescent and adult rats. Pharmacol Biochem Behav. 2004;77(1):21–8.PubMedCrossRef Elliott BM, et al. Effects of nicotine on elevated plus maze and locomotor activity in male and female adolescent and adult rats. Pharmacol Biochem Behav. 2004;77(1):21–8.PubMedCrossRef
86.
go back to reference Caldarone BJ, King SL, Picciotto MR. Sex differences in anxiety-like behavior and locomotor activity following chronic nicotine exposure in mice. Neurosci Lett. 2008;439(2):187–91.PubMedPubMedCentralCrossRef Caldarone BJ, King SL, Picciotto MR. Sex differences in anxiety-like behavior and locomotor activity following chronic nicotine exposure in mice. Neurosci Lett. 2008;439(2):187–91.PubMedPubMedCentralCrossRef
88.
go back to reference Batty GD, Der G, Deary IJ. Effect of maternal smoking during pregnancy on offspring’s cognitive ability: empirical evidence for complete confounding in the US national longitudinal survey of youth. Pediatrics. 2006;118(3):943–50.PubMedCrossRef Batty GD, Der G, Deary IJ. Effect of maternal smoking during pregnancy on offspring’s cognitive ability: empirical evidence for complete confounding in the US national longitudinal survey of youth. Pediatrics. 2006;118(3):943–50.PubMedCrossRef
89.
go back to reference Julvez J, et al. Maternal smoking habits and cognitive development of children at age 4 years in a population-based birth cohort. Int J Epidemiol. 2007;36(4):825–32.PubMedCrossRef Julvez J, et al. Maternal smoking habits and cognitive development of children at age 4 years in a population-based birth cohort. Int J Epidemiol. 2007;36(4):825–32.PubMedCrossRef
90.
go back to reference Micalizzi L, Knopik VS. Maternal smoking during pregnancy and offspring executive function: What do we know and what are the next steps? Dev Psychopathol. 2018;30(4):1333–54.PubMedCrossRef Micalizzi L, Knopik VS. Maternal smoking during pregnancy and offspring executive function: What do we know and what are the next steps? Dev Psychopathol. 2018;30(4):1333–54.PubMedCrossRef
91.
go back to reference Benowitz NL, Hukkanen J, Jacob P 3rd. Nicotine chemistry, metabolism, kinetics and biomarkers. Handb Exp Pharmacol. 2009;192:29–60.CrossRef Benowitz NL, Hukkanen J, Jacob P 3rd. Nicotine chemistry, metabolism, kinetics and biomarkers. Handb Exp Pharmacol. 2009;192:29–60.CrossRef
92.
go back to reference Hoegberg BG, et al. Regulation of α4β2α5 nicotinic acetylcholinergic receptors in rat cerebral cortex in early and late adolescence: Sex differences in response to chronic nicotine. Neuropharmacology. 2015;99:347–55.PubMedPubMedCentralCrossRef Hoegberg BG, et al. Regulation of α4β2α5 nicotinic acetylcholinergic receptors in rat cerebral cortex in early and late adolescence: Sex differences in response to chronic nicotine. Neuropharmacology. 2015;99:347–55.PubMedPubMedCentralCrossRef
93.
Metadata
Title
Maternal e-cigarette use can disrupt postnatal blood-brain barrier (BBB) integrity and deteriorates motor, learning and memory function: influence of sex and age
Authors
Sabrina Rahman Archie
Ali Ehsan Sifat
Yong Zhang
Heidi Villalba
Sejal Sharma
Saeideh Nozohouri
Thomas J. Abbruscato
Publication date
01-12-2023
Publisher
BioMed Central
Published in
Fluids and Barriers of the CNS / Issue 1/2023
Electronic ISSN: 2045-8118
DOI
https://doi.org/10.1186/s12987-023-00416-5

Other articles of this Issue 1/2023

Fluids and Barriers of the CNS 1/2023 Go to the issue