1
|
Shenassa ED, Botteri E, Stensheim H. Feeding Method, Nicotine Exposure, and Growth during Infancy. JOURNAL OF PEDIATRICS. CLINICAL PRACTICE 2024; 14:200127. [PMID: 39950049 PMCID: PMC11824624 DOI: 10.1016/j.jpedcp.2024.200127] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 02/16/2025]
Abstract
Objective To answer 3 questions: (1) Are infants breastfed by smokers at risk of rapid weight and length gain? (2) Is rapid growth during infancy partially attributable to ingestion of smokers' breastmilk? (3) If so, what are the implications for breastfeeding by smokers? Study design Using data from the Norwegian Mother, Father and Child Cohort Study and Medical Birth Registry of Norway (n = 54 522), we examined changes in weight, length, weight-for-length z-score (WFLZ) during infancy in the context of maternal smoking (0, 1-10, or >10 cigarettes/day) and feeding method during the first 6 months (breastfed, formula fed, mixed fed). We fit generalized linear models, adding a smoking by feeding method interaction term to evaluate the effect of ingesting smokers' breastmilk. Results Breastfed infants of both light and heavy smokers experienced WFLZ gains of 0.05 (95% CI, 0.01-0.09) and 0.13 (95% CI, 0.07-0.18), respectively. Among mixed-fed infants, only heavy maternal smoking predicted WFLZ gain (0.10; 95% CI, 0.05-0.16). Among exclusively formula-fed infants, maternal smoking did not predict rapid growth. Interaction models suggest that infants ever breastfed (ie, breastfed and mixed-fed groups combined) by heavy smokers gained weight (100 g; 95% CI, 30-231) and length (2.8 mm; 95% CI, 0.1-5.6), attributable to ingesting smoker's breastmilk. Conclusions Infants breastfed by smokers experience rapid growth; some of these gains are attributable to ingesting smokers' breastmilk. Among infants breasted by light smokers, these gains are within the range of normative growth patterns for healthy, breastfed infants.
Collapse
Affiliation(s)
- Edmond D. Shenassa
- Department of Epidemiology and Biostatistics, University of Maryland, College Park, MD
- Department of Epidemiology and Biostatistics, School of Public Health, Brown University, Providence, RI
- Department of Epidemiology and Biostatistics, School of Medicine, University of Maryland, Baltimore, MD
| | - Edoardo Botteri
- Department of Research, Cancer Registry of Norway, Oslo, Norway
- Section for Colorectal Cancer Screening, Cancer Registry of Norway, Oslo, Norway
| | - Hanne Stensheim
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| |
Collapse
|
2
|
Souza LL, Rossetti CL, Peixoto TC, Manhães AC, de Moura EG, Lisboa PC. Neonatal nicotine exposure affects adult rat hepatic pathways involved in endoplasmic reticulum stress and macroautophagy in a sex-dependent manner. J Dev Orig Health Dis 2023; 14:639-647. [PMID: 38037831 DOI: 10.1017/s2040174423000326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) involves changes in hepatic pathways, as lipogenesis, oxidative stress, endoplasmic reticulum (ER) stress, and macroautophagy. Maternal nicotine exposure exclusively during lactation leads to fatty liver (steatosis) only in the adult male offspring, not in females. Therefore, our hypothesis is that neonatal exposure to nicotine sex-dependently affects the signaling pathways involved in hepatic homeostasis of the offspring, explaining the hepatic lipid accumulation phenotype only in males. For this, between postnatal days 2 and 16, Wistar rat dams were implanted with osmotic minipumps, which released nicotine (NIC; 6 mg/Kg/day) or vehicle. The livers of offspring were evaluated at postnatal day 180. Only the male offspring that had been exposed to nicotine neonatally showed increased protein expression of markers of unfolded protein response (UPR), highlighting the presence of ER stress, as well as disruption of the activation of the macroautophagy repair pathway. These animals also had increased expression of diacylglycerol O-acyltransferase 1 and 4-hydroxynonenal, suggesting increased triglyceride esterification and oxidative stress. These parameters were not altered in the female offspring that had been neonatally exposed to nicotine, however they exhibited increased phospho adenosine monophosphate-activated protein kinase pAMPK expression, possibly as a protective mechanism. Thus, the disturbance in the hepatic homeostasis by UPR, macroautophagy, and oxidative stress modifications seem to be the molecular mechanisms underlying the liver steatosis in the adult male offspring of the nicotine-programming model. This highlights the importance of maternal smoking cessation during breastfeeding to decrease the risk of NAFLD development, especially in males.
Collapse
Affiliation(s)
- Luana Lopes Souza
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Camila Lüdke Rossetti
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thamara Cherem Peixoto
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alex Christian Manhães
- Laboratory of Neurophysiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Egberto Gaspar de Moura
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia Cristina Lisboa
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Nicotine exposure during breastfeeding alters the expression of endocannabinoid system biomarkers in female but not in male offspring at adulthood. J Dev Orig Health Dis 2023; 14:415-425. [PMID: 36815400 DOI: 10.1017/s2040174423000028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Early nicotine exposure compromises offspring's phenotype at long-term in both sexes. We hypothesize that offspring exposed to nicotine during breastfeeding show deregulated central and peripheral endocannabinoid system (ECS), compromising several aspects of their metabolism. Lactating rats received nicotine (NIC, 6 mg/Kg/day) or saline from postnatal day (PND) 2 to 16 through implanted osmotic minipumps. Offspring were analyzed at PND180. We evaluated protein expression of N-acylphosphatidylethanolamide-phospholipase D (NAPE-PLD), fatty acid amide hydrolase (FAAH), diacylglycerol lipase (DAGL), monoacylglycerol lipase (MAGL) and cannabinoid receptors (CB1 and/or CB2) in lateral hypothalamus, paraventricular nucleus of the hypothalamus, liver, visceral adipose tissue (VAT), adrenal and thyroid. NIC offspring from both sexes did not show differences in hypothalamic ECS markers. Peripheral ECS markers showed no alterations in NIC males. In contrast, NIC females had lower liver DAGL and CB1, higher VAT DAGL, higher adrenal NAPE-PLD and higher thyroid FAAH. Endocannabinoids biosynthesis was affected by nicotine exposure during breastfeeding only in females; alterations in peripheral tissues suggest lower action in liver and higher action in VAT, adrenal and thyroid. Effects of nicotine exposure during lactation on ECS markers are sex- and tissue-dependent. This characterization helps understanding the phenotype of the adult offspring in this model and may contribute to the development of new pharmacological targets for the treatment of several metabolic diseases that originate during development.
Collapse
|
4
|
Miranda RA, de Moura EG, Lisboa PC. Adverse perinatal conditions and the developmental origins of thyroid dysfunction-Lessons from Animal Models. Endocrine 2023; 79:223-234. [PMID: 36036880 DOI: 10.1007/s12020-022-03177-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/17/2022] [Indexed: 02/04/2023]
Abstract
PURPOSE Nutritional, hormonal, and environmental status during development can predispose the individual to obesity and endocrine diseases later in life, an association known as metabolic programming. In general, weight loss or gain are seen in thyroid disorders, and thyroid function can be affected by body adiposity. In addition, hyper- and hypothyroidism can be related to metabolic programming. Our aim was to gather evidence that regardless of the type or critical window of metabolic imprinting, offspring exposed to certain adverse perinatal conditions have a higher risk of developing thyroid dysfunction. METHODS We reviewed literature data that relate insults occurring during pregnancy and/or lactation to short- and long-term offspring thyroid dysfunction in animal models. RESULTS Few studies have addressed the hypothalamic-pituitary-thyroid axis and thyroid dysfunction related to metabolic programming. The literature shows that under- and overnutrition, exposure to endocrine disruptors, early weaning, maternal thyroid disease and maternal high-fat diet can induce alterations in offspring thyroid function in a sex-dependent manner. CONCLUSION Based on the few available data, mainly in rodent models, we can conclude that diet, hormones, and environmental contaminants are related to the developmental origins of later thyroid dysfunction by interrupting the normal maturation of the thyroid gland.
Collapse
Affiliation(s)
- Rosiane Aparecida Miranda
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Egberto Gaspar de Moura
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Patrícia Cristina Lisboa
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
5
|
Amaro A, Baptista FI, Matafome P. Programming of future generations during breastfeeding: The intricate relation between metabolic and neurodevelopment disorders. Life Sci 2022; 298:120526. [DOI: 10.1016/j.lfs.2022.120526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/27/2022]
|
6
|
Jaakkola JM, Rovio SP, Pahkala K, Viikari J, Rönnemaa T, Jula A, Niinikoski H, Mykkänen J, Juonala M, Hutri-Kähönen N, Kähönen M, Lehtimäki T, Raitakari OT. Childhood exposure to parental smoking and life-course overweight and central obesity. Ann Med 2021; 53:208-216. [PMID: 33305629 PMCID: PMC7901689 DOI: 10.1080/07853890.2020.1853215] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 11/14/2020] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE To evaluate the association between childhood parental smoking exposure and the risk of overweight/obesity from childhood to adulthood. METHODS This study leverages the data from two longitudinal population based cohort studies, the Cardiovascular Risk in Young Finns Study between years 1980-2011/2012 (YFS; N = 2,303; baseline age 3-18 years) and the Special Turku Coronary Risk Factor Intervention Project between years 1989-2009/2010 (STRIP; N = 632; baseline age 7 months). Weight, height and waist circumference were measured from childhood to adulthood. Overweight/obesity was defined as body mass index ≥25 kg/m2 in adults and using the Cole criteria in children. Central obesity was defined as waist circumference > 100/90 cm in men/women and as a waist-to-height ratio > 0.50 in children. Statistical analyses were adjusted for age, sex, socioeconomic status, smoking, birth weight, parental ages, diet and physical activity. RESULTS Childhood parental smoking exposure was associated with increased risk for life-course overweight/obesity (YFS: RR1.13, 95%CI 1.02-1.24; STRIP: RR1.57, 95%CI 1.10-2.26) and central obesity (YFS: RR1.18, 95%CI 1.01-1.38; STRIP: RR1.45, 95%CI 0.98-2.15). CONCLUSIONS Childhood exposure to parental smoking is associated with increased risk of overweight/obesity over the life-course. KEY MESSAGES Exposure to parental smoking in childhood was associated with increased risk of overweight/obesity, central obesity and adiposity measured by skinfold thickness from childhood to adulthood.
Collapse
Affiliation(s)
- Johanna M. Jaakkola
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Suvi P. Rovio
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Katja Pahkala
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Paavo Nurmi Centre, Sports and Exercise Medicine Unit, Department of Health and Physical activity, University of Turku, Turku, Finland
| | - Jorma Viikari
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Tapani Rönnemaa
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Antti Jula
- Department of Chronic Disease Prevention, Institute for Health and Welfare, Turku, Finland
| | - Harri Niinikoski
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Juha Mykkänen
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Markus Juonala
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Nina Hutri-Kähönen
- Department of Pediatrics, Tampere University Hospital, and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Olli T. Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| |
Collapse
|
7
|
Lisboa PC, Miranda RA, Souza LL, Moura EG. Can breastfeeding affect the rest of our life? Neuropharmacology 2021; 200:108821. [PMID: 34610290 DOI: 10.1016/j.neuropharm.2021.108821] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/17/2021] [Accepted: 09/30/2021] [Indexed: 12/31/2022]
Abstract
The breastfeeding period is one of the most important critical windows in our development, since milk, our first food after birth, contains several compounds, such as macronutrients, micronutrients, antibodies, growth factors and hormones that benefit human health. Indeed, nutritional, and environmental alterations during lactation, change the composition of breast milk and induce alterations in the child's development, such as obesity, leading to the metabolic dysfunctions, cardiovascular diseases and neurobehavioral disorders. This review is based on experimental animal models, most of them in rodents, and summarizes the impact of an adequate breast milk supply in view of the developmental origins of health and disease (DOHaD) concept, which has been proposed by researchers in the areas of epidemiology and basic science from around the world. Here, experimental advances in understanding the programming during breastfeeding were compiled with the purpose of generating knowledge about the genesis of chronic noncommunicable diseases and to guide the development of public policies to deal with and prevent the problems arising from this phenomenon. This review article is part of the special issue on "Cross talk between periphery and brain".
Collapse
Affiliation(s)
- Patricia C Lisboa
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Rosiane A Miranda
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luana L Souza
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Egberto G Moura
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
8
|
Rodrigues VST, Moura EG, Peixoto TC, Soares PN, Lopes BP, Oliveira E, Manhães AC, Atella GC, Kluck GEG, Cabral SS, Trindade PL, Daleprane JB, Lisboa PC. Changes in gut-brain axis parameters in adult rats of both sexes with different feeding pattern that were early nicotine-exposed. Food Chem Toxicol 2021; 158:112656. [PMID: 34740714 DOI: 10.1016/j.fct.2021.112656] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 10/20/2021] [Accepted: 10/29/2021] [Indexed: 01/09/2023]
Abstract
Nicotine is an endocrine disruptor and imprinting factor during breastfeeding that can cause food intake imbalance in the adulthood. As nicotine affects the intestinal microbiota, altering the composition of the bacterial communities and short-chain fatty acids (SCFAs) synthesis in a sex-dependent manner, we hypothesized that nicotine could program the gut-brain axis, consequently modifying the eating pattern of adult male and female rats in a model of maternal nicotine exposure (MNE) during breastfeeding. Lactating Wistar rat dams received minipumps that release 6 mg/kg/day of nicotine (MNE group) or saline for 14 days. The progeny received standard diet from weaning until euthanasia (26 weeks of age). We measured: in vivo electrical activity of the vagus nerve; c-Fos expression in the nucleus tractus solitarius, gastrointestinal peptides receptors, intestinal brain-derived neurotrophic factor (BDNF), SCFAs and microbiota. MNE females showed hyperphagia despite normal adiposity, while MNE males had unchanged food intake, despite obesity. Adult MNE offspring showed decreased Bacteroidetes and increased Firmicutes, Actinobacteria and Proteobacteria. MNE females had lower fecal acetate while MNE males showed higher vagus nerve activity. In summary nicotine exposure through the milk induces long-term intestinal dysbiosis, which may affect eating patterns of adult offspring in a sex-dependent manner.
Collapse
Affiliation(s)
- V S T Rodrigues
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - E G Moura
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - T C Peixoto
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - P N Soares
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - B P Lopes
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - E Oliveira
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - A C Manhães
- Neurophysiology Laboratory, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - G C Atella
- Laboratory of Lipids and Lipoprotein Biochemistry, Biochemistry Institute, Federal University of Rio de Janeiro, RJ, Brazil
| | - G E G Kluck
- Laboratory of Lipids and Lipoprotein Biochemistry, Biochemistry Institute, Federal University of Rio de Janeiro, RJ, Brazil
| | - S S Cabral
- Laboratory of Lipids and Lipoprotein Biochemistry, Biochemistry Institute, Federal University of Rio de Janeiro, RJ, Brazil
| | - P L Trindade
- Laboratory for Studies of Interactions Between Nutrition and Genetics, Nutrition Institute, Rio de Janeiro State University, RJ, Brazil
| | - J B Daleprane
- Laboratory for Studies of Interactions Between Nutrition and Genetics, Nutrition Institute, Rio de Janeiro State University, RJ, Brazil
| | - P C Lisboa
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
9
|
Al-Sawalha NA, Bdeir R, Sohaib A, Saad M, Inghaimesh T, Khabour OF, Alzoubi KH, Shihadeh A. Effect of E-cigarettes aerosol exposure during lactation in rats: Hormonal and biochemical aspects. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 88:103759. [PMID: 34695539 PMCID: PMC8957699 DOI: 10.1016/j.etap.2021.103759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/07/2021] [Accepted: 10/20/2021] [Indexed: 05/03/2023]
Abstract
Electronic cigarettes (e-cigarettes) have been marketed as a less lethal substitute for smoking traditional cigarettes. This study aims to investigate the impact of e-cigarettes aerosol exposure on lactating dams and pups, whose dams were exposed. Lactating dams received fresh air (control) or e-cigarettes aerosol during lactation (day 4-21). Maternal exposure to e-cigarettes aerosol during lactation induced significant reduction (P < 0.0001) in the fat content of the milk and serum Leptin level (P < 0.005) compared to control dams. Furthermore, pups whose dams were exposed to e-cigarettes during lactation showed an increased level of glucose, thyroxine and decreased level of insulin. The exposure to e-cigarettes aerosol during lactation altered the composition of milk as well as the hormonal and biochemical profile in dams and pups. This result, if observed in women using e-cigarettes, suggests that e-cigarettes' use during lactation may have consequences on the milk production and hormonal and biochemical profile in breastfeeding mothers and nursing babies.
Collapse
Affiliation(s)
- Nour A Al-Sawalha
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan.
| | - Roba Bdeir
- Faculty of Pharmacy, Jadara University, Irbid, Jordan
| | - Aiman Sohaib
- Faculty of Pharmacy, Jadara University, Irbid, Jordan
| | - Marwan Saad
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Tasneem Inghaimesh
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Omar F Khabour
- Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, UAE; Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Alan Shihadeh
- Mechanical Engineering Department, American University of Beirut, Beirut 1107 2020, Lebanon; Center for the Study of Tobacco Products, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
10
|
Di Dalmazi G, Giuliani C. Plant constituents and thyroid: A revision of the main phytochemicals that interfere with thyroid function. Food Chem Toxicol 2021; 152:112158. [PMID: 33789121 DOI: 10.1016/j.fct.2021.112158] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 01/06/2023]
Abstract
In the past few decades, there has been a lot of interest in plant constituents for their antioxidant, anti-inflammatory, anti-microbial and anti-proliferative properties. However, concerns have been raised on their potential toxic effects particularly when consumed at high dose. The anti-thyroid effects of some plant constituents have been known for some time. Indeed, epidemiological observations have shown the causal association between staple food based on brassicaceae or soybeans and the development of goiter and/or hypothyroidism. Herein, we review the main plant constituents that interfere with normal thyroid function such as cyanogenic glucosides, polyphenols, phenolic acids, and alkaloids. In detail, we summarize the in vitro and in vivo studies present in the literature, focusing on the compounds that are more abundant in foods or that are available as dietary supplements. We highlight the mechanism of action of these compounds on thyroid cells by giving a particular emphasis to the experimental studies that can be significant for human health. Furthermore, we reveal that the anti-thyroid effects of these plant constituents are clinically evident only when they are consumed in very large amounts or when their ingestion is associated with other conditions that impair thyroid function.
Collapse
Affiliation(s)
- Giulia Di Dalmazi
- Center for Advanced Studies and Technology (CAST) and Department of Medicine and Aging Science, University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy; Department of Medicine and Aging Science, Translational Medicine PhD Program, University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy.
| | - Cesidio Giuliani
- Center for Advanced Studies and Technology (CAST) and Department of Medicine and Aging Science, University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy.
| |
Collapse
|
11
|
Neonatal nicotine exposure changes insulin status in fat depots: sex-related differences. J Dev Orig Health Dis 2021; 13:252-262. [PMID: 33818369 DOI: 10.1017/s2040174421000131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nicotine is the main psychoactive substance present in cigarette smoke that is transferred to the baby by breast milk. In rats, maternal nicotine exposure during breastfeeding induces obesogenesis and hormone dysfunctions in adult male offspring. As glucocorticoid (GC), insulin, and vitamin D change both adipogenesis and lipogenesis processes, we assessed parameters related to metabolism and action of these hormones in visceral and subcutaneous adipose tissues (VAT and SAT) of adult male and female rats in a model of neonatal nicotine exposure. At postnatal (PN) day 2, dams were kept with six pups (three per sex) and divided into nicotine and control groups for implantation of osmotic minipumps that released 6 mg/kg nicotine or saline, respectively. At PN180, fat mass, hormone levels, and protein contents of biomarkers of the GC activation and receptor (11beta-hydroxysteroid dehydrogenase type 1 and glucocorticoid receptor alpha), insulin signaling pathway [insulin receptor beta (IRβ), phosphorylated insulin receptor substrate 1, insulin receptor substrate 1 (IRS1), phosphorylated serine/threonine kinase (pAKT), serine/threonine kinase, glucose transporter type 4 (GLUT4)], and vitamin D activation and receptor (1α-hydroxylase and vitamin D receptor) were evaluated. While nicotine-exposed males showed increased fat mass, hypercorticosteronemia, hyperinsulinemia, and higher 25-hydroxyvitamin D, these alterations were not observed in nicotine-exposed females. Nicotine-exposed males only showed lower IRS1 in VAT, while the females had hyperglycemia, higher pAKT in VAT, while lower IRβ, IRS1, and GLUT4 in SAT. Parameters related to metabolism and action of GC and vitamin D were unaltered in both sexes. We evidence that exposure exclusively to nicotine during breastfeeding affects the hormone status and fat depots of the adult progeny in a sex-dependent manner.
Collapse
|
12
|
Pietrobon CB, Lisboa PC, Bertasso IM, Peixoto TC, Soares PN, de Oliveira E, Rabelo K, de Carvalho JJ, Manhães AC, de Moura EG. Pancreatic steatosis in adult rats induced by nicotine exposure during breastfeeding. Endocrine 2021; 72:104-115. [PMID: 33420949 DOI: 10.1007/s12020-020-02579-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 11/27/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE Maternal nicotine exposure negatively impacts offspring's health and metabolism, leading to obesity and insulin resistance. Here we investigated the pancreatic islet function, glycemic homeostasis, and insulin signaling in adult rat offspring that were nicotine-exposed during breastfeeding. METHODS For this, lactating Wistar rat dams were divided into two groups: Nicotine (implanted with osmotic minipumps containing 6 mg/Kg, NIC) and Control (saline, CON). Solutions were released from postnatal (PN) day 2-16. At PN110 and PN170, 10 offspring per litter/sex/group were submitted to the oral glucose tolerance test (OGTT). PN180 offspring were killed and glycemia, insulinemia, adiponectinemia, pancreas morphology as well as pancreatic islet protein expression (related to insulin secretion) and skeletal muscle (related to insulin action) were evaluated. Males and females were compared to their respective controls. RESULTS Adult NIC offspring of both sexes showed glucose intolerance in the OGTT. Despite normoglycemia, NIC males showed hyperinsulinemia while females, although normoinsulinemic, had hyperglycemia. Both sexes showed increased IRI, reduced adiponectin/visceral fat mass ratio and higher ectopic deposition of lipids in the pancreatic tissue adipocytes. In pancreatic islets, NIC males showed lower PDX-1 expression while females had higher PDX-1 and GLUT2 expressions plus lower α2 adrenergic receptor. In the muscle, NIC offspring of both sexes showed reduction of GLUT4 expression; NIC males also had lower insulin receptor and pAKT expressions. CONCLUSIONS Thus, glycemic homeostasis and peripheral insulin signaling in adult offspring of both sexes are affected by nicotine exposure through the milk, increasing the risk for type 2 diabetes development.
Collapse
Affiliation(s)
- Carla Bruna Pietrobon
- Laboratory of Endocrine Physiology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia Cristina Lisboa
- Laboratory of Endocrine Physiology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Iala Milene Bertasso
- Laboratory of Endocrine Physiology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thamara Cherem Peixoto
- Laboratory of Endocrine Physiology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia Novaes Soares
- Laboratory of Endocrine Physiology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elaine de Oliveira
- Laboratory of Endocrine Physiology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kíssila Rabelo
- Laboratory of Ultrastructure and Tissue Biology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorge José de Carvalho
- Laboratory of Ultrastructure and Tissue Biology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alex Christian Manhães
- Neurophysiology Laboratory, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Egberto Gaspar de Moura
- Laboratory of Endocrine Physiology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Fu X, Zong T, Yang P, Li L, Wang S, Wang Z, Li M, Li X, Zou Y, Zhang Y, Htet Aung LH, Yang Y, Yu T. Nicotine: Regulatory roles and mechanisms in atherosclerosis progression. Food Chem Toxicol 2021; 151:112154. [PMID: 33774093 DOI: 10.1016/j.fct.2021.112154] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/22/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023]
Abstract
Smoking is an independent risk factor for atherosclerosis. The smoke produced by tobacco burning contains more than 7000 chemicals, among which nicotine is closely related to the occurrence and development of atherosclerosis. Nicotine, a selective cholinergic agonist, accelerates the formation of atherosclerosis by stimulating nicotinic acetylcholine receptors (nAChRs) located in neuronal and non-neuronal tissues. This review introduces the pathogenesis of atherosclerosis and the mechanisms involving nicotine and its receptors. Herein, we focus on the various roles of nicotine in atherosclerosis, such as upregulation of growth factors, inflammation, and the dysfunction of endothelial cells, vascular smooth muscle cells (VSMC) as well as macrophages. In addition, nicotine can stimulate the generation of reactive oxygen species, cause abnormal lipid metabolism, and activate immune cells leading to the onset and progression of atherosclerosis. Exosomes, are currently a research hotspot, due to their important connections with macrophages and the VSMC, and may represent a novel application into future preventive treatment to promote the prevention of smoking-related atherosclerosis. In this review, we will elaborate on the regulatory mechanism of nicotine on atherosclerosis, as well as the effects of interference with nicotine receptors and the use of exosomes to prevent atherosclerosis development.
Collapse
Affiliation(s)
- Xiuxiu Fu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Tingyu Zong
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Panyu Yang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Lin Li
- Department of Vascular Surgery, The Qingdao Hiser Medical Center, Qingdao, Shandong Province, China
| | - Shizhong Wang
- The Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 66000, People's Republic of China
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Min Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Yulin Zou
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Ying Zhang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Lynn Htet Htet Aung
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China
| | - Yanyan Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, No. 308 Ningxia Road, Qingdao, 266021, People's Republic of China.
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China; Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China.
| |
Collapse
|
14
|
Peixoto TC, Gaspar de Moura E, Quitete FT, Simino LA, Torsoni AS, Torsoni MA, Manhaes AC, Lisboa PC. Early life nicotine exposure alters mRNA and microRNA expressions related to thyroid function and lipid metabolism in liver and BAT of adult wistar rats. Mol Cell Endocrinol 2021; 523:111141. [PMID: 33359828 DOI: 10.1016/j.mce.2020.111141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 02/06/2023]
Abstract
In rats, maternal nicotine exposure during lactation induces obesity, thyroid dysfunction, brown adipose tissue (BAT) hypofunction and liver alterations in adult offspring. Both thyroid function and lipid metabolism are influenced by gene silencing mediated by microRNAs (miRNAs). Here we investigated long-term effects of early nicotine exposure on molecular and epigenetic mechanisms closely related to thyroid and lipid metabolism, through the expression of mRNAs and miRNAs in BAT and liver of adult male and female offspring. At postnatal day 2 (PND2), lactating control (CON) or nicotine (NIC) dams were subcutaneously implanted with osmotic minipumps containing, respectively, saline or 6 mg/kg nicotine. Litters were adjusted to 3 males and 3 females. Offspring's euthanasia occurred at PND180. In the BAT, NIC females showed higher Dio2 mRNA expression, while miR-382* expression was not altered in both sexes. In the liver, NIC offspring of both sexes showed lower Dio1 mRNA expression and higher miR-224 expression, while only NIC females had higher miR-383 and miR-21 expressions. NIC offspring of both sexes showed higher mRNA expression of SCD1 in the liver; NIC males had decreased CPT1 expression, whereas NIC females had increased FASN, miR-370 and miR-122 expressions. Regardless of sex, alterations in liver Dio1, miR-224 and SCD1 expressions are involved in the disturbances caused by maternal nicotine exposure during breastfeeding. Interestingly, females had more altered miRs in the liver. Early nicotine exposure induces a sex dimorphism, particularly regarding hepatic lipid metabolism, through miRs expression.
Collapse
Affiliation(s)
- Thamara Cherem Peixoto
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, 20551-030, Brazil
| | - Egberto Gaspar de Moura
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, 20551-030, Brazil
| | - Fernanda Torres Quitete
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, 20551-030, Brazil
| | - Laís Angélica Simino
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas, Limeira, SP, 13484-350, Brazil
| | - Adriana Souza Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas, Limeira, SP, 13484-350, Brazil
| | - Marcio Alberto Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas, Limeira, SP, 13484-350, Brazil
| | - Alex Christian Manhaes
- Laboratory of Neurophysiology, Department of Physiological Sciences, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, 20551-030, Brazil
| | - Patricia Cristina Lisboa
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, 20551-030, Brazil.
| |
Collapse
|
15
|
Al-Sawalha NA, Gaugazeh HT, Alzoubi KH, Khabour OF. Maternal waterpipe tobacco smoke exposure during lactation induces hormonal and biochemical changes in rat dams and offspring. Basic Clin Pharmacol Toxicol 2021; 128:315-321. [PMID: 32946675 DOI: 10.1111/bcpt.13493] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/04/2020] [Accepted: 09/11/2020] [Indexed: 01/24/2023]
Abstract
Waterpipe smoking is common among pregnant and breastfeeding women. Herein, the effects of waterpipe tobacco smoke (WTS) exposure during lactation on milk composition, hormonal levels and biochemical profile in dams and pups were investigated. Lactating Wistar rats were randomly assigned to receive either WTS (2 hours per day) or fresh air (control group). Milk was collected on day 21 and analysed for protein, lactose and total fat. Blood, from dams and pups, was analysed for insulin, glucose, lipid profile, leptin, prolactin and corticosterone. WTS exposure during lactation increased the blood level of HDL and corticosterone in dams (P < .05). However, the level of milk lactose and blood glucose was reduced in dams after the exposure to WTS during lactation (P < .05). WTS during lactation significantly increased levels of triglycerides, LDL and leptin (P < .05), and a trend of increase in blood level of nicotine and prolactin in pups. Levels of other parameters were not affected by WTS exposure in dams and pups. In conclusion, WTS exposure during lactation altered the milk composition and altered lipid profile, glucose homeostasis and hormonal levels in dams and pups. It is necessary to adopt strategies to enhance tobacco cessation during breastfeeding.
Collapse
Affiliation(s)
- Nour A Al-Sawalha
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Hasanh T Gaugazeh
- Department of Forensic Medicine and Toxicology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H Alzoubi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Omar F Khabour
- Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
16
|
Peixoto TC, Moura EG, Soares PN, Rodrigues VST, Claudio-Neto S, Oliveira E, Manhães AC, Lisboa PC. Nicotine exposure during lactation causes disruption of hedonic eating behavior and alters dopaminergic system in adult female rats. Appetite 2021; 160:105115. [PMID: 33453337 DOI: 10.1016/j.appet.2021.105115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 11/18/2020] [Accepted: 01/08/2021] [Indexed: 12/30/2022]
Abstract
Tobacco smoke during gestation is associated with increased consumption of palatable foods by the offspring in humans and rats. Postpartum relapse is observed in lactating women who quit smoking during pregnancy, putting their children at risk of adverse health outcomes caused by secondhand smoke. Nicotine is transferred through milk and alters the dopaminergic reward system of adult male rats, reducing dopamine action in the nucleus accumbens (NAc) and hypothalamic arcuate nucleus. Here, we evaluated the long-term effects of nicotine-only exposure during lactation on eating behavior, anxiety, locomotion, dopaminergic system, hypothalamic leptin signaling and nicotinic receptor in the adult female rat progeny. Two days after birth (PN2), Wistar rat dams were separated into control and nicotine (Nic) groups for implantation of osmotic minipumps that released respectively saline or 6 mg/kg nicotine. Lactating dams were kept with 6 pups. After weaning (PN21; nicotine withdrawal), only the female offspring were studied. Euthanasia occurred at PN180. Nic females showed hyperphagia, preference for a high-sucrose diet, increased anxiety-like behavior, lower tyrosine hydroxylase (TH), lower dopamine transporter and higher dopamine receptor (Drd2) in NAc; lower Drd1 in prefrontal cortex and lower TH in dorsal striatum (DS). These animals showed changes that can explain their hyperphagia, such as: lower leptin signaling pathway (Leprb, pJAK2, pSTAT3) and Chrna7 expression in hypothalamus. Neonatal nicotine exposure affects the brain reward system of the female progeny differently from males, mainly decreasing dopamine production in NAc and DS. Therefore, Nic females are more susceptible to develop food addiction and obesity.
Collapse
Affiliation(s)
- T C Peixoto
- Laboratory of Endocrine Physiology, RJ, Brazil
| | - E G Moura
- Laboratory of Endocrine Physiology, RJ, Brazil
| | - P N Soares
- Laboratory of Endocrine Physiology, RJ, Brazil
| | | | - S Claudio-Neto
- Neurophysiology Laboratory, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - E Oliveira
- Laboratory of Endocrine Physiology, RJ, Brazil
| | - A C Manhães
- Neurophysiology Laboratory, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - P C Lisboa
- Laboratory of Endocrine Physiology, RJ, Brazil.
| |
Collapse
|
17
|
Li GL, Ping J, Chen HJ, Zhang WX, Fan J, Peng DS, Zhang L, Yan YE. Maternal nicotine exposure impairs brown adipose tissue via AMPK-SIRT1-PGC-1α signals in male offspring. Life Sci 2021; 264:118695. [PMID: 33130079 DOI: 10.1016/j.lfs.2020.118695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022]
Abstract
AIMS Maternal nicotine exposure during pregnancy and lactation is associated with obesity in offspring. Brown adipose tissue (BAT) is correlated with energy metabolism and obesity. In this study, we explored the mechanism of maternal nicotine exposure on BAT changes in male offspring. MAIN METHODS Pregnant rats were randomly assigned to nicotine (1.0 mg/kg twice per day, subcutaneous administration) or control groups. In vitro, C3H10T1/2 cells were induced to differentiate into mature brown adipocytes, and 0-50 μM nicotine was given to C3H10T1/2 cells during the differentiation process. KEY FINDINGS Nicotine-exposed males had white-like adipocytes and abnormal mitochondria structure in iBAT at 26 weeks. The expression of mitochondrial genes, UCP1 and AMPK-SIRT1-PGC-1α pathway were downregulated in the nicotine group at 26 weeks rather than 4 weeks. In vitro, 50 μM nicotine decreased the expression of mitochondrial genes, UCP1 and AMPK-SIRT1-PGC-1α pathway in brown adipocytes. SIGNIFICANCE Maternal nicotine exposure showed the "programming" effect on the decreased brown-like phenotype in BAT of adult male offspring via downregulating AMPK-SIRT1-PGC-1α pathway. This impairment of BAT may be a potential mechanism of nicotine-induced obesity in male offspring.
Collapse
Affiliation(s)
- Gai-Ling Li
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Jie Ping
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Hui-Jian Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Wan-Xia Zhang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Jie Fan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Dang-Sheng Peng
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Li Zhang
- Demonstration Center for Experimental Basic Medicine Education of Wuhan University, China
| | - You-E Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China.
| |
Collapse
|
18
|
Sex- and age-dependent differences in nicotine susceptibility evoked by developmental exposure to tobacco smoke and/or ethanol in mice. J Dev Orig Health Dis 2020; 12:940-951. [PMID: 33292889 DOI: 10.1017/s2040174420001191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Either tobacco smoking or alcohol consumption during pregnancy sex-selectively increases susceptibility to drugs of abuse later in life. Considering that pregnant smoking women are frequently intermittent consumers of alcoholic beverages, here, we investigated whether a short-term ethanol exposure restricted to the brain growth spurt period when combined with chronic developmental exposure to tobacco smoke aggravates susceptibility to nicotine in adolescent and adult mice. Swiss male and female mice were exposed to tobacco smoke (SMK; research cigarettes 3R4F, whole-body exposure, 8 h/daily) or ambient air during the gestational period and until the tenth postnatal day (PN). Ethanol (ETOH, 2 g/Kg, 25%, i.p.) or saline was injected in the pups every other day from PN2 to PN10. There were no significant differences in cotinine (nicotine metabolite) and ethanol serum levels among SMK, ETOH and SMK + ETOH groups. During adolescence (PN30) and adulthood (PN90), nicotine (NIC, 0.5 mg/Kg) susceptibility was evaluated in the conditioned place preference and open field tests. NIC impact was more evident in females: SMK, ETOH and SMK + ETOH adolescent females were equally more susceptible to nicotine-induced place preference than control animals. At adulthood, SMK and SMK + ETOH adult females exhibited a nicotine-evoked hyperlocomotor profile in the open field, with a stronger effect in the SMK + ETOH group. Our results indicate that ethanol exposure during the brain growth spurt, when combined to developmental exposure to tobacco smoke, increases nicotine susceptibility with stronger effects in adult females. This result represents a worsened outcome from the early developmental dual exposure and may predispose nicotine use/abuse later in life.
Collapse
|
19
|
Miranda RA, de Moura EG, Soares PN, Peixoto TC, Lopes BP, de Andrade CBV, de Oliveira E, Manhães AC, de Faria CC, Fortunato RS, Lisboa PC. Thyroid redox imbalance in adult Wistar rats that were exposed to nicotine during breastfeeding. Sci Rep 2020; 10:15646. [PMID: 32973319 PMCID: PMC7519108 DOI: 10.1038/s41598-020-72725-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 09/04/2020] [Indexed: 12/12/2022] Open
Abstract
Maternal nicotine exposure causes several consequences in offspring phenotype, such as obesity and thyroid dysfunctions. Nicotine exposure can increase oxidative stress levels, which could lead to thyroid dysfunction. However, the mechanism by which nicotine exposure during breastfeeding leads to thyroid gland dysfunction remains elusive. We aimed to investigate the long-term effects of maternal nicotine exposure on redox homeostasis in thyroid gland, besides other essential steps for thyroid hormone synthesis in rats from both sexes. Lactating Wistar rats were implanted with osmotic minipumps releasing nicotine (NIC, 6 mg/kg/day) or saline (control) from postnatal day 2 to 16. Offspring were analyzed at 180-day-old. NIC males showed lower plasma TSH, T3 and T4 while NIC females had higher T3 and T4. In thyroid, NIC males had higher sodium-iodide symporter protein expression, whereas NIC females had higher thyroid-stimulating hormone receptor (TSHr) and thyroperoxidase (TPO) protein expression. TPO activity was lower in NIC males. Hydrogen peroxide generation was decreased in NIC males. Activities of superoxide dismutase, catalase and glutathione peroxidase were compromised in NIC animals from both sexes. 4-Hydroxynonenal was higher only in NIC females, while thiol was not affected in NIC animals from both sexes. NIC offspring also had altered expression of sex steroid receptors in thyroid gland. Both sexes showed similar thyroid morphology, with lower follicle and colloid size. Thyroid from female offspring exposed to nicotine during breastfeeding developed oxidative stress, while the male gland seemed to be protected from redox damage. Thyroid dysfunctions seem to be associated with redox imbalance in a sex-dependent manner.
Collapse
Affiliation(s)
- Rosiane Aparecida Miranda
- Physiological Sciences Department, Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Avenida 28 de Setembro, 87, Rio de Janeiro, RJ, 20551-031, Brazil
| | - Egberto Gaspar de Moura
- Physiological Sciences Department, Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Avenida 28 de Setembro, 87, Rio de Janeiro, RJ, 20551-031, Brazil
| | - Patrícia Novaes Soares
- Physiological Sciences Department, Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Avenida 28 de Setembro, 87, Rio de Janeiro, RJ, 20551-031, Brazil
| | - Thamara Cherem Peixoto
- Physiological Sciences Department, Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Avenida 28 de Setembro, 87, Rio de Janeiro, RJ, 20551-031, Brazil
| | - Bruna Pereira Lopes
- Physiological Sciences Department, Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Avenida 28 de Setembro, 87, Rio de Janeiro, RJ, 20551-031, Brazil
| | - Cherley Borba Vieira de Andrade
- Translational Endocrinology Laboratory, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elaine de Oliveira
- Physiological Sciences Department, Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Avenida 28 de Setembro, 87, Rio de Janeiro, RJ, 20551-031, Brazil
| | - Alex C Manhães
- Laboratory of Neurophysiology, Biology Institute, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Caroline Coelho de Faria
- Laboratory of Molecular Radiobiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo Soares Fortunato
- Laboratory of Molecular Radiobiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Cristina Lisboa
- Physiological Sciences Department, Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Avenida 28 de Setembro, 87, Rio de Janeiro, RJ, 20551-031, Brazil.
| |
Collapse
|
20
|
Miranda RA, Gaspar de Moura E, Lisboa PC. Tobacco smoking during breastfeeding increases the risk of developing metabolic syndrome in adulthood: Lessons from experimental models. Food Chem Toxicol 2020; 144:111623. [PMID: 32738371 DOI: 10.1016/j.fct.2020.111623] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/01/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022]
Abstract
Metabolic syndrome (MetS) is characterized by increased abdominal fat, dyslipidemia, diabetes mellitus and hypertension. A high MetS prevalence is strongly associated with obesity. Obesity is a public health problem in which several complex factors have been implicated, including environmental pollutants. For instance, maternal smoking seems to play a role in obesogenesis in childhood. Given the association between endocrine disruptors, obesity and metabolic programming, over the past 10 years, our research group has contributed to studies based on the hypothesis that early exposure to nicotine/tobacco causes offspring to become MetS-prone. The mechanism by which tobacco smoking during breastfeeding induces metabolic dysfunctions is not completely understood; however, increased metabolic programming has been shown in studies that focus on this topic. Here, we reviewed the literature mainly based in light of our latest data from experimental models. Nicotine or tobacco exposure during breastfeeding induces several endocrine dysfunctions in a sex- and tissue-specific manner. This review provides an updated summary regarding the hypothesis that early exposure to nicotine/tobacco causes offspring to become MetS-prone. An understanding of this issue can provide support to prevent long-term disorders, mainly related to the risk of obesity and its comorbidities, in future generations.
Collapse
Affiliation(s)
- Rosiane A Miranda
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Egberto Gaspar de Moura
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Patrícia Cristina Lisboa
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
21
|
Abstract
Abstract
Lactation is a critical period during which maternal nutritional and environmental challenges affect milk composition and, therefore, organ differentiation, structure, and function in offspring during the early postnatal period. Evidence to date shows that lactation is a vulnerable time during which transient insults can have lasting effects, resulting in altered health outcomes in offspring in adult life. Despite the importance of the developmental programming that occurs during this plastic period of neonatal life, there are few comprehensive reviews of the multiple challenges—especially to the dam—during lactation. This review presents milk data from rodent studies involving maternal nutritional challenges and offspring outcome data from studies involving maternal manipulations during lactation. Among the topics addressed are maternal nutritional challenges and the effects of litter size and artificial rearing on offspring metabolism and neural and endocrine outcomes. The lactation period is an opportunity to correct certain functional deficits resulting from prenatal challenges to the fetus, but, if not personalized, can also lead to undesirable outcomes related to catch up-growth and overnutrition.
Collapse
|
22
|
Nicotine exposure during breastfeeding reduces sympathetic activity in brown adipose tissue and increases in white adipose tissue in adult rats: Sex-related differences. Food Chem Toxicol 2020; 140:111328. [DOI: 10.1016/j.fct.2020.111328] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/13/2020] [Accepted: 04/06/2020] [Indexed: 12/13/2022]
|
23
|
Hibara A, Yamaguchi T, Kojima M, Yamano Y, Higuchi M. Nicotine inhibits expression of Prrx1 in pituitary stem/progenitor cells through epigenetic regulation, leading to a delayed supply of growth-hormone-producing cells. Growth Horm IGF Res 2020; 51:65-74. [PMID: 32146343 DOI: 10.1016/j.ghir.2020.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 12/27/2019] [Accepted: 02/17/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Nicotine, a toxic component of smoking, adversely affects animal growth and reproduction by decreasing secretion of anterior pituitary hormones. However, it has not been clarified whether nicotine inhibits the supply of endocrine cells in the pituitary gland. The present study investigated short- and long-term effects of persistent nicotine exposure on the pituitary glands of young animals. DESIGN Three-week-old male Wistar rats were exposed to nicotine (1 mg/kg body weight/day) for 7 days, and gene expression, cell numbers, and DNA methylation status were analyzed on the following day and 4 weeks after final treatments. RESULTS The expression level of the stem cell marker Sox2 was not changed by nicotine exposure throughout the experiment. On the other hand, nicotine inhibited expression of a progenitor cell marker, Prrx1, and growth hormone (Gh). Immunohistochemical analysis showed that the SOX2-positive cells positive for PRRX1 in nicotine-treated groups decreased to 61% (4-week-old) and 70% (8-week-old) of the saline-treated controls. In addition, the proportion of GH-positive cells in nicotine-treated group was 14% lower than that of saline-treated controls. Furthermore, first intron hypermethylation of Prrx1 was detected by a bisulfite sequence of genomic DNA from the anterior lobe of the rat pituitary gland. CONCLUSIONS We show that persistent nicotine exposure in young animals inhibits expression of Prrx1 in pituitary stem/progenitor cells through epigenetic regulation, leading to a delayed supply of GH-producing cells.
Collapse
Affiliation(s)
- Ayaka Hibara
- Laboratory of Veterinary Biochemistry, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101 Koyama-minami, Tottori-shi, Tottori 680-8553, Japan
| | - Takahiro Yamaguchi
- Laboratory of Veterinary Biochemistry, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101 Koyama-minami, Tottori-shi, Tottori 680-8553, Japan
| | - Miki Kojima
- Laboratory of Veterinary Biochemistry, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101 Koyama-minami, Tottori-shi, Tottori 680-8553, Japan
| | - Yoshiaki Yamano
- Laboratory of Veterinary Biochemistry, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101 Koyama-minami, Tottori-shi, Tottori 680-8553, Japan
| | - Masashi Higuchi
- Laboratory of Veterinary Biochemistry, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101 Koyama-minami, Tottori-shi, Tottori 680-8553, Japan.
| |
Collapse
|
24
|
Bertasso IM, Pietrobon CB, Lopes BP, Peixoto TC, Soares PN, Oliveira E, Manhães AC, Bonfleur ML, Balbo SL, Cabral SS, Gabriel Kluck GE, Atella GC, Gaspar de Moura E, Lisboa PC. Programming of hepatic lipid metabolism in a rat model of postnatal nicotine exposure - Sex-related differences. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 258:113781. [PMID: 31864076 DOI: 10.1016/j.envpol.2019.113781] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/04/2019] [Accepted: 12/08/2019] [Indexed: 06/10/2023]
Abstract
Maternal nicotine exposure during lactation induces liver damage in adult male rats. However, the mechanism in males is unknown and females have not been tested. Here, we determined the liver lipid composition and lipogenic enzymes in male and female offspring at two ages in a model of postnatal nicotine exposure. Osmotic minipumps were implanted in lactating Wistar rat dams at postnatal day (PND) 2 to release 6 mg/kg/day of nicotine (NIC group) or saline (CON group) for 14 days. Offspring received a standard diet from weaning until euthanasia at PND120 (1 pup/litter/sex) or PND180 (2 pups/litter/sex). At PND120, NIC males showed lower plasma triglycerides (TG), steatosis degree 1, higher hepatic cholesterol (CHOL) ester, free fatty acids, monoacylglycerol content as well as acetyl-coa carboxylase-1 (ACC-1) and fatty acid synthase (FAS) protein expression in the liver compared to CON males. At this age, NIC females had preserved hepatocytes architecture, higher plasma CHOL, higher CHOL ester and lower total CHOL content in the liver compared to CON females. At PND180, NIC males showed steatosis degrees 1 and 2, higher TG, lower free fatty acids and total CHOL content in the liver and an increase in ACC-1 hepatic protein expression. NIC females had higher plasma TG and CHOL levels, no change in hepatic morphology, lower CHOL ester and free fatty acids in the liver, which also showed higher total ACC-1 and FAS protein expression. Maternal nicotine exposure induces long-term liver dysfunction, with an alteration in hepatic cytoarchitecture that was aggravated with age in males. Concerning females, despite unchanged hepatic cytoarchitecture, lipid metabolism was compromised, which deserves further attention.
Collapse
Affiliation(s)
- Iala Milene Bertasso
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - Carla Bruna Pietrobon
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - Bruna Pereira Lopes
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - Thamara Cherem Peixoto
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - Patrícia Novaes Soares
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - Elaine Oliveira
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - Alex Christian Manhães
- Laboratory of Neurophysiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - Maria Lucia Bonfleur
- Laboratory of Endocrine Physiology and Metabolism, Center of Biological and Health Sciences, Western Paraná State University, Cascavel, PR, Brazil
| | - Sandra Lucinei Balbo
- Laboratory of Endocrine Physiology and Metabolism, Center of Biological and Health Sciences, Western Paraná State University, Cascavel, PR, Brazil
| | - Suellen Silva Cabral
- Laboratory of Lipids and Lipoprotein Biochemistry, Biochemistry Institute of Federal University of Rio de Janeiro, RJ, Brazil
| | - George Eduardo Gabriel Kluck
- Laboratory of Lipids and Lipoprotein Biochemistry, Biochemistry Institute of Federal University of Rio de Janeiro, RJ, Brazil
| | - Georgia Correa Atella
- Laboratory of Lipids and Lipoprotein Biochemistry, Biochemistry Institute of Federal University of Rio de Janeiro, RJ, Brazil
| | - Egberto Gaspar de Moura
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - Patrícia Cristina Lisboa
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
25
|
Peixoto TC, Moura EG, Oliveira E, Younes-Rapozo V, Soares PN, Rodrigues VST, Torsoni MA, Torsoni AS, Manhães AC, Lisboa PC. Hypothalamic Neuropeptides Expression and Hypothalamic Inflammation in Adult Rats that Were Exposed to Tobacco Smoke during Breastfeeding: Sex-Related Differences. Neuroscience 2019; 418:69-81. [PMID: 31487543 DOI: 10.1016/j.neuroscience.2019.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 02/06/2023]
Abstract
The hypothalamus controls food intake and energy expenditure. In rats, maternal exposure to nicotine during breastfeeding alters the hypothalamic circuitry of the adult offspring, resulting in leptin resistance, neuropeptides changes and gliosis. Tobacco smoke exposure during lactation causes greater adiposity, hyperphagia and hyperleptinemia in the adult progeny. To understand the central mechanisms underlying the obese phenotype of adult rats that were directly and indirectly exposed to cigarette smoke during lactation, we investigated leptin signaling, orexigenic and anorexigenic neuropeptides expression, as well as astrocyte and microglia markers in hypothalamus. From postnatal day (PND) 3 to 21, Wistar lactating rat dams and their pups were divided into two groups: SE, smoke-exposed in a cigarette-smoking machine (four times/day); Crtl, exposed to filtered air. Offspring of both sexes were euthanized at PND180. The leptin pathway was not altered in SE animals from both sexes. SE males showed increased NPY (arcuate nucleus, ARC), CRH (paraventricular nucleus, PVN), as well as higher GFAP fiber density (ARC and PVN) and IL6 protein content. TRH (PVN) immunohistochemistry was reduced. SE females had lower CART-positive cells (ARC) and lower α-MSH immunostaining intensity (PVN and lateral hypothalamus), with no change of GFAP or IL-6. The protein contents of CX3CR1 (marker of activated microglia) and α7nAChR (anti-inflammatory marker) were not altered in both SE males and females. Neonatal cigarette smoke is deleterious to the hypothalamic circuitry, inducing changes in energy homeostasis favoring hyperphagia and decreased energy expenditure at adulthood in both sexes; however sex-dependent mechanisms were observed.
Collapse
Affiliation(s)
- T C Peixoto
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - E G Moura
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - E Oliveira
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - V Younes-Rapozo
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - P N Soares
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - V S T Rodrigues
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - M A Torsoni
- Faculdade de Ciências Aplicadas, Universidade Estadual de Campinas, Limeira, Brazil
| | - A S Torsoni
- Faculdade de Ciências Aplicadas, Universidade Estadual de Campinas, Limeira, Brazil
| | - A C Manhães
- Laboratório de Neurofisiologia, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - P C Lisboa
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
26
|
Zhang WX, Chen HJ, Fan J, Li GL, Sun A, Lan LY, Zhang L, Yan YE. The association between maternal nicotine exposure and adipose angiogenesis in female rat offspring: A mechanism of adipose tissue function changes. Toxicol Lett 2019; 318:12-21. [PMID: 31622651 DOI: 10.1016/j.toxlet.2019.10.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/04/2019] [Accepted: 10/12/2019] [Indexed: 02/08/2023]
Abstract
Maternal smoking during pregnancy and lactation is associated with increased fat mass in the offspring, but the mechanism by which this occurs is not fully understood. Our study focused on the relationships among maternal nicotine exposure, adipose angiogenesis and adipose tissue function in female offspring. Pregnant rats were randomly assigned to nicotine or control groups. Microvascular density, lipid metabolism and α7nAChR-Egr1-FGF2 signaling pathway genes/proteins were tested in 4-, 12- and 26-week female offspring. In vitro, nicotine concentration- and time-response experiments were conducted in 3T3-L1. Lipid metabolism and α7nAChR-Egr1-FGF2 signaling pathway genes/proteins were tested. The conditioned media of differentiated 3T3-L1 treated with nicotine were used to observe tube formation in human umbilical vein endothelial cells (HUVECs). Nicotine-exposed females presented higher adipose microvascular density. The gene expression of α7nAChR, Egr1 and FGF2 was significantly increased in gonadal white adipose tissue (gWAT) and inguinal subcutaneous WAT (igSWAT) of nicotine-exposed females at 4 weeks of age. The protein expression of α7nAChR, Egr1 and FGF2 was increased in gWAT and igSWAT of nicotine-exposed females at 4 weeks of age, and increased in gWAT at 26 weeks. In vitro, nicotine increased the expression of lipid metabolism and α7nAChR-Egr1-FGF2 signaling pathway genes/proteins in a concentration- and time-dependent manner. In the tube formation experiment, adipocytes affected by nicotine promoted HUVEC angiogenesis. Therefore, maternal nicotine exposure promoted the early angiogenesis of adipose tissue via the α7nAChR-Egr1-FGF2 signaling pathway, and this angiogenesis mechanism was associated with increased adipogenesis in adipose tissue of female offspring.
Collapse
Affiliation(s)
- Wan-Xia Zhang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Hui-Jian Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Jie Fan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Gai-Ling Li
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Ao Sun
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Liu-Yi Lan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Li Zhang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - You-E Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.
| |
Collapse
|
27
|
Reduced adolescent risk-assessment and lower nicotinic beta-2 expression in rats exposed to nicotine through lactation by forcedly drinking dams. Neuroscience 2019; 413:64-76. [DOI: 10.1016/j.neuroscience.2019.06.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 01/06/2023]
|
28
|
Gibson L, Porter M. Drinking or Smoking While Breastfeeding and Later Cognition in Children. Pediatrics 2018; 142:peds.2017-4266. [PMID: 30061301 DOI: 10.1542/peds.2017-4266] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/09/2018] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Although prenatal alcohol and nicotine exposure are associated with reduced cognition in children, associations between consumption of alcohol during lactation and cognition have not been examined. We aimed to examine whether drinking or smoking while breastfeeding lowers children's cognitive scores. We hypothesized that increased drinking or smoking would be associated with dose-dependent cognitive reductions. METHODS Data were sourced from Growing Up in Australia: The Longitudinal Study of Australian Children. Participants were 5107 Australian infants recruited in 2004 and assessed every 2 years. Multivariable linear regression analyses assessed relationships between drinking and smoking habits of breastfeeding mothers and children's Matrix Reasoning, Peabody Picture Vocabulary Test-Third Edition and Who Am I? scores at later waves. RESULTS Increased or riskier wave 1 maternal alcohol consumption was associated with reductions in Matrix Reasoning scores at age 6 to 7 years in children who had been breastfed (B = -0.11; SE = 0.03; 95% confidence interval: -0.18 to -0.04; P = .01). This relationship was not evident in infants who had never breastfed (B = -0.02; SE = 0.10; 95% confidence interval = -0.20 to 0.17; P = .87). Smoking during lactation was not associated with any outcome variable. CONCLUSIONS Exposing infants to alcohol through breastmilk may cause dose-dependent reductions in their cognitive abilities. This reduction was observed at age 6 to 7 years but was not sustained at age 10 to 11 years. Although the relationship is small, it may be clinically significant when mothers consume alcohol regularly or binge drink. Further analyses will assess relationships between alcohol consumption or tobacco smoking during lactation and academic, developmental, physical, and behavioral outcomes in children.
Collapse
Affiliation(s)
- Louisa Gibson
- Department of Psychology, Faculty of Human Sciences, Macquarie University, Sydney, Australia
| | - Melanie Porter
- Department of Psychology, Faculty of Human Sciences, Macquarie University, Sydney, Australia
| |
Collapse
|
29
|
Gong Y, Zhang H, Geng N, Xing L, Fan J, Luo Y, Song X, Ren X, Wang F, Chen J. Short-chain chlorinated paraffins (SCCPs) induced thyroid disruption by enhancement of hepatic thyroid hormone influx and degradation in male Sprague Dawley rats. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 625:657-666. [PMID: 29304504 DOI: 10.1016/j.scitotenv.2017.12.251] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 05/22/2023]
Abstract
Short-chain chlorinated paraffins (SCCPs) are known to disturb thyroid hormone (TH) homeostasis in rodents. However, the mechanism remains to be fully characterized. In this study, male Sprague Dawley rats received SCCPs (0, 1, 10, or 100mg/kg/day) via gavage once a day for consecutive 28days. Plasma and hepatic TH concentrations, thyrocyte structure, as well as thyroid and hepatic mRNA and protein levels of genes associated with TH homeostasis were examined. Moreover, we performed molecular docking to predict interactions between constitutive androstane receptor (CAR), a key regulator in xenobiotic-induced TH metabolism, with different SCCP molecules. Exposure to SCCPs significantly decreased the circulating free thyroxine (T4) and triiodothyronine (T3) levels, but increased thyroid-stimulating hormone (TSH) levels by a feedback mechanism. Decreased hepatic T4 and increased hepatic T3 levels were also seen after 100mg/kg/day SCCPs exposure. SCCPs didn't show any significant effects on the expression of thyroid TH synthesis genes or thyrocyte structure. However, stimulation effects were observed for mRNA and protein levels of hepatic uridine diphosphoglucuronosyl transferase (UGT) 1A1 and organic anion transporter 2, suggesting an accelerated TH metabolism in rat liver. The increased cytochrome P450 2B1 but not 1A1 mRNA and protein levels indicated that the CAR signaling was activated by SCCPs exposure. According to docking analysis, SCCPs form hydrophobic interactions with CAR and the binding affinity shows dependency on chlorine content. Overall, our data showed that CAR implicated enhancement of hepatic TH influx and degradation could be the main cause for SCCPs induced TH deficiency in male rats.
Collapse
Affiliation(s)
- Yufeng Gong
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Haijun Zhang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Ningbo Geng
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Liguo Xing
- Safety Evaluation Center of Shenyang Research Institute of Chemical Industry Ltd., Shenyang 110021, China
| | - Jingfeng Fan
- National Marine Environmental Monitoring Center, Dalian 116023, China
| | - Yun Luo
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyao Song
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Xiaoqian Ren
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feidi Wang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiping Chen
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| |
Collapse
|
30
|
Peixoto TC, Moura EG, Oliveira E, Younes-Rapozo V, Soares PN, Rodrigues VST, Santos TR, Peixoto-Silva N, Carvalho JC, Calvino C, Conceição EPS, Guarda DS, Claudio-Neto S, Manhães AC, Lisboa PC. Neonatal tobacco smoke reduces thermogenesis capacity in brown adipose tissue in adult rats. ACTA ACUST UNITED AC 2018; 51:e6982. [PMID: 29694503 PMCID: PMC5937726 DOI: 10.1590/1414-431x20186982] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 01/15/2018] [Indexed: 12/30/2022]
Abstract
Maternal smoking is a risk factor for progeny obesity. We have previously shown, in a rat model of neonatal tobacco smoke exposure, a mild increase in food intake and a considerable increase in visceral adiposity in the adult offspring. Males also had secondary hyperthyroidism, while females had only higher T4. Since brown adipose tissue (BAT) hypofunction is related to obesity, here we tested the hypothesis that higher levels of thyroid hormones are not functional in BAT, suggesting a lower metabolic rate. We evaluated autonomic nerve activity in BAT and its function in adult rats that were exposed to tobacco smoke during lactation. At birth, litters were adjusted to 3 male and 3 female pups/litter. From postnatal day (PND) 3 to 21, Wistar lactating rats and their pups were divided into SE group, smoke-exposed in a cigarette smoking machine (4 times/day) and C group, exposed to filtered air. Offspring were sacrificed at PND180. Adult SE rats of both genders had lower interscapular BAT autonomic nervous system activity, with higher BAT mass but no change in morphology. BAT UCP1 and CPT1a protein levels were decreased in the SE groups of both genders. Male SE rats had lower β3-AR, TRα1, and TRβ1 expression while females showed lower PGC1α expression. BAT Dio2 mRNA and hypothalamic POMC and MC4R levels were similar between groups. Hypothalamic pAMPK level was higher in SE males and lower in SE females. Thus, neonatal cigarette smoke exposure induces lower BAT thermogenic capacity, which can be obesogenic at adulthood.
Collapse
Affiliation(s)
- T C Peixoto
- Laboratório de Fisiologia Endócrina, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - E G Moura
- Laboratório de Fisiologia Endócrina, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - E Oliveira
- Laboratório de Fisiologia Endócrina, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - V Younes-Rapozo
- Laboratório de Fisiologia Endócrina, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - P N Soares
- Laboratório de Fisiologia Endócrina, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - V S T Rodrigues
- Laboratório de Fisiologia Endócrina, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - T R Santos
- Laboratório de Fisiologia Endócrina, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - N Peixoto-Silva
- Laboratório de Fisiologia Endócrina, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - J C Carvalho
- Laboratório de Fisiologia Endócrina, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - C Calvino
- Laboratório de Fisiologia Endócrina, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - E P S Conceição
- Laboratório de Fisiologia Endócrina, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - D S Guarda
- Laboratório de Fisiologia Endócrina, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - S Claudio-Neto
- Laboratório de Neurofisiologia, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - A C Manhães
- Laboratório de Neurofisiologia, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - P C Lisboa
- Laboratório de Fisiologia Endócrina, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
31
|
Albers L, Sobotzki C, Kuß O, Ajslev T, Batista RF, Bettiol H, Brabin B, Buka SL, Cardoso VC, Clifton VL, Devereux G, Gilman SE, Grzeskowiak LE, Heinrich J, Hummel S, Jacobsen GW, Jones G, Koshy G, Morgen CS, Oken E, Paus T, Pausova Z, Rifas-Shiman SL, Sharma AJ, da Silva AA, Sørensen TI, Thiering E, Turner S, Vik T, von Kries R. Maternal smoking during pregnancy and offspring overweight: is there a dose-response relationship? An individual patient data meta-analysis. Int J Obes (Lond) 2018; 42:1249-1264. [PMID: 29717267 DOI: 10.1038/s41366-018-0050-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 11/13/2017] [Accepted: 12/27/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND/OBJECTIVES A number of meta-analyses suggest an association between any maternal smoking in pregnancy and offspring overweight obesity. Whether there is a dose-response relationship across number of cigarettes and whether this differs by sex remains unclear. SUBJECT/METHODS Studies reporting number of cigarettes smoked during pregnancy and offspring BMI published up to May 2015 were searched. An individual patient data meta-analysis of association between the number of cigarettes smoked during pregnancy and offspring overweight (defined according to the International Obesity Task Force reference) was computed using a generalized additive mixed model with non-linear effects and adjustment for confounders (maternal weight status, breastfeeding, and maternal education) and stratification for sex. RESULTS Of 26 identified studies, 16 authors provided data on a total of 238,340 mother-child-pairs. A linear positive association was observed between the number of cigarettes smoked and offspring overweight for up to 15 cigarettes per day with an OR increase per cigarette of 1.03, 95% CI = [1.02-1.03]. The OR flattened with higher cigarette use. Associations were similar in males and females. Sensitivity analyses supported these results. CONCLUSIONS A linear dose-response relationship of maternal smoking was observed in the range of 1-15 cigarettes per day equally in boys and girls with no further risk increase for doses above 15 cigarettes.
Collapse
Affiliation(s)
- Lucia Albers
- Division of Epidemiology, Institute of Social Paediatrics and Adolescents Medicine, Ludwig-Maximilians-University Munich, Munich, Germany.
| | - Christina Sobotzki
- Division of Epidemiology, Institute of Social Paediatrics and Adolescents Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Oliver Kuß
- German Diabetes Center, Institute of Biometrics and Epidemiology, Düsseldorf, 40225, Germany
| | - Teresa Ajslev
- Department of Clinical Epidemiology (formerly Institute of Preventive Medicine), Bispebjerg and Frederiksberg Hospitals, The Capital Region, Denmark
| | - Rosangela Fl Batista
- Departamento de Saúde Pública, Universidade Federal do Maranhão, São Luís, MA, Brazil
| | - Heloisa Bettiol
- Departamento de Puericultura e Pediatria, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Bernard Brabin
- Child and Reproductive Health Group, Liverpool School of Tropical Medicine, Liverpool, UK.,Department of Community Child Health,Royal Liverpool Children's Hospital, NHS Trust Alder Hey, Liverpool, UK.,Emma Kinderziekenhuis, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Stephen L Buka
- Department of Epidemiology, Brown University School of Public Health, Providence, Rhode Island, USA
| | - Viviane C Cardoso
- Departamento de Puericultura e Pediatria, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Vicki L Clifton
- Adelaide Medical School, The Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
| | | | - Stephen E Gilman
- Health Behavior Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA.,Department of Social and Behavioral Sciences, Harvard TH Chan School of Public Health, Boston, MA, USA.,Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA.,Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Luke E Grzeskowiak
- Adelaide Medical School, The Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
| | - Joachim Heinrich
- Institute of Occupational, Social, and Environmental Medicine, University Hospital, Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Occupational, Social, and Environmental Medicine, University Hospital, Neuherberg, Germany
| | - Sandra Hummel
- Forschergruppe Diabetes der Technischen Universität München, Munich, Germany.,Institut für Diabetesforschung der Forschergruppe Diabetes e.V. am Helmholtz Zentrum München, Munich, Germany
| | - Geir W Jacobsen
- Department of Public Health and General Practice, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Graeme Jones
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Gibby Koshy
- Child and Reproductive Health Group, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Camilla Schmidt Morgen
- Department of Clinical Epidemiology (formerly Institute of Preventive Medicine), Bispebjerg and Frederiksberg Hospitals, The Capital Region, Denmark
| | - Emily Oken
- Obesity Prevention Program, Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Tomas Paus
- Rotman Research Institute and Departments of Psychology and Psychiatry, University of Toronto, Toronto, Canada
| | - Zdenka Pausova
- Hospital for Sick Children and Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Canada
| | - Sheryl L Rifas-Shiman
- Obesity Prevention Program, Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | | | - Antônio Am da Silva
- Departamento de Saúde Pública, Universidade Federal do Maranhão, São Luís, MA, Brazil
| | - Thorkild Ia Sørensen
- Department of Clinical Epidemiology (formerly Institute of Preventive Medicine), Bispebjerg and Frederiksberg Hospitals, The Capital Region, Denmark.,Novo Nordisk Foundation Centre for Basic Metabolic Research, and Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Elisabeth Thiering
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Epidemiology I, Neuherberg, Germany
| | | | - Torstein Vik
- Department of Laboratory Medicine, Children and Women's Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rüdiger von Kries
- Division of Epidemiology, Institute of Social Paediatrics and Adolescents Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
32
|
Zhang WX, Li YP, Fan J, Chen HJ, Li GL, Ouyang YQ, Yan YE. Perinatal nicotine exposure increases obesity susceptibility by peripheral leptin resistance in adult female rat offspring. Toxicol Lett 2018; 283:91-99. [DOI: 10.1016/j.toxlet.2017.11.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/09/2017] [Accepted: 11/13/2017] [Indexed: 12/11/2022]
|
33
|
Lisboa PC, Soares PN, Peixoto TC, Carvalho JC, Calvino C, Rodrigues VST, Bernardino DN, Younes-Rapozo V, Manhães AC, de Oliveira E, de Moura EG. Effects of cigarette smoke exposure during suckling on food intake, fat mass, hormones, and biochemical profile of young and adult female rats. Endocrine 2017; 57:60-71. [PMID: 28527122 DOI: 10.1007/s12020-017-1320-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 05/01/2017] [Indexed: 01/31/2023]
Abstract
PURPOSE Children from smoking mothers have a higher risk of developing obesity and associated comorbidities later in life. Different experimental models have been used to assess the mechanisms involved with this increased risk. Using a rat model of neonatal nicotine exposure via implantation of osmotic minipumps in lactating dams, we have previously shown marked sexual dimorphisms regarding metabolic and endocrine outcomes in the adult progeny. Considering that more than four thousand substances are found in tobacco smoke besides nicotine, we then studied a rat model of neonatal tobacco smoke exposure: adult male offspring had hyperphagia, obesity, hyperglycemia, hypertriglyceridemia, secondary hyperthyroidism and lower adrenal hormones. Since litters were culled to include only males and since sexual dimorphisms had already been identified in the nicotine exposure model, here we also evaluated the effects of tobacco smoke exposure during lactation on females. METHODS Wistar rat dams and their pups were separated into two groups of 8 litters each: SMOKE (4 cigarettes per day, from postnatal day 3 to 21) and CONTROL (filtered air). Offspring of both sexes were euthanized at PN21 and PN180. RESULTS Changes in male offspring corroborated previous data. At weaning, females showed lower body mass gain and serum triglycerides, but no alterations in visceral fat and hormones. At adulthood, females had higher body mass, hyperphagia, central obesity, hyperleptinemia, hypercholesterolemia, hypercorticosteronemia, but no change in serum TSH and T3, and adrenal catecholamine CONCLUSIONS: Sexual dimorphisms were observed in several parameters, thus indicating that metabolic and hormonal changes due to smoke exposure during development are sex-dependent.
Collapse
Affiliation(s)
- Patricia Cristina Lisboa
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Patricia Novaes Soares
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Thamara Cherem Peixoto
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Janaine Cavalcanti Carvalho
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Camila Calvino
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Dayse Nascimento Bernardino
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Viviane Younes-Rapozo
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Alex Christian Manhães
- Laboratory of Neurophysiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Elaine de Oliveira
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Egberto Gaspar de Moura
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
34
|
Jones MR, Magid HS, Al-Rifai M, McEvoy JW, Kaufman JD, Hinckley Stukovsky KD, Szklo M, Polak J, Burke GL, Post WS, Blaha MJ, Navas-Acien A. Secondhand Smoke Exposure and Subclinical Cardiovascular Disease: The Multi-Ethnic Study of Atherosclerosis. J Am Heart Assoc 2016; 5:e002965. [PMID: 27993830 PMCID: PMC5210438 DOI: 10.1161/jaha.115.002965] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 10/14/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND Few studies have evaluated the association between secondhand smoke (SHS) and subclinical cardiovascular disease among ethnically diverse populations. This study assesses the impact of SHS on inflammation and atherosclerosis (carotid intima-media thickness, coronary artery calcification, and peripheral arterial disease). METHODS AND RESULTS We examined 5032 nonsmoking adults aged 45 to 84 years without prior cardiovascular disease participating in the Multi-Ethnic Study of Atherosclerosis (MESA) from 2000 to 2002. SHS exposure was determined by self-report, and urinary cotinine was measured in a representative subset (n=2893). The multi-adjusted geometric mean ratios (95% CIs) for high-sensitivity C-reactive protein and interleukin-6 comparing 407 participants with SHS ≥12 h/wk versus 3035 unexposed participants were 1.13 (1.02-1.26) and 1.04 (0.98-1.11), respectively. The multi-adjusted geometric mean ratio for carotid intima-media thickness was 1.02 (0.97-1.07). Fibrinogen and coronary artery calcification were not associated with SHS. The prevalence of peripheral arterial disease (ankle-brachial index ≤0.9 or ≥1.4) was associated with detectable urinary cotinine (odds ratio, 2.10; 95% CI, 1.09-4.04) but not with self-reported SHS. Urinary cotinine was not associated with inflammation or carotid intima-media thickness. CONCLUSIONS Despite limited exposure assessment, this study supports the association of SHS exposure with inflammation and peripheral arterial disease.
Collapse
Affiliation(s)
- Miranda R Jones
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Hoda S Magid
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Mahmoud Al-Rifai
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Ciccarone Center for the Prevention of Heart Disease, Johns Hopkins University, Baltimore, MD
| | - John W McEvoy
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Ciccarone Center for the Prevention of Heart Disease, Johns Hopkins University, Baltimore, MD
| | - Joel D Kaufman
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA
| | - Karen D Hinckley Stukovsky
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA
| | - Moyses Szklo
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Joseph Polak
- Department of Radiology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA
| | - Gregory L Burke
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston- Salem, NC
| | - Wendy S Post
- Division of Cardiology, Johns Hopkins University, Baltimore, MD
| | - Michael J Blaha
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Ciccarone Center for the Prevention of Heart Disease, Johns Hopkins University, Baltimore, MD
| | - Ana Navas-Acien
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
35
|
Epigenomic and metabolic responses of hypothalamic POMC neurons to gestational nicotine exposure in adult offspring. Genome Med 2016; 8:93. [PMID: 27609221 PMCID: PMC5015242 DOI: 10.1186/s13073-016-0348-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 08/23/2016] [Indexed: 01/06/2023] Open
Abstract
Background Epidemiological and animal studies have reported that prenatal nicotine exposure (PNE) leads to obesity and type-2 diabetes in offspring. Central leptin-melanocortin signaling via hypothalamic arcuate proopiomelanocortin (POMC) neurons is crucial for the regulation of energy and glucose balance. Furthermore, hypothalamic POMC neurons were recently found to mediate the anorectic effects of nicotine through activation of acetylcholine receptors. Here, we hypothesized that PNE impairs leptin-melanocortinergic regulation of energy balance in first-generation offspring by altering expression of long non-coding RNAs (lncRNAs) putatively regulating development and/or function of hypothalamic POMC neurons. Methods C57BL/6J females were exposed ad libitum to nicotine through drinking water and crossed with C57BL/6J males. Nicotine exposure was sustained during pregnancy and discontinued at parturition. Offspring development was monitored from birth into adulthood. From the age of 8 weeks, central leptin-melanocortin signaling, diabetes, and obesity susceptibility were assessed in male offspring fed a low-fat or high-fat diet for 16 weeks. Nicotine-exposed and non-exposed C57BL/6J females were also crossed with C57BL/6J males expressing the enhanced green fluorescent protein specifically in POMC neurons. Transgenic male offspring were subjected to laser microdissections and RNA sequencing (RNA-seq) analysis of POMC neurons for determination of nicotine-induced gene expression changes and regulatory lncRNA/protein-coding gene interactions. Results Contrary to expectation based on previous studies, PNE did not impair but rather enhanced leptin-melanocortinergic regulation of energy and glucose balance via POMC neurons in offspring. RNA-seq of laser microdissected POMC neurons revealed only one consistent change, upregulation of Gm15851, a lncRNA of yet unidentified function, in nicotine-exposed offspring. RNA-seq further suggested 82 cis-regulatory lncRNA/protein-coding gene interactions, 19 of which involved coding genes regulating neural development and/or function, and revealed expression of several previously unidentified metabolic, neuroendocrine, and neurodevelopment pathways in POMC neurons. Conclusions PNE does not result in obesity and type 2 diabetes but instead enhances leptin-melanocortinergic feeding and body weight regulation via POMC neurons in adult offspring. PNE leads to selective upregulation of Gm15851, a lncRNA, in adult offspring POMC neurons. POMC neurons express several lncRNAs and pathways possibly regulating POMC neuronal development and/or function. Electronic supplementary material The online version of this article (doi:10.1186/s13073-016-0348-2) contains supplementary material, which is available to authorized users.
Collapse
|
36
|
Fan J, Ping J, Zhang WX, Rao YS, Liu HX, Zhang J, Yan YE. Prenatal and lactation nicotine exposure affects morphology and function of brown adipose tissue in male rat offspring. Ultrastruct Pathol 2016; 40:288-95. [DOI: 10.1080/01913123.2016.1223243] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
37
|
Shenassa ED, Wen X, Braid S. Exposure to Tobacco Metabolites via Breast Milk and Infant Weight Gain: A Population-Based Study. J Hum Lact 2016; 32:462-71. [PMID: 26644421 DOI: 10.1177/0890334415619154] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 11/02/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND Although the immutable benefits of breastfeeding are well documented, information on the potential consequences of exposure to tobacco metabolites specifically via breastfeeding is sparse. OBJECTIVE The aim was to conduct the first study of the association between exposure to tobacco metabolites specifically through breastfeeding and infant weight gain. METHODS We used historical data from the US Collaborative Perinatal Project. Mothers were classified as nonsmokers, light smokers (1-19 cigarettes/day), and heavy (20+ cigarettes/day) smokers. In-hospital feeding type was observed during a nursery stay after delivery. We conducted stratified analyses among average-for-gestational-age (AGA; N = 23 571) and small-for-gestational-age (SGA; N = 2552) infants. We isolated the effect of exposure to tobacco metabolites specifically through breastfeeding. RESULTS Overall, maternal smoking was associated with change in weight-for-length z-score in a dose-response manner. Change in weight z-score was most pronounced among SGA infants of heavy smokers (breastfed: 0.53; 95% confidence interval [CI], 0.12-0.94; formula fed: 0.17; 95% CI, 0.03-0.30). Exposure to tobacco metabolites specifically through breastfeeding was not associated with additional weight gain among AGA infants. Among the much smaller sample of SGA infants, exposure specifically through breastfeeding was associated with marginally significant additional weight gain (0.46; 95% CI, 0.00-0.91) among infants of heavy smokers. CONCLUSION Our findings are in accord with recommendations by health agencies for smokers to breastfeed. However, SGA infants exposed to tobacco metabolites via breastfeeding by heavy smokers appear to gain weight more rapidly than other infants. Practical implications of our findings are discussed.
Collapse
Affiliation(s)
- Edmond D Shenassa
- Maternal and Child Health Program, School of Public Health, University of Maryland, College Park, MD, USA Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD, USA Department of Epidemiology, School of Medicine, University of Maryland, Baltimore, MD, USA Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA
| | - Xiaozhong Wen
- Division of Behavioral Medicine, Department of Pediatrics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Susan Braid
- Department of Family and Community Health, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
38
|
Fan J, Ping J, Xiang J, Rao YS, Zhang WX, Chen T, Zhang L, Yan YE. Effects of prenatal and lactation nicotine exposure on glucose homeostasis, lipogenesis and lipid metabolic profiles in mothers and offspring. Toxicol Res (Camb) 2016; 5:1318-1328. [PMID: 30090436 DOI: 10.1039/c6tx00237d] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/20/2016] [Indexed: 01/07/2023] Open
Abstract
There is increasing evidence suggesting that maternal nicotine (NIC) exposure alone can lead to many deleterious consequences in the fetus. In this study, we aimed to evaluate the effects of prenatal and lactation NIC exposure on glucose homeostasis, lipogenesis and lipid metabolism in mothers and pups. After maternal NIC exposure (from gestational day 9 to weaning), NIC mothers showed lower body weight, decreased parametrial white adipose tissue (pWAT) and inguinal WAT weights, lower homeostasis model assessment of beta cell function, higher serum total cholesterol (TC) and low-density lipoprotein cholesterol levels, higher Castelli index values, lower hepatic mRNA levels of sterol regulatory element binding protein-1c (SREBP1c), lipoprotein lipase, acetyl-CoA carboxylase, fatty acid synthase (FAS) and glucose transporters 4 (GLUT4), as well as lower SREBP1c, FAS, leptin and GLUT4 mRNA levels in pWAT. However, female NIC pups presented higher body weights and serum TC levels, and increased trends for high density lipoprotein-cholesterol and Castelli index I. Male NIC pups had higher body weight, serum TC levels and Castelli index I values, and lower glycemia levels. Additionally, hepatic and adipose FAS gene expression from the female NIC pups presented a decreasing trend, while the male NIC pups had lower hepatic FAS expression and higher adipose FAS expression. In conclusion, prenatal and lactation NIC exposure induced deleterious effects on the glucose homeostasis, lipogenesis and lipid metabolism in both mothers and pups, which may promote several important metabolic disorders in the progeny. Additionally, there are gender-specific effects on pups.
Collapse
Affiliation(s)
- Jie Fan
- Department of Pharmacology , Basic Medical School of Wuhan University , Wuhan , 430071 , China . ; ; Tel: +86 27 68759222
| | - Jie Ping
- Department of Pharmacology , Basic Medical School of Wuhan University , Wuhan , 430071 , China . ; ; Tel: +86 27 68759222
| | - Jie Xiang
- Wuhan Medical treatment center , Wuhan , 430032 , China
| | - Yi-Song Rao
- Department of Pharmacology , Basic Medical School of Wuhan University , Wuhan , 430071 , China . ; ; Tel: +86 27 68759222
| | - Wan-Xia Zhang
- Department of Pharmacology , Basic Medical School of Wuhan University , Wuhan , 430071 , China . ; ; Tel: +86 27 68759222
| | - Ting Chen
- Department of Pharmacology , Basic Medical School of Wuhan University , Wuhan , 430071 , China . ; ; Tel: +86 27 68759222
| | - Li Zhang
- Department of Pharmacology , Basic Medical School of Wuhan University , Wuhan , 430071 , China . ; ; Tel: +86 27 68759222
| | - You-E Yan
- Department of Pharmacology , Basic Medical School of Wuhan University , Wuhan , 430071 , China . ; ; Tel: +86 27 68759222
| |
Collapse
|
39
|
Younes-Rapozo V, Moura EG, Manhães AC, Pinheiro CR, Carvalho JC, Barradas PC, de Oliveira E, Lisboa PC. Neonatal Nicotine Exposure Leads to Hypothalamic Gliosis in Adult Overweight Rats. J Neuroendocrinol 2015; 27:887-98. [PMID: 26453898 DOI: 10.1111/jne.12328] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/23/2015] [Accepted: 10/06/2015] [Indexed: 01/22/2023]
Abstract
Astrocytes and microglia, the immune competent cells of central nercous system, can be activated in response to metabolic signals such as obesity and hyperleptinaemia. In rats, maternal exposure to nicotine during lactation leads to central obesity, hyperleptinaemia, leptin resistance and alterations in hypothalamic neuropeptides in the offspring during adulthood. In the present study, we studied the activation of astrocytes and microglia, as well as the pattern of inflammatory mediators, in adult offspring of this experimental model. On postnatal day 2 (P2), osmotic minipumps releasing nicotine (NIC) (-6 mg/kg/day) or saline for 14 days were s.c. implanted in dams. Male offspring were killed on P180 and hypothalamic immunohistochemistry, retroperitoneal white adipose tissue (WAT) polymerase chain reaction analysis and multiplex analysis for plasma inflammatory mediators were carried out. At P180, NIC astrocyte cell number was higher in the arcuate nucleus (ARC) (medial: +82%; lateral: +110%), in the paraventricular nucleus (PVN) (+144%) and in the lateral hypothalamus (+121%). NIC glial fibrillary acidic protein fibre density was higher in the lateral ARC (+178%) and in the PVN (+183%). Interleukin-6 was not affected in the hypothalamus. NIC monocyte chemotactic protein 1 was only higher in the periventricular nucleus (+287%). NIC microglia (iba-1-positive) cell number was higher (+68%) only in the PVN, as was the chemokine (C-X3-C motif) receptor 1 density (+93%). NIC interleukin-10 was lower in the WAT (-58%) and plasma (-50%). Thus, offspring of mothers exposed to nicotine during lactation present hypothalamic astrogliosis at adulthood and microgliosis in the PVN.
Collapse
Affiliation(s)
- V Younes-Rapozo
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - E G Moura
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - A C Manhães
- Laboratório de Neurofisiologia, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - C R Pinheiro
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - J C Carvalho
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - P C Barradas
- Laboratório de Neurobiologia, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - E de Oliveira
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - P C Lisboa
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
40
|
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. [DOI: 10.1016/j.physbeh.2015.05.040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 05/26/2015] [Accepted: 05/31/2015] [Indexed: 11/18/2022]
|
41
|
McPherson NO, Bell VG, Zander-Fox DL, Fullston T, Wu LL, Robker RL, Lane M. When two obese parents are worse than one! Impacts on embryo and fetal development. Am J Physiol Endocrinol Metab 2015. [PMID: 26199280 DOI: 10.1152/ajpendo.00230.2015] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The prevalence of overweight and obesity in reproductive-age adults is increasing worldwide. While the effects of either paternal or maternal obesity on gamete health and subsequent fertility and pregnancy have been reported independently, the combination of having both parents overweight/obese on fecundity and offspring health has received minimal attention. Using a 2 × 2 study design in rodents we established the relative contributions of paternal and maternal obesity on fetal and embryo development and whether combined paternal and maternal obesity had an additive effect. Here, we show that parental obesity reduces fetal and placental weights without altering pregnancy establishment and is not dependent on an in utero exposure to a high-fat diet. Interestingly combined parental obesity seemed to accumulate both the negative influences of paternal and maternal obesity had alone on embryo and fetal health rather than an amplification, manifested as reduced embryo developmental competency, reduced blastocyst cell numbers, impaired mitochondrial function, and alterations to active and repressive embryonic chromatin marks, resulting in aberrant placental gene expression and reduced fetal liver mtDNA copy numbers. Further understanding both the maternal cytoplasmic and paternal genetic interactions during this early developmental time frame will be vital for understanding how developmental programming is regulated and for the proposition of interventions to mitigate their effects.
Collapse
Affiliation(s)
- N O McPherson
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia; Freemasons Centre for Men's Health, University of Adelaide, South Australia, Australia; Repromed, Dulwich, South Australia, Australia; and
| | - V G Bell
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia; Freemasons Centre for Men's Health, University of Adelaide, South Australia, Australia; Repromed, Dulwich, South Australia, Australia; and
| | - D L Zander-Fox
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia; Repromed, Dulwich, South Australia, Australia; and
| | - T Fullston
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia
| | - L L Wu
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia
| | - R L Robker
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia
| | - M Lane
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia; Monash In Vitro Fertilisation Group, Richmond, Australia
| |
Collapse
|
42
|
Leach PT, Gould TJ. Thyroid hormone signaling: Contribution to neural function, cognition, and relationship to nicotine. Neurosci Biobehav Rev 2015; 57:252-63. [PMID: 26344666 DOI: 10.1016/j.neubiorev.2015.09.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 08/25/2015] [Accepted: 09/02/2015] [Indexed: 01/11/2023]
Abstract
Cigarette smoking is common despite its adverse effects on health, such as cardiovascular disease and stroke. Understanding the mechanisms that contribute to the addictive properties of nicotine makes it possible to target them to prevent the initiation of smoking behavior and/or increase the chance of successful quit attempts. While highly addictive, nicotine is not generally considered to be as reinforcing as other drugs of abuse. There are likely other mechanisms at work that contribute to the addictive liability of nicotine. Nicotine modulates aspects of the endocrine system, including the thyroid, which is critical for normal cognitive functioning. It is possible that nicotine's effects on thyroid function may alter learning and memory, and this may underlie some of its addictive potential. Here, we review the literature on thyroid function and cognition, with a focus on how nicotine alters thyroid hormone signaling and the potential impact on cognition. Changes in cognition are a major symptom of nicotine addiction. Current anti-smoking therapies have modest success at best. If some of the cognitive effects of nicotine are mediated through the thyroid hormone system, then thyroid hormone agonists may be novel treatments for smoking cessation therapies. The content of this review is important because it clarifies the relationship between smoking and thyroid function, which has been ill-defined in the past. This review is timely because the reduction in smoking rates we have seen in recent decades, due to public awareness campaigns and public smoking bans, has leveled off in recent years. Therefore, novel treatment approaches are needed to help reduce smoking rates further.
Collapse
Affiliation(s)
- Prescott T Leach
- Temple University Department of Psychology, Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Thomas J Gould
- Temple University Department of Psychology, Neuroscience Program, Temple University, Philadelphia, PA 19122, United States.
| |
Collapse
|
43
|
Exposure to nicotine increases dopamine receptor content in the mesocorticolimbic pathway of rat dams and offspring during lactation. Pharmacol Biochem Behav 2015. [DOI: 10.1016/j.pbb.2015.07.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
44
|
Pinheiro C, Moura E, Manhães A, Fraga M, Claudio-Neto S, Abreu-Villaça Y, Oliveira E, Lisboa P. Concurrent maternal and pup postnatal tobacco smoke exposure in Wistar rats changes food preference and dopaminergic reward system parameters in the adult male offspring. Neuroscience 2015; 301:178-92. [DOI: 10.1016/j.neuroscience.2015.06.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/30/2015] [Accepted: 06/02/2015] [Indexed: 12/26/2022]
|
45
|
Lisboa PC, de Oliveira E, Manhães AC, Santos-Silva AP, Pinheiro CR, Younes-Rapozo V, Faustino LC, Ortiga-Carvalho TM, Moura EG. Effects of maternal nicotine exposure on thyroid hormone metabolism and function in adult rat progeny. J Endocrinol 2015; 224:315-25. [PMID: 25653393 DOI: 10.1530/joe-14-0473] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Postnatal nicotine exposure leads to obesity and hypothyroidism in adulthood. We studied the effects of maternal nicotine exposure during lactation on thyroid hormone (TH) metabolism and function in adult offspring. Lactating rats received implants of osmotic minipumps releasing nicotine (NIC, 6 mg/kg per day s.c.) or saline (control) from postnatal days 2 to 16. Offspring were killed at 180 days. We measured types 1 and 2 deiodinase activity and mRNA, mitochondrial α-glycerol-3-phosphate dehydrogenase (mGPD) activity, TH receptor (TR), uncoupling protein 1 (UCP1), hypothalamic TRH, pituitary TSH, and in vitro TRH-stimulated TSH secretion. Expression of deiodinase mRNAs followed the same profile as that of the enzymatic activity. NIC exposure caused lower 5'-D1 and mGPD activities; lower TRβ1 content in liver as well as lower 5'-D1 activity in muscle; and higher 5'-D2 activity in brown adipose tissue (BAT), heart, and testis, which are in accordance with hypothyroidism. Although deiodinase activities were not changed in the hypothalamus, pituitary, and thyroid of NIC offspring, UCP1 expression was lower in BAT. Levels of both TRH and TSH were lower in offspring exposed to NIC, which presented higher basal in vitro TSH secretion, which was not increased in response to TRH. Thus, the hypothyroidism in NIC offspring at adulthood was caused, in part, by in vivo TRH-TSH suppression and lower sensitivity to TRH. Despite the hypothyroid status of peripheral tissues, these animals seem to develop an adaptive mechanism to preserve thyroxine to triiodothyronine conversion in central tissues.
Collapse
Affiliation(s)
- P C Lisboa
- Laboratory of Endocrine PhysiologyLaboratory of NeurophysiologyBiology Institute, State University of Rio de Janeiro, Rio de Janeiro 20551-030, BrazilCarlos Chagas Filho Biophysic InstituteFederal University of Rio de Janeiro, Rio de Janeiro 21949-900, Brazil
| | - E de Oliveira
- Laboratory of Endocrine PhysiologyLaboratory of NeurophysiologyBiology Institute, State University of Rio de Janeiro, Rio de Janeiro 20551-030, BrazilCarlos Chagas Filho Biophysic InstituteFederal University of Rio de Janeiro, Rio de Janeiro 21949-900, Brazil
| | - A C Manhães
- Laboratory of Endocrine PhysiologyLaboratory of NeurophysiologyBiology Institute, State University of Rio de Janeiro, Rio de Janeiro 20551-030, BrazilCarlos Chagas Filho Biophysic InstituteFederal University of Rio de Janeiro, Rio de Janeiro 21949-900, Brazil
| | - A P Santos-Silva
- Laboratory of Endocrine PhysiologyLaboratory of NeurophysiologyBiology Institute, State University of Rio de Janeiro, Rio de Janeiro 20551-030, BrazilCarlos Chagas Filho Biophysic InstituteFederal University of Rio de Janeiro, Rio de Janeiro 21949-900, Brazil
| | - C R Pinheiro
- Laboratory of Endocrine PhysiologyLaboratory of NeurophysiologyBiology Institute, State University of Rio de Janeiro, Rio de Janeiro 20551-030, BrazilCarlos Chagas Filho Biophysic InstituteFederal University of Rio de Janeiro, Rio de Janeiro 21949-900, Brazil
| | - V Younes-Rapozo
- Laboratory of Endocrine PhysiologyLaboratory of NeurophysiologyBiology Institute, State University of Rio de Janeiro, Rio de Janeiro 20551-030, BrazilCarlos Chagas Filho Biophysic InstituteFederal University of Rio de Janeiro, Rio de Janeiro 21949-900, Brazil Laboratory of Endocrine PhysiologyLaboratory of NeurophysiologyBiology Institute, State University of Rio de Janeiro, Rio de Janeiro 20551-030, BrazilCarlos Chagas Filho Biophysic InstituteFederal University of Rio de Janeiro, Rio de Janeiro 21949-900, Brazil
| | - L C Faustino
- Laboratory of Endocrine PhysiologyLaboratory of NeurophysiologyBiology Institute, State University of Rio de Janeiro, Rio de Janeiro 20551-030, BrazilCarlos Chagas Filho Biophysic InstituteFederal University of Rio de Janeiro, Rio de Janeiro 21949-900, Brazil
| | - T M Ortiga-Carvalho
- Laboratory of Endocrine PhysiologyLaboratory of NeurophysiologyBiology Institute, State University of Rio de Janeiro, Rio de Janeiro 20551-030, BrazilCarlos Chagas Filho Biophysic InstituteFederal University of Rio de Janeiro, Rio de Janeiro 21949-900, Brazil
| | - E G Moura
- Laboratory of Endocrine PhysiologyLaboratory of NeurophysiologyBiology Institute, State University of Rio de Janeiro, Rio de Janeiro 20551-030, BrazilCarlos Chagas Filho Biophysic InstituteFederal University of Rio de Janeiro, Rio de Janeiro 21949-900, Brazil
| |
Collapse
|
46
|
Early weaning by maternal prolactin inhibition leads to higher neuropeptide Y and astrogliosis in the hypothalamus of the adult rat offspring. Br J Nutr 2015; 113:536-45. [DOI: 10.1017/s0007114514003882] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The suppression of prolactin production with bromocriptine (BRO) in the last 3 d of lactation reduces milk yield (early weaning) and increases the transfer of leptin through the milk, causing hyperleptinaemia in pups. In adulthood, several changes occur in the offspring as a result of metabolic programming, including overweight, higher visceral fat mass, hypothyroidism, hyperglycaemia, insulin resistance, hyperleptinaemia and central leptin resistance. In the present study, we investigated whether overweight rats programmed by early weaning with maternal BRO treatment have hypothalamic alterations in adulthood. We analysed the expression of neuropeptide Y (NPY), cocaine- and amphetamine-regulated transcript (CART), pro-opiomelanocortin (POMC) and α-melanocyte-stimulating hormone (α-MSH) by immunohistochemistry in the following hypothalamic nuclei: medial and lateral arcuate nucleus (ARC); paraventricular nucleus (PVN); lateral hypothalamus (LH). Additionally, we sought to determine whether these programmed rats exhibited hypothalamic inflammation as indicated by astrogliosis. NPY immunostaining showed a denser NPY-positive fibre network in the ARC and PVN (+82 % in both nuclei) of BRO offspring. Regarding the anorexigenic neuropeptides, no difference was found for CART, POMC and α-MSH. The number of astrocytes was higher in all the nuclei of BRO rats. The fibre density of glial fibrillary acidic protein was also increased in both medial and lateral ARC (6·06-fold increase and 9·13-fold increase, respectively), PVN (5·75-fold increase) and LH (2·68-fold increase) of BRO rats. We suggest that early weaning has a long-term effect on the expression of NPY as a consequence of developmental plasticity, and the presence of astrogliosis indicates hypothalamic inflammation that is closely related to overweight and hyperleptinaemia observed in our model.
Collapse
|
47
|
Primo CC, Ruela PBF, Brotto LDDA, Garcia TR, Lima EDF. Effects of maternal nicotine on breastfeeding infants. REVISTA PAULISTA DE PEDIATRIA 2014; 31:392-7. [PMID: 24142324 PMCID: PMC4182966 DOI: 10.1590/s0103-05822013000300018] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 04/23/2013] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To assess scientific evidence about the effects of maternal nicotine on infant by an integrative review. DATA SOURCES Studies published in Portuguese, English and Spanish, from 1990 to 2009, with abstracts available in the Latin American Health Sciences Literature (Lilacs) and Medical Literature Analysis and Retrieval System On-Line (Medline) databases. The descriptors were: "breastfeeding", "lactation" and "smoking". DATA SYNTHESIS The main identified effects of nicotine on infants were: changes in sleep and wakefulness patterns; reduction of iodine supply; hystopathological damage on liver and lung; intracellular oxidative damage; reduction of pancreatic ß cells; and decreased glucose tolerance. CONCLUSIONS It is recommended to inform mothers about harmful chemicals contained in cigarettes that can be secreted into breast milk. They should be strongly encouraged to stop smoking during lactation.
Collapse
|
48
|
Riedel C, Schönberger K, Yang S, Koshy G, Chen YC, Gopinath B, Ziebarth S, von Kries R. Parental smoking and childhood obesity: higher effect estimates for maternal smoking in pregnancy compared with paternal smoking--a meta-analysis. Int J Epidemiol 2014; 43:1593-606. [PMID: 25080528 DOI: 10.1093/ije/dyu150] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Some studies reported similar effect estimates for the impact of maternal smoking in pregnancy and paternal smoking on childhood obesity, whereas others suggested higher effects for maternal smoking. We performed a meta-analysis to compare the effect of in utero exposure to maternal smoking and that of paternal or household smoking exposure in utero or after birth with mutual adjustment. METHODS Meta-analysis of observational studies identified in MEDLINE, EMBASE and Web of Knowledge published in 1900-2013. Study inclusion criterion was assessment of the association of maternal smoking during pregnancy and paternal or household smoking (anyone living in the household who smokes) at any time with childhood overweight and obesity. The analyses were based on all studies with mutually adjusted effect estimates for maternal and paternal/household smoking applying a random-effects model. RESULTS Data for 109,838 mother/child pairs were reported in 12 studies. The pooled odds ratios (ORs) for overweight 1.33 [95% confidence interval (CI) 1.23;1.44] (n=6, I2=0.00%) and obesity 1.60 (95% CI 1.37;1.88) (n=4, I2=32.47%) for maternal smoking during pregnancy were higher than for paternal smoking: 1.07 (95% CI 1.00;1.16) (n=6, I2=41.34%) and 1.23 (95% CI 1.10;1.38) (n=4, I2=14.61%), respectively. Similar estimates with widely overlapping confidence limits were found for maternal smoking during pregnancy and childhood overweight and obesity: 1.35 (95% CI 1.20;1.51) (n=3, I2=0.00%) and 1.28 (95% CI 1.07;1.54) (n=3, I2=0.00%) compared with household smoking 1.22 (95% CI 1.06;1.39) (n=3, I2=72.14%) and 1.31 (95% CI 1.15;1.50)] (n=3, I2=0.00%). CONCLUSIONS Higher effect estimates for maternal smoking in pregnancy compared with paternal smoking in mutually adjusted models may suggest a direct intrauterine effect.
Collapse
Affiliation(s)
- Christina Riedel
- Ludwig Maximilian University of Munich, Institute of Social Paediatrics and Adolescent Medicine, Munich, Germany, Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Canada, Child and Reproductive Health Group, Liverpool School of Tropical Medicine, Liverpool, UK, Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan, Department of Family Medicine, Taipei City Hospital, ZhongXing Branch, Taipei, Taiwan and Centre for Vision Research, Department of Ophthalmology and Westmead Millennium Institute, University of Sydney, Sydney, Australia
| | - Katharina Schönberger
- Ludwig Maximilian University of Munich, Institute of Social Paediatrics and Adolescent Medicine, Munich, Germany, Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Canada, Child and Reproductive Health Group, Liverpool School of Tropical Medicine, Liverpool, UK, Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan, Department of Family Medicine, Taipei City Hospital, ZhongXing Branch, Taipei, Taiwan and Centre for Vision Research, Department of Ophthalmology and Westmead Millennium Institute, University of Sydney, Sydney, Australia
| | - Seungmi Yang
- Ludwig Maximilian University of Munich, Institute of Social Paediatrics and Adolescent Medicine, Munich, Germany, Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Canada, Child and Reproductive Health Group, Liverpool School of Tropical Medicine, Liverpool, UK, Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan, Department of Family Medicine, Taipei City Hospital, ZhongXing Branch, Taipei, Taiwan and Centre for Vision Research, Department of Ophthalmology and Westmead Millennium Institute, University of Sydney, Sydney, Australia
| | - Gibby Koshy
- Ludwig Maximilian University of Munich, Institute of Social Paediatrics and Adolescent Medicine, Munich, Germany, Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Canada, Child and Reproductive Health Group, Liverpool School of Tropical Medicine, Liverpool, UK, Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan, Department of Family Medicine, Taipei City Hospital, ZhongXing Branch, Taipei, Taiwan and Centre for Vision Research, Department of Ophthalmology and Westmead Millennium Institute, University of Sydney, Sydney, Australia
| | - Yang-Ching Chen
- Ludwig Maximilian University of Munich, Institute of Social Paediatrics and Adolescent Medicine, Munich, Germany, Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Canada, Child and Reproductive Health Group, Liverpool School of Tropical Medicine, Liverpool, UK, Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan, Department of Family Medicine, Taipei City Hospital, ZhongXing Branch, Taipei, Taiwan and Centre for Vision Research, Department of Ophthalmology and Westmead Millennium Institute, University of Sydney, Sydney, Australia Ludwig Maximilian University of Munich, Institute of Social Paediatrics and Adolescent Medicine, Munich, Germany, Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Canada, Child and Reproductive Health Group, Liverpool School of Tropical Medicine, Liverpool, UK, Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan, Department of Family Medicine, Taipei City Hospital, ZhongXing Branch, Taipei, Taiwan and Centre for Vision Research, Department of Ophthalmology and Westmead Millennium Institute, University of Sydney, Sydney, Australia
| | - Bamini Gopinath
- Ludwig Maximilian University of Munich, Institute of Social Paediatrics and Adolescent Medicine, Munich, Germany, Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Canada, Child and Reproductive Health Group, Liverpool School of Tropical Medicine, Liverpool, UK, Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan, Department of Family Medicine, Taipei City Hospital, ZhongXing Branch, Taipei, Taiwan and Centre for Vision Research, Department of Ophthalmology and Westmead Millennium Institute, University of Sydney, Sydney, Australia
| | - Stephanie Ziebarth
- Ludwig Maximilian University of Munich, Institute of Social Paediatrics and Adolescent Medicine, Munich, Germany, Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Canada, Child and Reproductive Health Group, Liverpool School of Tropical Medicine, Liverpool, UK, Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan, Department of Family Medicine, Taipei City Hospital, ZhongXing Branch, Taipei, Taiwan and Centre for Vision Research, Department of Ophthalmology and Westmead Millennium Institute, University of Sydney, Sydney, Australia
| | - Rüdiger von Kries
- Ludwig Maximilian University of Munich, Institute of Social Paediatrics and Adolescent Medicine, Munich, Germany, Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Canada, Child and Reproductive Health Group, Liverpool School of Tropical Medicine, Liverpool, UK, Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan, Department of Family Medicine, Taipei City Hospital, ZhongXing Branch, Taipei, Taiwan and Centre for Vision Research, Department of Ophthalmology and Westmead Millennium Institute, University of Sydney, Sydney, Australia
| |
Collapse
|
49
|
Riedel C, Fenske N, Müller MJ, Plachta-Danielzik S, Keil T, Grabenhenrich L, von Kries R. Differences in BMI z-scores between offspring of smoking and nonsmoking mothers: a longitudinal study of German children from birth through 14 years of age. ENVIRONMENTAL HEALTH PERSPECTIVES 2014; 122:761-767. [PMID: 24695368 PMCID: PMC4080541 DOI: 10.1289/ehp.1307139] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 03/31/2014] [Indexed: 05/30/2023]
Abstract
BACKGROUND Children of mothers who smoked during pregnancy have a lower birth weight but have a higher chance to become overweight during childhood. OBJECTIVES We followed children longitudinally to assess the age when higher body mass index (BMI) z-scores became evident in the children of mothers who smoked during pregnancy, and to evaluate the trajectory of changes until adolescence. METHODS We pooled data from two German cohort studies that included repeated anthropometric measurements until 14 years of age and information on smoking during pregnancy and other risk factors for overweight. We used longitudinal quantile regression to estimate age- and sex-specific associations between maternal smoking and the 10th, 25th, 50th, 75th, and 90th quantiles of the BMI z-score distribution in study participants from birth through 14 years of age, adjusted for potential confounders. We used additive mixed models to estimate associations with mean BMI z-scores. RESULTS Mean and median (50th quantile) BMI z-scores at birth were smaller in the children of mothers who smoked during pregnancy compared with children of nonsmoking mothers, but BMI z-scores were significantly associated with maternal smoking beginning at the age of 4-5 years, and differences increased over time. For example, the difference in the median BMI z-score between the daughters of smokers versus nonsmokers was 0.12 (95% CI: 0.01, 0.21) at 5 years, and 0.30 (95% CI: 0.08, 0.39) at 14 years of age. For lower BMI z-score quantiles, the association with smoking was more pronounced in girls, whereas in boys the association was more pronounced for higher BMI z-score quantiles. CONCLUSIONS A clear difference in BMI z-score (mean and median) between children of smoking and nonsmoking mothers emerged at 4-5 years of age. The shape and size of age-specific effect estimates for maternal smoking during pregnancy varied by age and sex across the BMI z-score distribution.
Collapse
|
50
|
Zinkhan EK, Lang BY, Yu B, Wang Y, Jiang C, Fitzhugh M, Dahl M, Campbell MS, Fung C, Malleske D, Albertine KH, Joss-Moore L, Lane RH. Maternal tobacco smoke increased visceral adiposity and serum corticosterone levels in adult male rat offspring. Pediatr Res 2014; 76:17-23. [PMID: 24727947 DOI: 10.1038/pr.2014.58] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 01/24/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND Maternal tobacco smoke (MTS) predisposes human and rat offspring to visceral obesity in early adulthood. Glucocorticoid excess also causes visceral obesity. We hypothesized that in utero MTS would increase visceral adiposity and alter the glucocorticoid pathway in young adult rats. METHODS We developed a novel model of in utero MTS exposure in pregnant rats by exposing them to cigarette smoke from E11.5 to term. Neonatal rats were cross-fostered to control dams and weaned to standard rat chow through young adulthood (postnatal day 60). RESULTS We demonstrated increased visceral adiposity (193%)*, increased visceral adipose 11-β hydroxysteroid dehydrogenase 1 mRNA (204%)*, increased serum corticosterone (147%)*, and no change in glucocorticoid receptor protein in adult male MTS rat offspring. Female rats exposed to MTS in utero demonstrated no change in visceral or subcutaneous adiposity, decreased serum corticosterone (60%)*, and decreased adipose glucocorticoid receptor protein (66%)*. *P < 0.05. CONCLUSION We conclude that in utero MTS exposure increased visceral adiposity and altered in the glucocorticoid pathway in a sex-specific manner. We speculate that in utero MTS exposure programs adipose dysfunction in adult male rat offspring via alteration in the glucocorticoid pathway.
Collapse
Affiliation(s)
- Erin K Zinkhan
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah
| | - Brook Y Lang
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah
| | - Baifeng Yu
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah
| | - Yan Wang
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah
| | - Chengshe Jiang
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah
| | - Melanie Fitzhugh
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah
| | - Marjanna Dahl
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah
| | - Michael S Campbell
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah
| | - Camille Fung
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah
| | - Daniel Malleske
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah
| | - Kurt H Albertine
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah
| | - Lisa Joss-Moore
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah
| | - Robert H Lane
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|