151
|
Mirzakhani H, Carey VJ, McElrath TF, Qiu W, Hollis BW, O’Connor GT, Zeiger RS, Bacharier L, Litonjua AA, Weiss ST. Impact of Preeclampsia on the Relationship between Maternal Asthma and Offspring Asthma. An Observation from the VDAART Clinical Trial. Am J Respir Crit Care Med 2019; 199:32-42. [PMID: 30153046 PMCID: PMC6353019 DOI: 10.1164/rccm.201804-0770oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/27/2018] [Indexed: 01/27/2023] Open
Abstract
RATIONALE Maternal asthma and preeclampsia have independently been reported to be associated with increased asthma incidence in children of affected mothers. Maternal asthma is also associated with increased risk of preeclampsia development. However, the joint effect of these maternal conditions on child asthma risk is unknown. OBJECTIVES To study whether development of preeclampsia among pregnant women with asthma was associated with higher risk of childhood asthma in the VDAART (Vitamin D Antenatal Asthma Reduction Trial). METHODS A total of 806 pregnant women and their offspring at high risk of asthma or atopy, who were followed from VDAART enrollment (10-18 wk of gestation) through the child's third birthday, were included in this cohort analysis. Preeclampsia status was determined by chart review, obstetrician diagnosis, and adjudication by a panel of obstetricians. Child asthma was the main outcome as determined by parental report of a physician diagnosis, and the risk of child asthma was also examined if accompanied by recurrent wheeze. The main risk variable of interest was a four-level ordered variable defined for each mother, with values without asthma without preeclampsia, without asthma with preeclampsia, with asthma without preeclampsia, and with asthma with preeclampsia during their pregnancy. We examined the trend of outcome proportions across these categories. To account for differences in maternal and child characteristics, we used a Weibull regression model for interval-censored data to compare the incidence of child asthma by age of 3 years across the maternal variable categories. MEASUREMENTS AND MAIN RESULTS The incidence of asthma in 3-year-old children was 9.90% (44/445), 17.95% (7/39), 22.11% (65/294), and 32.14% (9/28) among those born to mothers without asthma and without preeclampsia, mothers without asthma with preeclampsia, mothers with asthma without preeclampsia, and mothers with asthma with preeclampsia, respectively. The incidences demonstrated an increasing trend in risk of child asthma across the maternal groups (P for trend <0.001). After accounting for potential confounders and using time to report of childhood asthma as analysis outcome, risk of asthma was greater among children born to mothers with asthma without preeclampsia, compared with mothers without asthma without preeclampsia (adjusted hazard ratio, 2.18; 95% confidence interval, 1.46-3.26). This risk was 50% greater for children born to mothers with asthma who developed preeclampsia during pregnancy (adjusted hazard ratio, 2.68; 95% confidence interval, 1.30-5.61). The trend in asthma and recurrent wheeze proportions across the maternal groups' children also indicated a higher risk for children born to mothers with asthma with preeclampsia (adjusted hazard ratio, 4.73; 95% confidence interval, 2.20-10.07; P for trend <0.001). CONCLUSIONS Preeclampsia is associated with increased risk of early life childhood asthma in children less than 3 years old over and above that associated with maternal asthma alone. The results implicate the interplay between maternal factors as strong predictors of offspring asthma and in utero maternal-fetal immune perturbations and developmental dysregulations associated with preeclampsia.
Collapse
Affiliation(s)
- Hooman Mirzakhani
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Vincent J. Carey
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Thomas F. McElrath
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Weiliang Qiu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bruce W. Hollis
- Department of Pediatrics, Medical College of South Carolina, Charleston, South Carolina
| | - George T. O’Connor
- Pulmonary Center, Department of Medicine, Boston Medical Center, Boston University, Boston, Massachusetts
| | - Robert S. Zeiger
- Department of Allergy and Research and Evaluation, Kaiser Permanente Southern California, Region, San Diego and Pasadena, California
| | - Leonard Bacharier
- Division of Pediatric Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Washington University, St. Louis, Missouri
| | - Augusto A. Litonjua
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, Golisano Children’s Hospital at Strong, University of Rochester Medical Center, Rochester, New York; and
| | - Scott T. Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Partners Center for Personalized Medicine, Partners Health Care, Boston, Massachusetts
| |
Collapse
|
152
|
Kampmann U, Knorr S, Fuglsang J, Ovesen P. Determinants of Maternal Insulin Resistance during Pregnancy: An Updated Overview. J Diabetes Res 2019; 2019:5320156. [PMID: 31828161 PMCID: PMC6885766 DOI: 10.1155/2019/5320156] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/11/2019] [Accepted: 11/05/2019] [Indexed: 01/08/2023] Open
Abstract
Insulin resistance changes over time during pregnancy, and in the last half of the pregnancy, insulin resistance increases considerably and can become severe, especially in women with gestational diabetes and type 2 diabetes. Numerous factors such as placental hormones, obesity, inactivity, an unhealthy diet, and genetic and epigenetic contributions influence insulin resistance in pregnancy, but the causal mechanisms are complex and still not completely elucidated. In this review, we strive to give an overview of the many components that have been ascribed to contribute to the insulin resistance in pregnancy. Knowledge about the causes and consequences of insulin resistance is of extreme importance in order to establish the best possible treatment during pregnancy as severe insulin resistance can result in metabolic dysfunction in both mother and offspring on a short as well as long-term basis.
Collapse
Affiliation(s)
- Ulla Kampmann
- Steno Diabetes Center Aarhus, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Sine Knorr
- Steno Diabetes Center Aarhus, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Jens Fuglsang
- Department of Obstetrics and Gynecology, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Per Ovesen
- Department of Obstetrics and Gynecology, Aarhus University Hospital, 8200 Aarhus N, Denmark
| |
Collapse
|
153
|
Wu Y, Peterson KE, Sánchez BN, Dolinoy DC, Mercado-Garcia A, Téllez-Rojo MM, Goodrich JM. Association of blood leukocyte DNA methylation at LINE-1 and growth-related candidate genes with pubertal onset and progression. Epigenetics 2018; 13:1222-1233. [PMID: 30582410 PMCID: PMC6986794 DOI: 10.1080/15592294.2018.1556198] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 10/01/2018] [Accepted: 11/21/2018] [Indexed: 01/15/2023] Open
Abstract
Puberty is a developmentally plastic phase. Variations in pubertal tempo have implications for the risk of later adult diseases. Influences on pubertal tempo have been widely discussed, but the underlying biological mechanisms remain unclear. Epigenetic modifications are known to regulate development processes; they could play an important role in affecting pubertal outcomes. We conducted a population-based analysis to investigate the association of peripubertal blood DNA methylation at LINE-1 and growth-related candidate genes with pubertal onset and progression in healthy adolescents. The analytic sample included 114 males and 129 females aged 10 to 18 years. DNA methylation at growth-related candidate loci IGF2, H19, HSD11B2, as well as LINE-1 repetitive elements were quantified. Cox survival and ordinal regression models were used to examine sex- and locus-specific associations of epigenetic markers with pubertal development using physician-assessed Tanner stages and self-reported menarche, adjusted for covariates. Among boys, DNA methylation at H19 was associated with later pubarche. HSD11B2 methylation was associated with earlier onset of pubic hair and genitalia development and slower pubertal progression. IGF2 was associated with later onset of genital development. Among girls, LINE-1 methylation was associated with later onset of breast development. For each percent increase of methylation at H19, there was 5% increased odds in the earlier onset of breast development. DNA methylation of IGF2 was associated with earlier onset of pubic hair. DNA methylation at genes known to influence early-life growth may also influence pubertal outcomes.
Collapse
Affiliation(s)
- Yue Wu
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Karen E. Peterson
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Human Growth and Development, University of Michigan, Ann Arbor, MI, USA
| | - Brisa N. Sánchez
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Dana C. Dolinoy
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Human Growth and Development, University of Michigan, Ann Arbor, MI, USA
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Adriana Mercado-Garcia
- Center for Research on Nutrition and Health, National Institute of Public Health, Cuernavaca, Morelos, México
| | - Martha M. Téllez-Rojo
- Center for Research on Nutrition and Health, National Institute of Public Health, Cuernavaca, Morelos, México
| | - Jaclyn M. Goodrich
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| |
Collapse
|
154
|
Hoffmann A, Spengler D. The Mitochondrion as Potential Interface in Early-Life Stress Brain Programming. Front Behav Neurosci 2018; 12:306. [PMID: 30574076 PMCID: PMC6291450 DOI: 10.3389/fnbeh.2018.00306] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/26/2018] [Indexed: 12/23/2022] Open
Abstract
Mitochondria play a central role in cellular energy-generating processes and are master regulators of cell life. They provide the energy necessary to reinstate and sustain homeostasis in response to stress, and to launch energy intensive adaptation programs to ensure an organism’s survival and future well-being. By this means, mitochondria are particularly apt to mediate brain programming by early-life stress (ELS) and to serve at the same time as subcellular substrate in the programming process. With a focus on mitochondria’s integrated role in metabolism, steroidogenesis and oxidative stress, we review current findings on altered mitochondrial function in the brain, the placenta and peripheral blood cells following ELS-dependent programming in rodents and recent insights from humans exposed to early life adversity (ELA). Concluding, we propose a role of the mitochondrion as subcellular intersection point connecting ELS, brain programming and mental well-being, and a role as a potential site for therapeutic interventions in individuals exposed to severe ELS.
Collapse
Affiliation(s)
- Anke Hoffmann
- Epigenomics of Early Life, Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Dietmar Spengler
- Epigenomics of Early Life, Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
155
|
Fournier T, Poulain JP. Eating According to One's Genes? Exploring the French Public's Understanding of and Reactions to Personalized Nutrition. QUALITATIVE HEALTH RESEARCH 2018; 28:2195-2207. [PMID: 30132729 DOI: 10.1177/1049732318793417] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In this article, we analyze qualitatively the understanding of and reactions to personalized nutrition (PN) among the French public. Focus groups were conducted to identify the opinions and discourses about two applications of knowledge from nutritional (epi)genomics: a biotechnology (nutrigenetic testing) and a public awareness campaign (the "first thousand days of life" initiative). Our objective was to understand to what extent PN could lead to changes in eating practices as well as in the representations of food-health relationships within France, a country characterized by a strong commitment to commensality and a certain "nutritional relativism." Although discourses on nutritional genomics testify to a resistance to food medicalization, nutritional epigenomics appears as more performative because it introduces the question of transgenerational transmission, thus parental responsibility.
Collapse
Affiliation(s)
- Tristan Fournier
- 1 Institut de Recherche Interdisciplinaire sur les enjeux Sociaux (Iris), Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Jean-Pierre Poulain
- 2 Université de Toulouse, Toulouse, France
- 3 Centre d'Etude et de Recherche Travail Organisation Pouvoir (Certop), Centre National de la Recherche Scientifique (CNRS), Toulouse, France
- 4 Laboratoire International Associé (LIA) "Food, Cultures and Health", Centre National de la Recherche Scientifique (CNRS), Taylor's University, Subang Jaya, Malaysia
| |
Collapse
|
156
|
Abstract
Research in the field of gerontology has traditionally focused on later life stages. There is increasing evidence, however, that both the rate of age-related functional decline and the later-life health status can be programmed during early development. The central role of epigenetic mechanisms (methylation of DNA, histone modifications and regulation by non-coding RNAs) in mediating these long-term effects has been elucidated. Both rate and direction of age-associated change of epigenetic patterns (“epigenetic drift”) were shown to be largely dependent on early-life environmental conditions. Inter-individual divergences in epigenetic profiles may arise following the stochastic errors in maintaining epigenetic marks, but they may also be adaptively mediated by specific environmental cues. Recent cohort studies indicate that ticking rate of epigenetic clock, estimated by a DNA methylation-based methods, may be developmentally adjusted, and that individual’s discrepancies among epigenetic and chronological age would be likely programmed early in development. In this Perspective article, recent findings suggesting the importance of early-life determinants for life-course dynamics of epigenetic drift are summarized and discussed.
Collapse
|
157
|
Larval crowding results in hormesis-like effects on longevity in Drosophila: timing of eclosion as a model. Biogerontology 2018; 20:191-201. [PMID: 30456589 DOI: 10.1007/s10522-018-9786-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/14/2018] [Indexed: 01/20/2023]
Abstract
There is increasing evidence that stress during development can affect adult-life health status and longevity. In the present study, we examined life span (LS), fly weight, fecundity and expression levels of longevity-associated genes (Hsp70, InR, dSir2, dTOR and dFOXO) in adult Drosophila melanogaster flies reared in normal [low density (LD), ~ 300-400 eggs per jar] or crowded [high density (HD), more than 3000 eggs per jar] conditions by using the order (day) of emergence as an index of the developmental duration (HD1-5 groups). Developmental time showed a significant trend to increase while weight showed a significant trend to decrease with increasing the timing of emergence. In both males and females eclosed during first 2 days in HD conditions (HD1 and HD2 groups), both mean and maximum LSs were significantly increased in comparison to LD group. In males, mean LS was increased by 24.0% and 23.5% in HD1 and HD2 groups, respectively. In females, corresponding increments in mean LS were 23.8% (HD1 group) and 29.3% (HD2 group). In HD groups, a strong negative association with developmental time has been found for both male and female mean and male maximum LSs; no association with growth rate was observed for female maximum LS. The female reproductive activity (fecundity) tended to decrease with subsequent days of eclosion. In HD groups, the levels of expression of all studied longevity-associated genes tended to increase with the timing of eclosion in males; no differences were observed in females. On the basis of findings obtained, it can be assumed that the development in conditions of larval overpopulation (if not too extended) could trigger hormetic response thereby extending the longevity. Further studies are, however, needed to confirm this assumption.
Collapse
|
158
|
Early post-conception maternal cortisol, children’s HPAA activity and DNA methylation profiles. J Dev Orig Health Dis 2018; 10:73-87. [DOI: 10.1017/s2040174418000880] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
AbstractThe hypothalamic–pituitary–adrenal axis (HPAA) plays a critical role in the functioning of all other biological systems. Thus, studying how the environment may influence its ontogeny is paramount to understanding developmental origins of health and disease. The early post-conceptional (EPC) period could be particularly important for the HPAA as the effects of exposures on organisms’ first cells can be transmitted through all cell lineages. We evaluate putative relationships between EPC maternal cortisol levels, a marker of physiologic stress, and their children’s pre-pubertal HPAA activity (n=22 dyads). Maternal first-morning urinary (FMU) cortisol, collected every-other-day during the first 8 weeks post-conception, was associated with children’s FMU cortisol collected daily around the start of the school year, a non-experimental challenge, as well as salivary cortisol responses to an experimental challenge (all Ps<0.05), with some sex-related differences. We investigated whether epigenetic mechanisms statistically mediated these links and, therefore, could provide cues as to possible biological pathways involved. EPC cortisol was associated with >5% change in children’s buccal epithelial cells’ DNA methylation for 867 sites, while children’s HPAA activity was associated with five CpG sites. Yet, no CpG sites were related to both, EPC cortisol and children’s HPAA activity. Thus, these epigenetic modifications did not statistically mediate the observed physiological links. Larger, prospective peri-conceptional cohort studies including frequent bio-specimen collection from mothers and children will be required to replicate our analyses and, if our results are confirmed, identify biological mechanisms mediating the statistical links observed between maternal EPC cortisol and children’s HPAA activity.
Collapse
|
159
|
Vaiserman A, Koliada A, Lushchak O. Developmental programming of aging trajectory. Ageing Res Rev 2018; 47:105-122. [PMID: 30059788 DOI: 10.1016/j.arr.2018.07.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/17/2018] [Accepted: 07/19/2018] [Indexed: 12/12/2022]
Abstract
There is accumulating evidence that aging phenotype and longevity may be developmentally programmed. Main mechanisms linking developmental conditions to later-life health outcomes include persistent changes in epigenetic regulation, (re)programming of major endocrine axes such as growth hormone/insulin-like growth factor axis and hypothalamic-pituitary-adrenal axis and also early-life immune maturation. Recently, evidence has also been generated on the role of telomere biology in developmental programming of aging trajectory. In addition, persisting changes of intestinal microbiota appears to be crucially involved in these processes. In this review, experimental and epidemiological evidence on the role of early-life conditions in programming of aging phenotypes are presented and mechanisms potentially underlying these associations are discussed.
Collapse
|
160
|
Gartstein MA, Skinner MK. Prenatal influences on temperament development: The role of environmental epigenetics. Dev Psychopathol 2018; 30:1269-1303. [PMID: 29229018 PMCID: PMC5997513 DOI: 10.1017/s0954579417001730] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
This review summarizes current knowledge and outlines future directions relevant to questions concerning environmental epigenetics and the processes that contribute to temperament development. Links between prenatal adversity, epigenetic programming, and early manifestations of temperament are important in their own right, also informing our understanding of biological foundations for social-emotional development. In addition, infant temperament attributes represent key etiological factors in the onset of developmental psychopathology, and studies elucidating their prenatal foundations expand our understanding of developmental origins of health and disease. Prenatal adversity can take many forms, and this overview is focused on the environmental effects of stress, toxicants, substance use/psychotropic medication, and nutrition. Dysregulation associated with attention-deficit/hyperactivity-disruptive disorders was noted in the context of maternal substance use and toxicant exposures during gestation, as well as stress. Although these links can be made based on the existing literature, currently few studies directly connect environmental influences, epigenetic programming, and changes in brain development/behavior. The chain of events starting with environmental inputs and resulting in alterations to gene expression, physiology, and behavior of the organism is driven by epigenetics. Epigenetics provides the molecular mechanism of how environmental factors impact development and subsequent health and disease, including early brain and temperament development.
Collapse
Affiliation(s)
- Maria A. Gartstein
- Department of Psychology, Washington State University, Pullman, WA-99164-4820, USA
| | - Michael K. Skinner
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA-99164-4236, USA
| |
Collapse
|
161
|
Song S, Hua C, Zhao F, Li M, Fu Q, Hooiveld GJEJ, Muller M, Li C, Zhou G. Purified Dietary Red and White Meat Proteins Show Beneficial Effects on Growth and Metabolism of Young Rats Compared to Casein and Soy Protein. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:9942-9951. [PMID: 30176144 DOI: 10.1021/acs.jafc.8b02521] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
This study compared the effects of casein, soy protein (SP), red (RMP), and white meat (WMP) proteins on growth and metabolism of young rats. Compared to casein, the ratio of daily feed intake to daily body weight gain of rats was not changed by meat protein but reduced by SP by 93.3% ( P < 0.05). Feeding RMP and WMP reduced the liver total cholesterol (TC) contents by 24.3% and 17.8%, respectively ( P < 0.05). Only RMP increased plasma HDL-cholesterol concentrations (by 12.7%, P < 0.05), whereas SP increased plasma triacylglycerol, TC, and LDL-cholesterol concentrations by 23.7%, 19.5%, and 61.5%, respectively ( P < 0.05). Plasma essential and total amino acid concentrations were increased by WMP (by 18.8% and 12.4%, P < 0.05) but reduced by SP (by 28.3% and 37.7%, P < 0.05). Twenty-five liver proteins were differentially expressed in response to different protein sources. Therefore, meat proteins were beneficial for growth and metabolism of young rats compared to casein and SP.
Collapse
Affiliation(s)
- Shangxin Song
- School of Food Science, Nanjing Xiaozhuang University , 3601 Hongjing Road , Nanjing 211171 , People's Republic of China
| | - Chun Hua
- School of Food Science, Nanjing Xiaozhuang University , 3601 Hongjing Road , Nanjing 211171 , People's Republic of China
| | - Fan Zhao
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Animal Products Processing, MOA; Jiang Synergetic Innovation Center of Meat Processing and Quality Control , Nanjing Agricultural University , Nanjing 210095 , People's Republic of China
| | - Mengjie Li
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Animal Products Processing, MOA; Jiang Synergetic Innovation Center of Meat Processing and Quality Control , Nanjing Agricultural University , Nanjing 210095 , People's Republic of China
| | - Qingquan Fu
- School of Food Science, Nanjing Xiaozhuang University , 3601 Hongjing Road , Nanjing 211171 , People's Republic of China
| | - Guido J E J Hooiveld
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition , Wageningen University , Wageningen 6700 HB , The Netherlands
| | - Michael Muller
- Norwich Medical School , University of East Anglia , Norwich NR4 2QR , England
| | - Chunbao Li
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Animal Products Processing, MOA; Jiang Synergetic Innovation Center of Meat Processing and Quality Control , Nanjing Agricultural University , Nanjing 210095 , People's Republic of China
| | - Guanghong Zhou
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Animal Products Processing, MOA; Jiang Synergetic Innovation Center of Meat Processing and Quality Control , Nanjing Agricultural University , Nanjing 210095 , People's Republic of China
| |
Collapse
|
162
|
Gliga AR, Engström K, Kippler M, Skröder H, Ahmed S, Vahter M, Raqib R, Broberg K. Prenatal arsenic exposure is associated with increased plasma IGFBP3 concentrations in 9-year-old children partly via changes in DNA methylation. Arch Toxicol 2018; 92:2487-2500. [PMID: 29947889 PMCID: PMC6063321 DOI: 10.1007/s00204-018-2239-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/04/2018] [Indexed: 01/20/2023]
Abstract
Exposure to inorganic arsenic (As), a carcinogen and epigenetic toxicant, has been associated with lower circulating levels of insulin-like growth factor 1 (IGF1) and impaired growth in children of pre-school age. The aim of this study was to assess the potential impact of exposure to As on IGF1 and insulin-like growth factor-binding protein 3 (IGFBP3) as well as DNA methylation changes in 9-year-old children. To this end, we studied 9-year-old children from a longitudinal mother-child cohort in rural Bangladesh (n = 551). Prenatal and concurrent exposure to As was assessed via concentrations in maternal urine at gestational week 8 and in child urine at 9 years, measured by HPLC-HG-ICPMS. Plasma IGF1 and IGFBP3 concentrations were quantified with immunoassays. DNA methylation was measured in blood mononuclear cells at 9 years in a sub-sample (n = 113) using the Infinium HumanMethylation450K BeadChip. In multivariable-adjusted linear regression models, prenatal As (natural log-transformed), but not children's concurrent urinary As, was positively associated with IGFBP3 concentrations (β = 76, 95% CI 19, 133). As concentrations were not associated with IGF1. DNA methylation analysis revealed CpGs associated with both prenatal As and IGFBP3. Mediation analysis suggested that methylation of 12 CpG sites for all children was mediator of effect for the association between prenatal As and IGFBP3. We also found differentially methylated regions, generally hypermethylated, that were associated with both prenatal As and IGFBP3. In all, our study revealed that prenatal exposure to As was positively associated with IGFBP3 concentrations in children at 9 years, independent of IGF1, and this association may, at least in part, be epigenetically mediated.
Collapse
Affiliation(s)
- Anda R Gliga
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Karin Engström
- Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
| | - Maria Kippler
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Helena Skröder
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Sultan Ahmed
- Division of Infectious Diseases, icddr,b, Dhaka, Bangladesh
| | - Marie Vahter
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Rubhana Raqib
- Division of Infectious Diseases, icddr,b, Dhaka, Bangladesh
| | - Karin Broberg
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
163
|
The effect of gestational period on the association between maternal prenatal salivary cortisol and birth weight: A systematic review and meta-analysis. Psychoneuroendocrinology 2018; 94:49-62. [PMID: 29754005 DOI: 10.1016/j.psyneuen.2018.04.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/21/2018] [Accepted: 04/23/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Studies exploring the relations between maternal stress and fetal development show an association between increased maternal stress and adverse birth outcomes. A frequently proposed mechanism linking maternal prenatal stress and adverse birth outcomes is heightened concentrations of maternal cortisol. To date, studies exploring this association have reported conflicting results because of the diverse approaches taken to measuring cortisol and the wide variety of possible birth outcomes explored. To add clarity to the growing body of literature, this systematic review and meta-analysis reports empirical findings on the association between maternal prenatal salivary cortisol and newborn birth weight. METHODS Searches for relevant papers published up until November 2017 were run in MEDLINE, EMBASE, PsycINFO, and CINAHL. Non-English language papers were included and experts were contacted when necessary. We included data from human observational studies that were designed or had an underlying intention to measure maternal prenatal salivary cortisol and newborn birth weight. We only included data from measurements of salivary cortisol to prevent rendering of the review unsuitable for meta-analysis. Two independent reviewers assessed study eligibility and quality. For every maternal-fetal dyad, an area under the curve with respect to ground (AUCg) of maternal cortisol was calculated to determine a Pearson's correlation coefficient with a continuous measure of newborn birth weight. Correlation coefficients were then pooled across all stages of gestation. To examine if there are critical gestational periods in which the fetus may be more susceptible to elevated concentration of maternal salivary cortisol, a meta-analysis was performed on separate correlations calculated from gestational trimesters. RESULTS Nine studies with a total of 1606 maternal-fetal dyads demonstrated a negative correlation between pooled maternal salivary cortisol and birth weight (-0.24, 95% CI -0.28 to -0.20), but there was a high degree of heterogeneity between studies (I2 = 88.9%). To investigate heterogeneity, subgroup analysis by trimester of the pooled correlation between salivary cortisol and birth weight was performed with the following correlations found: first trimester, -0.18 (95% CI -0.32 to -0.03, I2 = 97.3%); second trimester, -0.20 (95% CI -0.28 to -0.12, I2 = 98.3%); and third trimester, -0.30 (95% CI -0.33 to -0.26, I2 = 85.4%). DISCUSSION A consistently negative association was observed between maternal cortisol and infant birth weight. The review highlights specific gaps in the literature on the relationship between maternal prenatal salivary cortisol and newborn birth weight. Although a significant negative correlation was found, substantial heterogeneity of effects and the likelihood of publication bias exist. The third trimester was revealed as a possible critical gestational period for heightened maternal cortisol concentration to affect birth weight. Challenges faced in this body of research and recommendations for future research are discussed.
Collapse
|
164
|
Air pollution during pregnancy and placental adaptation in the levels of global DNA methylation. PLoS One 2018; 13:e0199772. [PMID: 29979694 PMCID: PMC6034814 DOI: 10.1371/journal.pone.0199772] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 06/13/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Health in early life is crucial for health later in life. Exposure to air pollution during embryonic and early-life development can result in placental epigenetic modification and foetus reprogramming, which can influence disease susceptibility in later life. Objectives: The aim of this paper was to investigate the placental adaptation in the level of global DNA methylation and differential gene expression in the methylation cycle in new-borns exposed to high fine particulate matter in the foetal stage. STUDY DESIGN This is a nested case-control study. We enrolled pregnant healthy women attending prenatal care clinics in Tehran, Iran, who were residents of selected polluted and unpolluted regions, before the 14th week of pregnancy. We calculated the regional background levels of particle mass- particles with aerodynamics diameter smaller than 2.5 μm (PM2.5) and 10 μm (PM10)-of two regions of interest. At the time of delivery, placental tissue was taken for gene expression and DNA methylation analyses. We also recorded birth outcomes (the new-born's sex, birth date, birth weight and length, head and chest circumference, gestational age, Apgar score, and level of neonatal care required). RESULTS As regards PM2.5 and PM10 concentrations in different time windows of pregnancy, there were significantly independent positive correlations between PM10 and PM2.5 in the first trimester of all subjects and placental global DNA methylation levels (p-value = 0.01, p-value = 0.03, respectively). The gene expression analysis showed there was significant correlation between S-adenosylmethionine expression and PM2.5 (p = 0.003) and PM10 levels in the first trimester (p = 0.03). CONCLUSION Our data showed prenatal exposures to air pollutants in the first trimester could influence placental adaptation by DNA methylation.
Collapse
|
165
|
Costello KR, Schones DE. Chromatin modifications in metabolic disease: Potential mediators of long-term disease risk. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2018; 10:e1416. [PMID: 29369528 PMCID: PMC6002879 DOI: 10.1002/wsbm.1416] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/30/2017] [Accepted: 12/08/2017] [Indexed: 12/29/2022]
Abstract
Metabolic diseases such as obesity and diabetes are complex diseases resulting from multiple genetic and environmental factors, such as diet and activity levels. These factors are well known contributors to the development of metabolic diseases. One manner by which environmental factors can influence metabolic disease progression is through modifications to chromatin. These modifications can lead to altered gene regulatory programs, which alters disease risk. Furthermore, there is evidence that parents exposed to environmental factors can influence the metabolic health of offspring, especially if exposures are during intrauterine growth periods. In this review, we outline the evidence that chromatin modifications are associated with metabolic diseases, including diabetes and obesity. We also consider evidence that these chromatin modifications can lead to long-term disease risk and contribute to disease risk for future generations. This article is categorized under: Biological Mechanisms > Metabolism Developmental Biology > Developmental Processes in Health and Disease Physiology > Organismal Responses to Environment.
Collapse
Affiliation(s)
- Kevin R. Costello
- Department of Diabetes Complications and MetabolismIrell & Manella Graduate School, City of HopeDuarteCalifornia
| | - Dustin E. Schones
- Department of Diabetes Complications and MetabolismIrell & Manella Graduate School, City of HopeDuarteCalifornia
| |
Collapse
|
166
|
Ramirez-Silva I, Rivera JA, Trejo-Valdivia B, Stein AD, Martorell R, Romieu I, Barraza-Villarreal A, Avila-Jiménez L, Ramakrishnan U. Relative Weight Gain Through Age 4 Years Is Associated with Increased Adiposity, and Higher Blood Pressure and Insulinemia at 4-5 Years of Age in Mexican Children. J Nutr 2018; 148:1135-1143. [PMID: 29924321 PMCID: PMC6669951 DOI: 10.1093/jn/nxy068] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/10/2017] [Accepted: 03/12/2018] [Indexed: 12/25/2022] Open
Abstract
Background Rapid early weight gain has been associated with increased risk of obesity and cardiometabolic alterations, but evidence in low and middle-income countries is inconclusive. Objective We evaluated the relation between relative weight gain from 1 to 48 mo with adiposity and cardiometabolic risk factors at 4-5 y of age, and determined if adiposity is a mediator for cardiometabolic alterations. Methods We studied 428 Mexican children with anthropometric and blood pressure (BP) information from birth to 5 y of age from POSGRAD (Prenatal Omega-3 fatty acid Supplementation and child GRowth And Development), of whom 334 provided measures of adiposity and cardiometabolic risk markers at 4 y. We estimated relative weight gain by means of conditional weight-for-height z scores for the age intervals 1-6, 6-12, 12-24, and 24-48 mo. Associations between relative weight gain and adiposity and cardiometabolic risk markers (lipid profile, triglycerides, insulin, glucose, and BP) were analyzed by multivariate multiple linear models and path analysis. Results A 1-unit increase in conditional weight-for-height z score within each age interval was positively associated with adiposity at 5 y, with coefficients of 0.43-0.89 for body mass index (BMI) z score, 1.08-3.65 mm for sum of skinfolds, and 1.21-3.87 cm for abdominal circumference (all P < 0.01). Positive associations were documented from ages 6 to 48 mo with systolic BP (coefficient ranges: 1.19-1.78 mm Hg; all P < 0.05) and from ages 12 to 48 mo with diastolic BP (1.28-0.94 mm Hg; P < 0.05) at 5 y. Conditional weight-for-height z scores at 12-24 and 24-48 mo of age were more strongly associated with adiposity and BP relative to younger ages. A unit increase in conditional weight-for-height z scores from 12 to 24 mo was associated with 14% higher insulin levels (P < 0.05) at 4 y. Path analyses documented that the associations of conditional weight gain with BP were mediated by BMI and sum of skinfolds. Conclusion Relative weight gain at most periods during the first 4 y of life was associated with greater adiposity and higher systolic and diastolic BP at 5 y. These associations with BP were mediated by adiposity. Relative weight gain from 12 to 24 mo was associated with increased serum insulin concentrations at 4 y, but there were no associations with lipid profiles or glucose concentration.
Collapse
Affiliation(s)
| | | | | | - Aryeh D Stein
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Reynaldo Martorell
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Isabelle Romieu
- Nutrition and Metabolism Section, International Agency for Research on Cancer, Lyon, France
| | | | | | - Usha Ramakrishnan
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA
| |
Collapse
|
167
|
Affiliation(s)
- Michael P Kelly
- Primary Care Unit, Department of Public Health and Primary Care, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| |
Collapse
|
168
|
Hartman RJG, Huisman SE, den Ruijter HM. Sex differences in cardiovascular epigenetics-a systematic review. Biol Sex Differ 2018; 9:19. [PMID: 29792221 PMCID: PMC5966883 DOI: 10.1186/s13293-018-0180-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/14/2018] [Indexed: 01/05/2023] Open
Abstract
Background Differences in cardiovascular diseases are evident in men and women throughout life and are mainly attributed to the presence of sex hormones and chromosomes. Epigenetic mechanisms drive the regulation of the biological processes that may lead to CVD and are possibly influenced by sex. In order to gain an overview of the status quo on sex differences in cardiovascular epigenetics, we performed a systematic review. Materials and methods A systematic search was performed on PubMed and Embase for studies mentioning cardiovascular disease, epigenetics, and anything related to sex differences. The search returned 3071 publications to be screened. Primary included publications focused on cardiovascular and epigenetics research. Subsequently, papers were assessed for including both sexes in their studies and checked for appropriate sex stratification of results. Results Two independent screeners identified 75 papers in the proper domains that had included both sexes. Only 17% (13 papers out of 75) of these publications stratified some of their data according to sex. All remaining papers focused on DNA methylation solely as an epigenetic mechanism. Of the excluded papers that included only one sex, 86% (24 out 28) studied males, while 14% (4 out of 28) studied females. Conclusion Our overview indicates that the majority of studies into cardiovascular epigenetics do not show their data stratified by sex, despite the well-known sex differences in CVD. All included and sex-stratified papers focus on DNA methylation, indicating that a lot of ground is still to gain regarding other epigenetic mechanisms, like chromatin architecture, and histone modifications. More attention to sex in epigenetic studies is warranted as such integration will advance our understanding of cardiovascular disease mechanisms in men and women. Electronic supplementary material The online version of this article (10.1186/s13293-018-0180-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Robin J G Hartman
- Laboratory of Experimental Cardiology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Sarah E Huisman
- Laboratory of Experimental Cardiology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
169
|
Wang P, Zhang C, Lv Q, Bao C, Sun H, Ma G, Fang Y, Yi Z, Cai W. Association of DNA methylation in BDNF with escitalopram treatment response in depressed Chinese Han patients. Eur J Clin Pharmacol 2018; 74:1011-1020. [PMID: 29748862 DOI: 10.1007/s00228-018-2463-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 04/11/2018] [Indexed: 01/16/2023]
Abstract
PURPOSE The neurotrophin brain-derived neurotrophic factor (BDNF) has been found to be associated with both the pathophysiology of depression and antidepressants response. Gene expression differences were partly mediated by SNP, which might be identified as a predictor of antidepressant response. In the present study, we attempt to identify whether DNA methylation, another factor known to affect gene transcription, might also predict antidepressant response. METHODS A total of 85 depressed Chinese Han patients were followed-up 8 weeks after initiating escitalopram treatment. Treatment response was assessed by changes in the Hamilton Depression Rating Scale-17 (HAMD-17) score. The Life Events Scale (LES) and the Childhood Trauma Questionnaire (CTQ) were utilized as the assessment of previous life stress. The bisulfate sequencing was used to assess DNA methylation. Four single nucleotide polymorphisms (SNPs) in the BDNF gene were genotyped using PCR-RFLP or PCR sequencing. RESULTS We identified a DNA methylation predictor (P = 0.006-0.036) and a DNA methylation by LES interaction predictor (OR = 1.442 [1.057-1.968], P = 0.021) of general antidepressant treatment response. Lower mean BDNF DNA methylation was associated with impaired antidepressant response. Furthermore, the present data indicated that age, life stress, and SNPs genotype might be likely related to DNA methylation status. Average DNA methylation of BDNF at baseline was significantly lower than that at endpoint after 8 weeks of escitalopram treatment, which was based only on a subset of cases (n = 44). CONCLUSIONS Our results suggest that BDNF DNA hypomethylation and its interaction with lower LES score might result in impaired antidepressant treatment response. The pharmacoepigenetic study could eventually help in finding epigenetic biomarkers of antidepressant response.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Rd, Shanghai, 201203, People's Republic of China
| | - Cuizhen Zhang
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Rd, Shanghai, 201203, People's Republic of China
| | - Qinyu Lv
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 Wanping Rd, Shanghai, 200030, People's Republic of China
| | - Chenxi Bao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 Wanping Rd, Shanghai, 200030, People's Republic of China
| | - Hong Sun
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Rd, Shanghai, 201203, People's Republic of China
| | - Guo Ma
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Rd, Shanghai, 201203, People's Republic of China
| | - Yiru Fang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 Wanping Rd, Shanghai, 200030, People's Republic of China
| | - Zhenghui Yi
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 Wanping Rd, Shanghai, 200030, People's Republic of China.
| | - Weimin Cai
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Rd, Shanghai, 201203, People's Republic of China.
| |
Collapse
|
170
|
Flöter VL, Lorenz AK, Kirchner B, Pfaffl MW, Bauersachs S, Ulbrich SE. Impact of preimplantational oral low-dose estradiol-17β exposure on the endometrium: The role of miRNA. Mol Reprod Dev 2018. [DOI: 10.1002/mrd.22975] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Veronika L. Flöter
- ETH Zurich, Animal Physiology; Institute of Agricultural Sciences; Zürich Switzerland
- Department of Animal Physiology and Immunology, School of Life Sciences, Life Science Center Weihenstephan; Technical University Munich; Freising-Weihenstephan Germany
| | - Anne-Kathrin Lorenz
- ETH Zurich, Animal Physiology; Institute of Agricultural Sciences; Zürich Switzerland
- Department of Animal Physiology and Immunology, School of Life Sciences, Life Science Center Weihenstephan; Technical University Munich; Freising-Weihenstephan Germany
| | - Benedikt Kirchner
- Department of Animal Physiology and Immunology, School of Life Sciences, Life Science Center Weihenstephan; Technical University Munich; Freising-Weihenstephan Germany
| | - Michael W. Pfaffl
- Department of Animal Physiology and Immunology, School of Life Sciences, Life Science Center Weihenstephan; Technical University Munich; Freising-Weihenstephan Germany
| | - Stefan Bauersachs
- ETH Zurich, Animal Physiology; Institute of Agricultural Sciences; Zürich Switzerland
| | - Susanne E. Ulbrich
- ETH Zurich, Animal Physiology; Institute of Agricultural Sciences; Zürich Switzerland
| |
Collapse
|
171
|
Vaiserman AM. Birth weight predicts aging trajectory: A hypothesis. Mech Ageing Dev 2018; 173:61-70. [PMID: 29626501 DOI: 10.1016/j.mad.2018.04.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/10/2018] [Accepted: 04/03/2018] [Indexed: 01/26/2023]
Abstract
Increasing evidence suggests that risk for age-related disease and longevity can be programmed early in life. In human populations, convincing evidence has been accumulated indicating that intrauterine growth restriction (IUGR) resulting in low birth weight (<2.5 kg) followed by postnatal catch-up growth is associated with various aspects of metabolic syndrome, type 2 diabetes and cardiovascular disease in adulthood. Fetal macrosomia (birth weight > 4.5 kg), by contrast, is associated with high risk of non-diabetic obesity and cancers in later life. Developmental modification of epigenetic patterns is considered to be a central mechanism in determining such developmentally programmed phenotypes. Growth hormone/insulin-like growth factor (GH/IGF) axis is likely a key driver of these processes. In this review, evidence is discussed that suggests that different aging trajectories can be realized depending on developmentally programmed life-course dynamics of IGF-1. In this hypothetical scenario, IUGR-induced deficit of IGF-1 causes "diabetic" aging trajectory associated with various metabolic disorders in adulthood, while fetal macrosomia-induced excessive levels of IGF-1 lead to "cancerous" aging trajectory. If the above reasoning is correct, then both low and high birth weights are predictors of short life expectancy, while the normal birth weight is a predictor of "normal" aging and maximum longevity.
Collapse
Affiliation(s)
- Alexander M Vaiserman
- Institute of Gerontology NAMS of Ukraine, Vyshgorodskaya st. 67, Kiev 04114, Ukraine.
| |
Collapse
|
172
|
Neelankal John A, Jiang FX. An overview of type 2 diabetes and importance of vitamin D3-vitamin D receptor interaction in pancreatic β-cells. J Diabetes Complications 2018; 32:429-443. [PMID: 29422234 DOI: 10.1016/j.jdiacomp.2017.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 12/03/2017] [Accepted: 12/07/2017] [Indexed: 02/07/2023]
Abstract
One significant health issue that plagues contemporary society is that of Type 2 diabetes (T2D). This disease is characterised by higher-than-average blood glucose levels as a result of a combination of insulin resistance and insufficient insulin secretions from the β-cells of pancreatic islets of Langerhans. Previous developmental research into the pancreas has identified how early precursor genes of pancreatic β-cells, such as Cpal, Ngn3, NeuroD, Ptf1a, and cMyc, play an essential role in the differentiation of these cells. Furthermore, β-cell molecular characterization has also revealed the specific role of β-cell-markers, such as Glut2, MafA, Ins1, Ins2, and Pdx1 in insulin expression. The expression of these genes appears to be suppressed in the T2D β-cells, along with the reappearance of the early endocrine marker genes. Glucose transporters transport glucose into β-cells, thereby controlling insulin release during hyperglycaemia. This stimulates glycolysis through rises in intracellular calcium (a process enhanced by vitamin D) (Norman et al., 1980), activating 2 of 4 proteinases. The rise in calcium activates half of pancreatic β-cell proinsulinases, thus releasing free insulin from granules. The synthesis of ATP from glucose by glycolysis, Krebs cycle and oxidative phosphorylation plays a role in insulin release. Some studies have found that the β-cells contain high levels of the vitamin D receptor; however, the role that this plays in maintaining the maturity of the β-cells remains unknown. Further research is required to develop a more in-depth understanding of the role VDR plays in β-cell function and the processes by which the beta cell function is preserved.
Collapse
Affiliation(s)
- Abraham Neelankal John
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, Western Australia, Australia; School of Medicine and Pharmacology, University of Western Australia, Carwley, Western Australia, Australia
| | - Fang-Xu Jiang
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, Western Australia, Australia; School of Medicine and Pharmacology, University of Western Australia, Carwley, Western Australia, Australia.
| |
Collapse
|
173
|
Taylor RM, Smith R, Collins CE, Mossman D, Wong-Brown MW, Chan EC, Evans TJ, Attia JR, Smith T, Butler T, Hure AJ. Methyl-Donor and Cofactor Nutrient Intakes in the First 2-3 Years and Global DNA Methylation at Age 4: A Prospective Cohort Study. Nutrients 2018; 10:E273. [PMID: 29495543 PMCID: PMC5872691 DOI: 10.3390/nu10030273] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/13/2018] [Accepted: 02/13/2018] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND During the early postnatal period, the impact of nutrition on DNA methylation has not been well studied in humans. The aim was to quantify the relationship between one-carbon metabolism nutrient intake during the first three years of life and global DNA methylation levels at four years. DESIGN Childhood dietary intake was assessed using infant feeding questionnaires, food frequency questionnaires, 4-day weighed food records and 24-h food records. The dietary records were used to estimate the intake of methionine, folate, vitamins B2, B6 and B12 and choline. The accumulative nutrient intake specific rank from three months to three years of age was used for analysis. Global DNA methylation (%5-methyl cytosines (%5-mC)) was measured in buccal cells at four years of age, using an enzyme-linked immunosorbent assay (ELISA) commercial kit. Linear regression models were used to quantify the statistical relationships. RESULTS Data were collected from 73 children recruited from the Women and their Children's Health (WATCH) study. No association was found between one-carbon metabolism nutrient intake and global DNA methylation levels (P > 0.05). Global DNA methylation levels in males were significantly higher than in females (median %5-mC: 1.82 vs. 1.03, males and females respectively, (P < 0.05)). CONCLUSION No association was found between the intake of one-carbon metabolism nutrients during the early postnatal period and global DNA methylation levels at age four years. Higher global DNA methylation levels in males warrants further investigation.
Collapse
Affiliation(s)
- Rachael M. Taylor
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW 2308, Australia; (R.S.); (C.E.C.); (T.S.); (T.B.)
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, Callaghan, NSW 2308, Australia; (E.-C.C.); (J.R.A.); (A.J.H.)
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia; (D.M.); (M.W.W.-B.); (T.-J.E.)
| | - Roger Smith
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW 2308, Australia; (R.S.); (C.E.C.); (T.S.); (T.B.)
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, Callaghan, NSW 2308, Australia; (E.-C.C.); (J.R.A.); (A.J.H.)
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia; (D.M.); (M.W.W.-B.); (T.-J.E.)
| | - Clare E. Collins
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW 2308, Australia; (R.S.); (C.E.C.); (T.S.); (T.B.)
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, Callaghan, NSW 2308, Australia; (E.-C.C.); (J.R.A.); (A.J.H.)
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia; (D.M.); (M.W.W.-B.); (T.-J.E.)
- Faculty of Health and Medicine, School of Health Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
- Priority Research Centre in Physical Activity and Nutrition, University of Newcastle, Callaghan, NSW 2308, Australia
| | - David Mossman
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia; (D.M.); (M.W.W.-B.); (T.-J.E.)
- Department of Molecular Medicine, NSW Health Pathology, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia
| | - Michelle W. Wong-Brown
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia; (D.M.); (M.W.W.-B.); (T.-J.E.)
- Faculty of Health, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Eng-Cheng Chan
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, Callaghan, NSW 2308, Australia; (E.-C.C.); (J.R.A.); (A.J.H.)
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia; (D.M.); (M.W.W.-B.); (T.-J.E.)
| | - Tiffany-Jane Evans
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia; (D.M.); (M.W.W.-B.); (T.-J.E.)
- Clinical Research Design IT and Statistical Support (CReDITSS) Unit, Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - John R. Attia
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, Callaghan, NSW 2308, Australia; (E.-C.C.); (J.R.A.); (A.J.H.)
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia; (D.M.); (M.W.W.-B.); (T.-J.E.)
- Clinical Research Design IT and Statistical Support (CReDITSS) Unit, Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - Tenele Smith
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW 2308, Australia; (R.S.); (C.E.C.); (T.S.); (T.B.)
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, Callaghan, NSW 2308, Australia; (E.-C.C.); (J.R.A.); (A.J.H.)
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia; (D.M.); (M.W.W.-B.); (T.-J.E.)
| | - Trent Butler
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW 2308, Australia; (R.S.); (C.E.C.); (T.S.); (T.B.)
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, Callaghan, NSW 2308, Australia; (E.-C.C.); (J.R.A.); (A.J.H.)
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia; (D.M.); (M.W.W.-B.); (T.-J.E.)
| | - Alexis J. Hure
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, Callaghan, NSW 2308, Australia; (E.-C.C.); (J.R.A.); (A.J.H.)
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia; (D.M.); (M.W.W.-B.); (T.-J.E.)
- Priority Research Centre for Generational, Health and Ageing, University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
174
|
Ventura-Clapier R, Dworatzek E, Seeland U, Kararigas G, Arnal JF, Brunelleschi S, Carpenter TC, Erdmann J, Franconi F, Giannetta E, Glezerman M, Hofmann SM, Junien C, Katai M, Kublickiene K, König IR, Majdic G, Malorni W, Mieth C, Miller VM, Reynolds RM, Shimokawa H, Tannenbaum C, D'Ursi AM, Regitz-Zagrosek V. Sex in basic research: concepts in the cardiovascular field. Cardiovasc Res 2018; 113:711-724. [PMID: 28472454 DOI: 10.1093/cvr/cvx066] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 05/01/2017] [Indexed: 01/01/2023] Open
Abstract
Women and men, female and male animals and cells are biologically different, and acknowledgement of this fact is critical to advancing medicine. However, incorporating concepts of sex-specific analysis in basic research is largely neglected, introducing bias into translational findings, clinical concepts and drug development. Research funding agencies recently approached these issues but implementation of policy changes in the scientific community is still limited, probably due to deficits in concepts, knowledge and proper methodology. This expert review is based on the EUGenMed project (www.eugenmed.eu) developing a roadmap for implementing sex and gender in biomedical and health research. For sake of clarity and conciseness, examples are mainly taken from the cardiovascular field that may serve as a paradigm for others, since a significant amount of knowledge how sex and oestrogen determine the manifestation of many cardiovascular diseases (CVD) has been accumulated. As main concepts for implementation of sex in basic research, the study of primary cell and animals of both sexes, the study of the influence of genetic vs. hormonal factors and the analysis of sex chromosomes and sex specific statistics in genome wide association studies (GWAS) are discussed. The review also discusses methodological issues, and analyses strength, weaknesses, opportunities and threats in implementing sex-sensitive aspects into basic research.
Collapse
Affiliation(s)
- Renée Ventura-Clapier
- Signalisation et Physiopathologie Cardiovasculaire UMR-S 1180, Inserm, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Elke Dworatzek
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin Berlin, 10115 Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| | - Ute Seeland
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin Berlin, 10115 Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| | - Georgios Kararigas
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin Berlin, 10115 Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| | - Jean-Francois Arnal
- Faculté Médecine Toulouse-Rangueil, Université de Toulouse, Toulouse, France
| | - Sandra Brunelleschi
- Department of Health Sciences, School of Medicine, University of Eastern Piedmont, Novara, Italy
| | - Thomas C Carpenter
- College of Medicine and Veterinary Medicine, University of Edinburgh, EH16 4TJ Edinburgh, UK
| | - Jeanette Erdmann
- Institut für Kardiogenetik, Universität zu Lübeck, 23562 Lübeck, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany
| | - Flavia Franconi
- Department of Biomedical Science, University of Sassari,Sassari, Italy
| | - Elisa Giannetta
- Ricercatore TD in Endocrinologia, Dipartimento di Medicina Sperimentale, Sezione di Fisiopatologia Medica, Sapienza University of Rome, Roma, Italy
| | - Marek Glezerman
- International Society for Gender Medicine, Research Center for Medicine, Rabin Medical Center, and Tel Aviv University, Israel
| | - Susanna M Hofmann
- Medizinische Klinik und Poliklinik IV, Klinikum der LMU München, Munich 80336, Germany; Institute for Diabetes and Regeneration, Helmholtz Center Munich, Germany; German Center for Diabetes Research (DZD) München-Neuherberg, Germany
| | - Claudine Junien
- BDR Biologie du Développement et Reproduction Developmental Biology and Reproduction UMR, INRA, France
| | - Miyuki Katai
- Section of Gender Medicine, Department of General Medicine, Tokyo Women's Medical University, 162-8666 Tokyo, Japan
| | - Karolina Kublickiene
- Centre for Gender Medicine and Departments of Obstetrics and Gynecology and Renal Medicine, Karolinska Institutet, 14186 Stockholm, Sweden
| | - Inke R König
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany.,Institut für Medizinische Biometrie und Statistik, Universität zu Lübeck, 235620 Lübeck, Germany
| | - Gregor Majdic
- Institute for Preclinical Sciences, Veterinary Faculty, University of Ljubljana & Institute of Physiology, Medical Faculty, University of Maribor, Maribor, Slovenia
| | - Walter Malorni
- National Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Roma, Italy
| | - Christin Mieth
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtz-Gemeinschaft (MDC), Berlin, Germany
| | | | - Rebecca M Reynolds
- Center for Cardiovascular Science, Queen's Medical Research Institute, EH16 4TJ Edinburgh, UK
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Cara Tannenbaum
- Institute of Gender and Health, Canadian Institutes of Health Research (CIHR), Canada
| | - Anna Maria D'Ursi
- Medicinal Chemistry DIFARMA, Università di Salerno, 84084 Fisciano, Italy
| | - Vera Regitz-Zagrosek
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin Berlin, 10115 Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| |
Collapse
|
175
|
Li SC, Kabeer MH. Spatiotemporal switching signals for cancer stem cell activation in pediatric origins of adulthood cancer: Towards a watch-and-wait lifetime strategy for cancer treatment. World J Stem Cells 2018; 10:15-22. [PMID: 29531638 PMCID: PMC5840533 DOI: 10.4252/wjsc.v10.i2.15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/25/2018] [Accepted: 02/24/2018] [Indexed: 02/06/2023] Open
Abstract
Pediatric origin of cancer stem cell hypothesis holds great promise and potential in adult cancer treatment, however; the road to innovation is full of obstacles as there are plenty of questions left unanswered. First, the key question is to characterize the nature of such stem cells (concept). Second, the quantitative imaging of pediatric stem cells should be implemented (technology). Conceptually, pediatric stem cell origins of adult cancer are based on the notion that plasticity in early life developmental programming evolves local environments to cancer. Technologically, such imaging in children is lacking as all imaging is designed for adult patients. We postulate that the need for quantitative imaging to measure space-time changes of plasticity in early life developmental programming in children may trigger research and development of the imaging technology. Such quantitative imaging of pediatric origin of adulthood cancer will help develop a spatiotemporal monitoring system to determine cancer initiation and progression. Clinical validation of such speculative hypothesis-that cancer originates in a pediatric environment-will help implement a wait-and-watch strategy for cancer treatment.
Collapse
Affiliation(s)
- Shengwen Calvin Li
- Neuro-oncology and Stem Cell Research Laboratory, Children's Hospital of Orange County, Department of Neurology, University of California-Irvine School of Medicine, Orange, CA 92868-3874, United States
| | - Mustafa H Kabeer
- Children's Hospital of Orange County, Department of Surgery, University of California-Irvine School of Medicine, Orange, CA 92868-3874, United States
| |
Collapse
|
176
|
Embryotoxic cytokines—Potential roles in embryo loss and fetal programming. J Reprod Immunol 2018; 125:80-88. [DOI: 10.1016/j.jri.2017.12.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/13/2017] [Accepted: 12/21/2017] [Indexed: 12/22/2022]
|
177
|
Brown HM, Green ES, Tan TCY, Gonzalez MB, Rumbold AR, Hull ML, Norman RJ, Packer NH, Robertson SA, Thompson JG. Periconception onset diabetes is associated with embryopathy and fetal growth retardation, reproductive tract hyperglycosylation and impaired immune adaptation to pregnancy. Sci Rep 2018; 8:2114. [PMID: 29391475 PMCID: PMC5794861 DOI: 10.1038/s41598-018-19263-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 12/22/2017] [Indexed: 01/04/2023] Open
Abstract
Diabetes has been linked with impaired fertility but the underlying mechanisms are not well defined. Here we use a streptozotocin-induced diabetes mouse model to investigate the cellular and biochemical changes in conceptus and maternal tissues that accompany hyperglycaemia. We report that streptozotocin treatment before conception induces profound intra-cellular protein β-O-glycosylation (O-GlcNAc) in the oviduct and uterine epithelium, prominent in early pregnancy. Diabetic mice have impaired blastocyst development and reduced embryo implantation rates, and delayed mid-gestation growth and development. Peri-conception changes are accompanied by increased expression of pro-inflammatory cytokine Trail, and a trend towards increased Il1a, Tnf and Ifng in the uterus, and changes in local T-cell dynamics that skew the adaptive immune response to pregnancy, resulting in 60% fewer anti-inflammatory regulatory T-cells within the uterus-draining lymph nodes. Activation of the heat shock chaperones, a mechanism for stress deflection, was evident in the reproductive tract. Additionally, we show that the embryo exhibits elevated hyper-O-GlcNAcylation of both cytoplasmic and nuclear proteins, associated with activation of DNA damage (ɣH2AX) pathways. These results advance understanding of the impact of peri-conception diabetes, and provide a foundation for designing interventions to support healthy conception without propagation of disease legacy to offspring.
Collapse
Affiliation(s)
- Hannah M Brown
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia. .,Australian Research Council (ARC) Centre for Nanoscale Biophotonics, University of Adelaide, Adelaide, Australia.
| | - Ella S Green
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Tiffany C Y Tan
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Macarena B Gonzalez
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Alice R Rumbold
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - M Louise Hull
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Fertility SA, Adelaide, Australia
| | - Robert J Norman
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Fertility SA, Adelaide, Australia
| | - Nicolle H Packer
- ARC Centre for Nanoscale Biophotonics, Department of Molecular Sciences, Macquarie University, Sydney, Australia
| | - Sarah A Robertson
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Jeremy G Thompson
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Australian Research Council (ARC) Centre for Nanoscale Biophotonics, University of Adelaide, Adelaide, Australia
| |
Collapse
|
178
|
Osher D, Cantor P, Berg J, Steyer L, Rose T. Drivers of human development: How relationships and context shape learning and development1. APPLIED DEVELOPMENTAL SCIENCE 2018. [DOI: 10.1080/10888691.2017.1398650] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
| | - Pamela Cantor
- Turnaround for Children
- Harvard Graduate School of Education
| | | | | | - Todd Rose
- Harvard Graduate School of Education
- The Center for Individual Opportunity
| |
Collapse
|
179
|
McGowan PO, Matthews SG. Prenatal Stress, Glucocorticoids, and Developmental Programming of the Stress Response. Endocrinology 2018; 159:69-82. [PMID: 29136116 DOI: 10.1210/en.2017-00896] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 11/06/2017] [Indexed: 01/06/2023]
Abstract
The early environment has a major impact on the developing embryo, fetus, and infant. Parental adversity (maternal and paternal) and glucocorticoid exposure before conception and during pregnancy have profound effects on the development and subsequent function of the hypothalamic-pituitary-adrenal axis and related behaviors. These effects are species-, sex-, and age-specific and depend on the timing and duration of exposure. The impact of these early exposures can extend across multiple generations, via both the maternal and paternal lineage, and recent studies have begun to determine the mechanisms by which this occurs. Improved knowledge of the mechanisms by which adversity and glucocorticoids program stress systems will allow development of strategies to ameliorate and/or reverse these long-term effects.
Collapse
Affiliation(s)
- Patrick O McGowan
- Department of Biological Sciences, University of Toronto, Toronto, Ontario, Canada
- Center for Environmental Epigenetics and Development, University of Toronto, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Stephen G Matthews
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Obstetrics & Gynaecology, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| |
Collapse
|
180
|
Vaiserman AM, Koliada AK. Early-life adversity and long-term neurobehavioral outcomes: epigenome as a bridge? Hum Genomics 2017; 11:34. [PMID: 29246185 PMCID: PMC5732459 DOI: 10.1186/s40246-017-0129-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 12/04/2017] [Indexed: 12/12/2022] Open
Abstract
Accumulating evidence suggests that adversities at critical periods in early life, both pre- and postnatal, can lead to neuroendocrine perturbations, including hypothalamic-pituitary-adrenal axis dysregulation and inflammation persisting up to adulthood. This process, commonly referred to as biological embedding, may cause abnormal cognitive and behavioral functioning, including impaired learning, memory, and depressive- and anxiety-like behaviors, as well as neuropsychiatric outcomes in later life. Currently, the regulation of gene activity by epigenetic mechanisms is suggested to be a key player in mediating the link between adverse early-life events and adult neurobehavioral outcomes. Role of particular genes, including those encoding glucocorticoid receptor, brain-derived neurotrophic factor, as well as arginine vasopressin and corticotropin-releasing factor, has been demonstrated in triggering early adversity-associated pathological conditions. This review is focused on the results from human studies highlighting the causal role of epigenetic mechanisms in mediating the link between the adversity during early development, from prenatal stages through infancy, and adult neuropsychiatric outcomes. The modulation of epigenetic pathways involved in biological embedding may provide promising direction toward novel therapeutic strategies against neurological and cognitive dysfunctions in adult life.
Collapse
Affiliation(s)
- Alexander M Vaiserman
- Laboratory of Epigenetics, Institute of Gerontology, Vyshgorodskaya st. 67, Kiev, 04114, Ukraine.
| | - Alexander K Koliada
- Laboratory of Epigenetics, Institute of Gerontology, Vyshgorodskaya st. 67, Kiev, 04114, Ukraine
| |
Collapse
|
181
|
Grier A, Qiu X, Bandyopadhyay S, Holden-Wiltse J, Kessler HA, Gill AL, Hamilton B, Huyck H, Misra S, Mariani TJ, Ryan RM, Scholer L, Scheible KM, Lee YH, Caserta MT, Pryhuber GS, Gill SR. Impact of prematurity and nutrition on the developing gut microbiome and preterm infant growth. MICROBIOME 2017; 5:158. [PMID: 29228972 PMCID: PMC5725645 DOI: 10.1186/s40168-017-0377-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 11/23/2017] [Indexed: 05/17/2023]
Abstract
BACKGROUND Identification of factors that influence the neonatal gut microbiome is urgently needed to guide clinical practices that support growth of healthy preterm infants. Here, we examined the influence of nutrition and common practices on the gut microbiota and growth in a cohort of preterm infants. RESULTS With weekly gut microbiota samples spanning postmenstrual age (PMA) 24 to 46 weeks, we developed two models to test associations between the microbiota, nutrition and growth: a categorical model with three successive microbiota phases (P1, P2, and P3) and a model with two periods (early and late PMA) defined by microbiota composition and PMA, respectively. The more significant associations with phase led us to use a phase-based framework for the majority of our analyses. Phase transitions were characterized by rapid shifts in the microbiota, with transition out of P1 occurring nearly simultaneously with the change from meconium to normal stool. The rate of phase progression was positively associated with gestational age at birth, and delayed transition to a P3 microbiota was associated with growth failure. We found distinct bacterial metabolic functions in P1-3 and significant associations between nutrition, microbiota phase, and infant growth. CONCLUSION The phase-dependent impact of nutrition on infant growth along with phase-specific metabolic functions suggests a pioneering potential for improving growth outcomes by tailoring nutrient intake to microbiota phase.
Collapse
MESH Headings
- Bacteria/classification
- Bacteria/genetics
- Bacteria/isolation & purification
- Breast Feeding
- Cohort Studies
- DNA, Bacterial
- Feces/microbiology
- Female
- Gastrointestinal Microbiome
- Gestational Age
- Humans
- Infant
- Infant Health
- Infant, Newborn
- Infant, Premature/growth & development
- Infant, Premature/physiology
- Infant, Premature, Diseases/diet therapy
- Infant, Premature, Diseases/prevention & control
- Male
- Meconium/microbiology
- Nutritional Status
- RNA, Ribosomal, 16S
- Sequence Analysis, DNA
Collapse
Affiliation(s)
- Alex Grier
- Genomics Research Center, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Sanjukta Bandyopadhyay
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Jeanne Holden-Wiltse
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Haeja A Kessler
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Ann L Gill
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Brooke Hamilton
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Heidie Huyck
- Division of Neonatology, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Sara Misra
- Division of Neonatology, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Thomas J Mariani
- Division of Neonatology, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Rita M Ryan
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Lori Scholer
- Division of Neonatology, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Kristin M Scheible
- Division of Neonatology, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Yi-Horng Lee
- Division of Pediatric Surgery, Department of Surgery, Robert Wood Johnson University Hospital, New Brunswick, NJ, USA
| | - Mary T Caserta
- Division of Infectious Disease, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Gloria S Pryhuber
- Division of Neonatology, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Steven R Gill
- Genomics Research Center, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
| |
Collapse
|
182
|
Metwally M, Eslam M, George J. Genetic and Epigenetic Associations of NAFLD: Focus on Clinical Decision Making. CURRENT HEPATOLOGY REPORTS 2017; 16:335-345. [DOI: 10.1007/s11901-017-0372-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
|
183
|
Xia B, Gerstin E, Schones DE, Huang W, Steven de Belle J. Transgenerational programming of longevity through E(z)-mediated histone H3K27 trimethylation in Drosophila. Aging (Albany NY) 2017; 8:2988-3008. [PMID: 27889707 PMCID: PMC5191882 DOI: 10.18632/aging.101107] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 11/05/2016] [Indexed: 12/19/2022]
Abstract
Transgenerational effects on health and development of early-life nutrition have gained increased attention recently. However, the underlying mechanisms of transgenerational transmission are only starting to emerge, with epigenetics as perhaps the most important mechanism. We recently reported the first animal model to study transgenerational programming of longevity after early-life dietary manipulations, enabling investigations to identify underlying epigenetic mechanisms. We report here that post-eclosion dietary manipulation (PDM) with a low-protein (LP) diet upregulates the protein level of E(z), an H3K27 specific methyltransferase, leading to higher levels of H3K27 trimethylation (H3K27me3). This PDM-mediated change in H3K27me3 corresponded with a shortened longevity of F0 flies as well as their F2 offspring. Specific RNAi-mediated post-eclosion knockdown of E(z) or pharmacological inhibition of its enzymatic function with EPZ-6438 in the F0 parents improved longevity while rendering H3K27me3 low across generations. Importantly, addition of EPZ-6438 to the LP diet fully alleviated the longevity-reducing effect of the LP PDM, supporting the increased level of E(z)-dependent H3K27me3 as the primary cause and immediate early-life period as the critical time to program longevity through epigenetic regulation. These observations establish E(z)-mediated H3K27me3 as one epigenetic mechanism underlying nutritional programming of longevity and support the use of EPZ-6438 to extend lifespan.
Collapse
Affiliation(s)
- Brian Xia
- Canyon Crest Academy, San Diego, CA 92130, USA.,Department of Diabetes Complications and Metabolism, City of Hope, Duarte, CA 91010, USA.,Department of Biology, Dart Neuroscience LLC, San Diego, CA 92131, USA
| | - Ed Gerstin
- Canyon Crest Academy, San Diego, CA 92130, USA
| | - Dustin E Schones
- Department of Diabetes Complications and Metabolism, City of Hope, Duarte, CA 91010, USA
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, City of Hope, Duarte, CA 91010, USA
| | - J Steven de Belle
- Department of Biology, Dart Neuroscience LLC, San Diego, CA 92131, USA
| |
Collapse
|
184
|
|
185
|
Donkor HM, Grundt JH, Júlíusson PB, Eide GE, Hurum J, Bjerknes R, Markestad T. Social and somatic determinants of underweight, overweight and obesity at 5 years of age: a Norwegian regional cohort study. BMJ Open 2017; 7:e014548. [PMID: 28821510 PMCID: PMC5724122 DOI: 10.1136/bmjopen-2016-014548] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE To identify associations between the weight groups underweight (UW), overweight (OW) and obesity (OB) at 5 years of age and exposures related to pregnancy, anthropometric measures at birth, sociodemographic factors, and family health, anthropometric measures and habits. DESIGN Regional cohort study. SETTING Oppland County, Norway. METHODS Pregnancy data were obtained from a prospective perinatal register for children born in the county, and weight and height were measured by midwives at birth and by public health nurses at 5 years. Other information was obtained from questionnaires completed by parents. PARTICIPANTS Of 1895 eligible children, current weight and height were obtained for all, weight and length at birth and information from parents for 1119 (59%) and pregnancy register data for 749 (40%) of the children. The significance of potential explanatory variables from descriptive statistics was tested in multinomial logistic regression analysis. RESULTS The prevalence of UW, OW and OB among participants was 7.8%, 10.6% and 3.5%, respectively. UW was associated with anthropometric measures at birth and those of parents, but not with sociodemographic or behavioural characteristics. OW and OB were associated with anthropometric measures of parents and siblings and with a variety of unfavourable social characteristics, lack of prolonged breast feeding, sedentary behaviour and dental caries, but not with current dietary habits. After adjustments, OW and OB were marginally related to birth parameters and diet and unrelated to physical activity, but significantly related to parental body mass index, low parental education and maternal smoking. CONCLUSION The strong associations between sociodemographic and behavioural factors and OW and OB, but not with UW, may suggest that environmental factors are major contributing causes of OW and particularly OB at 5 years. These results may be helpful in targeting preventive measures against OW and OB.
Collapse
Affiliation(s)
- Hilde Mjell Donkor
- Department of Paediatrics, Innlandet Hospital Trust, Lillehammer, Norway
| | | | - Pétur Benedikt Júlíusson
- Department of Clinical Science, Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
| | - Geir Egil Eide
- Centre for Clinical Research, Haukeland University Hospital, Bergen, Norway
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Jørgen Hurum
- Department of Paediatrics, Innlandet Hospital Trust, Lillehammer, Norway
| | - Robert Bjerknes
- Department of Clinical Science, Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
| | - Trond Markestad
- Department of Clinical Science, Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
- Department of Research, Innlandet Hospital Trust, Lillehammer, Norway
| |
Collapse
|
186
|
Berti C, Agostoni C, Davanzo R, Hyppönen E, Isolauri E, Meltzer HM, Steegers-Theunissen RPM, Cetin I. Early-life nutritional exposures and lifelong health: immediate and long-lasting impacts of probiotics, vitamin D, and breastfeeding. Nutr Rev 2017; 75:83-97. [PMID: 28130504 DOI: 10.1093/nutrit/nuw056] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/16/2016] [Indexed: 12/21/2022] Open
Abstract
Pregnancy and infancy comprise the most critical stages for conditioning an individual's health, with a number of implications for subsequent risks of morbidity, mortality, and reproductive health. Nutrition may influence both the overall pregnancy outcome and the growth trajectory and immune system of the fetus and infant, with short- and long-term effects on the health of the offspring. Within this context, leading experts at Expo Milano 2015 in Milan, Italy, discussed up-to-date knowledge while providing suggestions and challenges before, during, and after pregnancy. This narrative review summarizes the key issues raised by the experts concerning the interplay between the nutritional environment from conception to early infancy and the offspring's immediate and lifelong health, with a particular focus on epigenetic mechanisms, probiotics, vitamin D, and breastfeeding. Taken together, the findings strengthen the awareness that nutritional exposures occurring from preconception to the postnatal period may be strong determinants of the offspring's health and may provide supportive evidence for current nutritional recommendations and guidelines for pregnant women and infants. Critical topics to be addressed in future research and translated into recommendations of public health relevance are also highlighted.
Collapse
Affiliation(s)
- Cristiana Berti
- Department of Biomedical and Clinical Sciences, School of Medicine and Center for Fetal Research Giorgio Pardi, University of Milan, Milan, Italy
| | - Carlo Agostoni
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Riccardo Davanzo
- Division of Neonatology and NICU, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Elina Hyppönen
- Centre for Population Health Research, Sansom Institute of Health Research and School of Health Sciences, University of South Australia, Adelaide, Australia.,South Australian Health and Medical Research Institute, Adelaide, Australia.,Population, Policy and Practice Programme, University College London Institute of Child Health, London, UK
| | - Erika Isolauri
- Department of Paediatrics, Turku University Hospital and University of Turku, Turku, Finland
| | - Helle M Meltzer
- Norwegian Institute of Public Health, Domain of Infection Control and Environmental Health, Oslo, Norway
| | - Régine P M Steegers-Theunissen
- Department of Obstetrics and Department of Gynaecology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Irene Cetin
- Department of Biomedical and Clinical Sciences, School of Medicine and Center for Fetal Research Giorgio Pardi, University of Milan, Milan, Italy
| |
Collapse
|
187
|
Liu J, Dias K, Plagnes-Juan E, Veron V, Panserat S, Marandel L. Long-term programming effect of embryonic hypoxia exposure and high-carbohydrate diet at first feeding on glucose metabolism in juvenile rainbow trout. ACTA ACUST UNITED AC 2017; 220:3686-3694. [PMID: 28798080 DOI: 10.1242/jeb.161406] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 08/07/2017] [Indexed: 12/23/2022]
Abstract
Environmental conditions experienced during early life play an important role in the long-term metabolic status of individuals. The present study investigated whether hypoxia exposure [for 24 h: 2.5 mg O2 l-1 (20% dissolved O2)] during the embryonic stage alone (hypoxic history) or combined with a 5-day high-carbohydrate (60%) diet stimulus at first feeding (HC dietary history) can affect glucose metabolism later in life, i.e. in juvenile fish. After 19 weeks of growth, we observed a decrease in final body mass in fish with an HC dietary history. Feed efficiency was significantly affected by both hypoxic and HC dietary histories. After a short challenge test (5 days) performed with a 30% carbohydrate diet in juvenile trout, our results also showed that, in trout that experienced hypoxic history, mRNA levels of gluconeogenic genes in liver and glucose transport genes in both liver and muscle were significantly increased at the juvenile stage. Besides, mRNA levels of glycolytic genes were decreased in fish with an HC dietary history. Both hypoxic and dietary histories barely affected plasma metabolites or global epigenetic modifications in juvenile fish after the challenge test. In conclusion, our results demonstrated that an acute hypoxic stimulus during early development alone or combined with a hyperglucidic stimulus at first feeding can modify growth performance and glucose metabolism at the molecular level in juvenile trout.
Collapse
Affiliation(s)
- Jingwei Liu
- INRA, Université de Pau et des pays de l'Adour, UMR1419 Nutrition Metabolism and Aquaculture, Aquapôle, F-64310 Saint-Pée-sur-Nivelle, France
| | - Karine Dias
- INRA, Université de Pau et des pays de l'Adour, UMR1419 Nutrition Metabolism and Aquaculture, Aquapôle, F-64310 Saint-Pée-sur-Nivelle, France
| | - Elisabeth Plagnes-Juan
- INRA, Université de Pau et des pays de l'Adour, UMR1419 Nutrition Metabolism and Aquaculture, Aquapôle, F-64310 Saint-Pée-sur-Nivelle, France
| | - Vincent Veron
- INRA, Université de Pau et des pays de l'Adour, UMR1419 Nutrition Metabolism and Aquaculture, Aquapôle, F-64310 Saint-Pée-sur-Nivelle, France
| | - Stéphane Panserat
- INRA, Université de Pau et des pays de l'Adour, UMR1419 Nutrition Metabolism and Aquaculture, Aquapôle, F-64310 Saint-Pée-sur-Nivelle, France
| | - Lucie Marandel
- INRA, Université de Pau et des pays de l'Adour, UMR1419 Nutrition Metabolism and Aquaculture, Aquapôle, F-64310 Saint-Pée-sur-Nivelle, France
| |
Collapse
|
188
|
Gouin JP, Zhou QQ, Booij L, Boivin M, Côté SM, Hébert M, Ouellet-Morin I, Szyf M, Tremblay RE, Turecki G, Vitaro F. Associations among oxytocin receptor gene (OXTR) DNA methylation in adulthood, exposure to early life adversity, and childhood trajectories of anxiousness. Sci Rep 2017; 7:7446. [PMID: 28785027 PMCID: PMC5547144 DOI: 10.1038/s41598-017-07950-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 07/06/2017] [Indexed: 12/17/2022] Open
Abstract
Recent models propose deoxyribonucleic acid methylation of key neuro-regulatory genes as a molecular mechanism underlying the increased risk of mental disorder associated with early life adversity (ELA). The goal of this study was to examine the association of ELA with oxytocin receptor gene (OXTR) methylation among young adults. Drawing from a 21-year longitudinal cohort, we compared adulthood OXTR methylation frequency of 46 adults (23 males and 23 females) selected for high or low ELA exposure based on childhood socioeconomic status and exposure to physical and sexual abuse during childhood and adolescence. Associations between OXTR methylation and teacher-rated childhood trajectories of anxiousness were also assessed. ELA exposure was associated with one significant CpG site in the first intron among females, but not among males. Similarly, childhood trajectories of anxiousness were related to one significant CpG site within the promoter region among females, but not among males. This study suggests that females might be more sensitive to the impact of ELA on OXTR methylation than males.
Collapse
Affiliation(s)
- J P Gouin
- Department of Psychology, Concordia University, Montreal, Canada.
- Research Unit on Children's Psychosocial Maladjustment (GRIP), University of Montreal, Montreal, Canada.
| | - Q Q Zhou
- Department of Psychology, Concordia University, Montreal, Canada
| | - L Booij
- Department of Psychology, Concordia University, Montreal, Canada
- Sainte-Justine Hospital Research Center, University of Montreal, Montreal, Canada
- Research Unit on Children's Psychosocial Maladjustment (GRIP), University of Montreal, Montreal, Canada
| | - M Boivin
- Research Unit on Children's Psychosocial Maladjustment (GRIP), Laval University, Québec, Canada
- Institute of Genetic, Neurobiological, and Social Foundations of Child Development, Tomsk State University, Tomsk, Russian Federation
- School of Psychology, Laval University, Québec, Canada
| | - S M Côté
- Department of Social and Preventive Medicine, University of Montreal, Montreal, Canada
- Research Unit on Children's Psychosocial Maladjustment (GRIP), University of Montreal, Montreal, Canada
- Bordeaux Population Health Research Center, INSERM and Bordeaux University, Bordeaux, France
| | - M Hébert
- Department of Sexology, Université du Québec à Montréal, Montreal, Canada
| | - I Ouellet-Morin
- Research Unit on Children's Psychosocial Maladjustment (GRIP), University of Montreal, Montreal, Canada
- Department of Criminology, University of Montreal, Montreal, Canada
| | - M Szyf
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Canada
| | - R E Tremblay
- Research Unit on Children's Psychosocial Maladjustment (GRIP), University of Montreal, Montreal, Canada
- Departments of Pediatrics and Psychology, University of Montreal, Montreal, Canada
- School of Public Health, University College Dublin, Dublin, Ireland
| | - G Turecki
- Research Unit on Children's Psychosocial Maladjustment (GRIP), University of Montreal, Montreal, Canada
- Department of Psychiatry, McGill University, Montreal, Canada
| | - F Vitaro
- Research Unit on Children's Psychosocial Maladjustment (GRIP), University of Montreal, Montreal, Canada
- School of Psychoeducation, University of Montreal, Montreal, Canada
| |
Collapse
|
189
|
Bento-Bernardes T, Toste FP, Pazos-Moura CC, Oliveira KJ. Maternal cinnamon extract intake during lactation leads to sex-specific endocrine modifications in rat offspring. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2017; 97:3855-3863. [PMID: 28182286 DOI: 10.1002/jsfa.8253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 10/25/2016] [Accepted: 02/02/2017] [Indexed: 06/06/2023]
Abstract
BACKGROUND Cinnamon supplementation has been associated with an improvement in glucose disposal and a reduction in fat mass in type 2 diabetes. Maternal nutrition during lactation impacts the health of the offspring throughout life. We hypothesize that cinnamon intake by lactating rats affects maternal physiology, leading to hormonal and metabolic changes in their offspring. To investigate this hypothesis, dams received aqueous cinnamon extract (400 mg cinnamon kg-1 body mass day-1 ) or water orally, during lactation. RESULTS Maternal cinnamon intake did not affect the body mass gain or food intake of dams or their offspring, although it decreased visceral white adipose tissue mass in dams and in their adult offspring of both sexes. Cinnamon-treated dams exhibited no differences in serum insulin, adiponectin, leptin or estradiol levels, although they presented higher serum progesterone. At weaning, cinnamon male pups exhibited lower insulinemia, whereas cinnamon female pups exhibited lower glycemia. Interestingly, in adulthood, only the female offspring exhibited an altered hormonal profile, with reduced serum leptin, adiponectin and insulin levels accompanied by lower glycemia. CONCLUSION The present study demonstrates that maternal cinnamon intake during lactation promotes mild changes in dams and can trigger sex-specific metabolic programming in pups that lasts into adulthood. © 2017 Society of Chemical Industry.
Collapse
Affiliation(s)
- Thais Bento-Bernardes
- Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Rio de Janeiro, Brazil
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda P Toste
- Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Carmen C Pazos-Moura
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Karen J Oliveira
- Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Rio de Janeiro, Brazil
| |
Collapse
|
190
|
Álvarez-Bueno C, Cavero-Redondo I, Lucas-de la Cruz L, Notario-Pacheco B, Martínez-Vizcaíno V. Association between pre-pregnancy overweight and obesity and children’s neurocognitive development: a systematic review and meta-analysis of observational studies. Int J Epidemiol 2017; 46:1653-1666. [DOI: 10.1093/ije/dyx122] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2017] [Indexed: 01/19/2023] Open
|
191
|
Epigenetic dysregulation of protocadherins in human disease. Semin Cell Dev Biol 2017; 69:172-182. [PMID: 28694114 DOI: 10.1016/j.semcdb.2017.07.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 07/03/2017] [Accepted: 07/06/2017] [Indexed: 12/12/2022]
Abstract
Protocadherins (Pcdhs) are a group of cell-cell adhesion molecules that are highly expressed in the nervous system and have a major function in dendrite development and neural circuit formation. However, the role protocadherins play in human health and disease remains unclear. Several recent studies have associated epigenetic dysregulation of protocadherins with possible implications for disease pathogenesis. In this review, we briefly recap the various epigenetic mechanisms regulating protocadherin genes, particularly the clustered Pcdhs. We further outline research describing altered epigenetic regulation of protocadherins in neurological and psychiatric disorders, as well as in cancer and during aging. We additionally present preliminary data on DNA methylation dynamics of clustered protocadherins during fetal brain development, as well as the epigenetic differences distinguishing adult neuronal and glial cells. A deeper understanding of the role of protocadherins in disease is crucial for designing novel diagnostic tools and therapies targeting brain disorders.
Collapse
|
192
|
Embryonic exposure to the widely-used herbicide atrazine disrupts meiosis and normal follicle formation in female mice. Sci Rep 2017; 7:3526. [PMID: 28615648 PMCID: PMC5471253 DOI: 10.1038/s41598-017-03738-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/03/2017] [Indexed: 01/28/2023] Open
Abstract
The widely-used herbicide atrazine (ATZ) is detected in ground and surface water in many countries. Several studies in animals have demonstrated that ATZ has endocrine-disrupting effects on male and female reproduction in many vertebrate species. In this study, we investigated the effects of ATZ exposure on meiosis, a key step in gametogenesis in mammals. The treatment was initiated before oocyte entry into meiosis, which occurs during the embryonic period in females. We found that embryonic exposure to ATZ increases the level of 8-oxo-guanine in the nucleus of meiotic cells, reflecting oxidative stress and affecting meiotic double-strand break repair, chromosome synapsis and crossover numbers. Finally, embryonic exposure to ATZ reduces the number of primordial follicles and increases the incidence of multi-oocyte follicles in adult mice. Our data demonstrate that embryonic exposure to ATZ disrupts prophase I of meiosis and affects normal follicle formation in female mice.
Collapse
|
193
|
Wang Z, Li C, Yang Z, Ma J, Zou Z. Fetal and infant exposure to severe Chinese famine increases the risk of adult dyslipidemia: Results from the China health and retirement longitudinal study. BMC Public Health 2017; 17:488. [PMID: 28610568 PMCID: PMC5470181 DOI: 10.1186/s12889-017-4421-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 05/14/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND To explore the associations between the Chinese famine exposure in early life and the dyslipidemia in adulthood. METHODS We selected 2752 participants from the baseline survey of China Health and Retirement Longitudinal Study (CHARLS) 2011-2012 to evaluate the associations of early life the Chinese famine exposure with risk of dyslipidemia in adulthood. Dyslipidemia was defined as TC (Total Cholesterol): HDL-C (High-Density Lipoprotein Cholesterol) ratio ≥ 5.0 or use cholesterol lowering drugs. Famine exposure cohorts were categorized by birthdates of participants. Binary logistics regression model was used to examine the associations of early-life famine exposure with the risk of dyslipidemia. RESULTS The dyslipidemia prevalence of the non-exposed cohort, fetal stage-, infant stage-, and preschool stage-exposed cohorts in adulthood was 15.7%, 23.1%, 22.0%, and 18.6%, respectively. Early-life exposure to the Chinese famine significantly increased LDL cholesterol concentrations in adulthood after adjusted for age. The risks of dyslipidemia in fetal (OR = 1.58; 95% CI: 1.23-2.03; P < 0.001) and infant (OR = 1.52; 95% CI: 1.15-2.00; P = 0.003) stage-exposed cohorts were significantly higher than the non-exposed cohort after adjusted for gender and current family economic status. Following gender stratification, we found that fetal (OR = 1.80; 95% CI: 1.26-2.57; P = 0.001), infant (OR = 1.75; 95% CI: 1.17-2.62; P = 0.006), and preschool (OR = 1.63; 95% CI: 1.10-2.42; P = 0.015) -stage exposure to severe famine aggravated the risk of dyslipidemia in female adults. However, the similar association was not observed for male adults. CONCLUSIONS Early-life exposure to severe Chinese famine could link with the higher dyslipidemia risk in female adulthood, but not in male adulthood. This gender-specific effect might be associated with the hypothesis that parents in China prefer boys to girls traditionally or survivors' bias.
Collapse
Affiliation(s)
- Zhenghe Wang
- Institute of Child and Adolescent Health, School of Public Health, Peking University Health Science Center, No 38 Xue Yuan Road, Haidian District, Beijing, 100191, China
| | - Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Zhongping Yang
- Institute of Child and Adolescent Health, School of Public Health, Peking University Health Science Center, No 38 Xue Yuan Road, Haidian District, Beijing, 100191, China
| | - Jun Ma
- Institute of Child and Adolescent Health, School of Public Health, Peking University Health Science Center, No 38 Xue Yuan Road, Haidian District, Beijing, 100191, China.
| | - Zhiyong Zou
- Institute of Child and Adolescent Health, School of Public Health, Peking University Health Science Center, No 38 Xue Yuan Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
194
|
Alsaied T, Omar K, James JF, Hinton RB, Crombleholme TM, Habli M. Fetal origins of adult cardiac disease: a novel approach to prevent fetal growth restriction induced cardiac dysfunction using insulin like growth factor. Pediatr Res 2017; 81:919-925. [PMID: 28099426 DOI: 10.1038/pr.2017.18] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 12/30/2016] [Indexed: 01/28/2023]
Abstract
BACKGROUND Fetal growth restriction (FGR) is a risk factor for adult cardiovascular disease. Intraplacental gene transfer of human insulin-like growth factor-1 (IGF-1) corrects birth weight in our mouse model of FGR. This study addresses long term effects of FGR on cardiac function and the potential preventive effect of IGF-1. STUDY DESIGN Laparotomy was performed on pregnant C57BL/6J mice at embryonic day 18 and pups were divided into three groups: Sham operated; FGR (induced by mesenteric uterine artery ligation); treatment (intraplacental injection of IGF-1 after uterine artery ligation). Pups were followed until 32 wk of life. Transthoracic echocardiography was performed starting at 12 wk. RESULTS Systolic cardiac function was significantly impaired in the FGR group with reduced fractional shortening compared with sham and treatment group starting at week 12 of life (20 ± 4 vs. 31 ± 5 vs. 32 ± 5, respectively, n = 12 for each group; P < 0.001) with no difference between the sham and treatment groups. CONCLUSION Intraplacental gene transfer of IGF-1 prevents FGR induced cardiac dysfunction. This suggests that in utero therapy may positively impact cardiac remodeling and prevent adult cardiovascular disease.
Collapse
Affiliation(s)
- Tarek Alsaied
- Cincinnati Children's Hospital Heart Institute, Cincinnati, Ohio
| | - Khaled Omar
- Colorado Fetal Care Center, Division of Pediatric General Thoracic and Fetal Surgery, Children's Hospital of Colorado, Denver, Colorado
| | - Jeanne F James
- Cincinnati Children's Hospital Heart Institute, Cincinnati, Ohio
| | - Robert B Hinton
- Cincinnati Children's Hospital Heart Institute, Cincinnati, Ohio
| | - Timothy M Crombleholme
- Colorado Fetal Care Center, Division of Pediatric General Thoracic and Fetal Surgery, Children's Hospital of Colorado, Denver, Colorado
| | - Mounira Habli
- Center for Molecular Fetal Therapy, Cincinnati Children's Hospital, Cincinnati, Ohio.,Department of General and Thoracic Pediatric Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
195
|
Pauwels S, Truijen I, Ghosh M, Duca RC, Langie SAS, Bekaert B, Freson K, Huybrechts I, Koppen G, Devlieger R, Godderis L. The effect of paternal methyl-group donor intake on offspring DNA methylation and birth weight. J Dev Orig Health Dis 2017; 8:311-321. [PMID: 28260562 DOI: 10.1017/s2040174417000046] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Most nutritional studies on the development of children focus on mother-infant interactions. Maternal nutrition is critically involved in the growth and development of the fetus, but what about the father? The aim is to investigate the effects of paternal methyl-group donor intake (methionine, folate, betaine, choline) on paternal and offspring global DNA (hydroxy)methylation, offspring IGF2 DMR DNA methylation, and birth weight. Questionnaires, 7-day estimated dietary records, whole blood samples, and anthropometric measurements from 74 fathers were obtained. A total of 51 cord blood samples were collected and birth weight was obtained. DNA methylation status was measured using liquid chromatography-tandem mass spectrometry (global DNA (hydroxy)methylation) and pyrosequencing (IGF2 DMR methylation). Paternal betaine intake was positively associated with paternal global DNA hydroxymethylation (0.028% per 100 mg betaine increase, 95% CI: 0.003, 0.053, P=0.03) and cord blood global DNA methylation (0.679% per 100 mg betaine increase, 95% CI: 0.057, 1.302, P=0.03). Paternal methionine intake was positively associated with CpG1 (0.336% per 100 mg methionine increase, 95% CI: 0.103, 0.569, P=0.006), and mean CpG (0.201% per 100 mg methionine increase, 95% CI: 0.001, 0.402, P=0.049) methylation of the IGF2 DMR in cord blood. Further, a negative association between birth weight/birth weight-for-gestational age z-score and paternal betaine/methionine intake was found. In addition, a positive association between choline and birth weight/birth weight-for-gestational age z-score was also observed. Our data indicate a potential impact of paternal methyl-group donor intake on paternal global DNA hydroxymethylation, offspring global and IGF2 DMR DNA methylation, and prenatal growth.
Collapse
Affiliation(s)
- S Pauwels
- 1Department of Public Health and Primary Care, Environment and Health,KU Leuven- University of Leuven,Leuven,Belgium
| | - I Truijen
- 1Department of Public Health and Primary Care, Environment and Health,KU Leuven- University of Leuven,Leuven,Belgium
| | - M Ghosh
- 1Department of Public Health and Primary Care, Environment and Health,KU Leuven- University of Leuven,Leuven,Belgium
| | - R C Duca
- 1Department of Public Health and Primary Care, Environment and Health,KU Leuven- University of Leuven,Leuven,Belgium
| | - S A S Langie
- 2Unit Environmental Risk and Health,Flemish Institute of Technological Research (VITO),Mol,Belgium
| | - B Bekaert
- 4Department of Imaging & Pathology,KU Leuven - University of Leuven,Leuven,Belgium
| | - K Freson
- 6Center for Molecular and Vascular Biology,KU Leuven - University of Leuven,Leuven,Belgium
| | - I Huybrechts
- 7Dietary Exposure Assessment Group,International Agency for Research on Cancer,Lyon,France
| | - G Koppen
- 2Unit Environmental Risk and Health,Flemish Institute of Technological Research (VITO),Mol,Belgium
| | - R Devlieger
- 8Department of Development and Regeneration,KU Leuven-University of Leuven,Leuven,Belgium
| | - L Godderis
- 1Department of Public Health and Primary Care, Environment and Health,KU Leuven- University of Leuven,Leuven,Belgium
| |
Collapse
|
196
|
The epigenetic effects of assisted reproductive technologies: ethical considerations. J Dev Orig Health Dis 2017; 8:436-442. [DOI: 10.1017/s2040174417000344] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The use of assisted reproductive technologies (ART) has increased significantly, allowing many coping with infertility to conceive. However, an emerging body of evidence suggests that ART could carry epigenetic risks for those conceived through the use of these technologies. In accordance with the Developmental Origins of Health and Disease hypothesis, ART could increase the risk of developing late-onset diseases through epigenetic mechanisms, as superovulation, fertilization methods and embryo culture could impair the embryo’s epigenetic reprogramming. Such epigenetic risks raise ethical issues for all stakeholders: prospective parents and children, health professionals and society. This paper focuses on ethical issues raised by the consideration of these risks when using ART. We apply two key ethical principles of North American bioethics (respect for autonomy and non-maleficence) and suggest that an ethical tension may emerge from conflicting duties to promote the reproductive autonomy of prospective parents on one hand, and to minimize risks to prospective children on the other. We argue that this tension is inherent to the entire enterprise of ART and thus cannot be addressed by individual clinicians in individual cases. We also consider the implications of the ‘non-identity problem’ in this context. We call for additional research that would allow a more robust evidence base for policy. We also call upon professional societies to provide clinicians with guidelines and educational resources to facilitate the communication of epigenetic risks associated with ART to patients, taking into consideration the challenges of communicating risk information whose validity is still uncertain.
Collapse
|
197
|
Tajbakhsh A, Mokhtari-Zaer A, Rezaee M, Afzaljavan F, Rivandi M, Hassanian SM, Ferns GA, Pasdar A, Avan A. Therapeutic Potentials of BDNF/TrkB in Breast Cancer; Current Status and Perspectives. J Cell Biochem 2017; 118:2502-2515. [PMID: 28230291 DOI: 10.1002/jcb.25943] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 02/21/2017] [Indexed: 12/14/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a potent neurotrophic factor that has been shown to stimulate breast cancer cell growth and metastasis via tyrosine kinase receptors TrkA, TrkB, and the p75NTR death receptor. The aberrant activation of BDNF/TrkB pathways can modulate several signaling pathways, including Akt/PI3K, Jak/STAT, NF-kB, UPAR/UPA, Wnt/β-catenin, and VEGF pathways as well as the ER receptor. Several microRNAs have been identified that are involved in the modulation of BDNF/TrkB pathways. These include miR-206, miR-204, MiR-200a/c, MiR-210, MiR-134, and MiR-191; and these may be of value as prognostic and predictive biomarkers for detecting patients at high risk of developing breast cancer. It has been also been demonstrated that a high expression of genes involved in the BDNF pathway in breast cancer is associated with poor clinical outcome and reduced survival of patients. Several approaches have been developed for targeting this pathway, for example TKr inhibitors (AZD6918, CEP-701) and RNA interference. The aim of the current review was to provide an overview of the role of BDNF/TrkB pathways in the pathogenesis of breast cancer and its value as a potential therapeutic target. J. Cell. Biochem. 118: 2502-2515, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Mokhtari-Zaer
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Centre and Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Rezaee
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Afzaljavan
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Rivandi
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, UK
| | - Alireza Pasdar
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Division of Applied Medicine, Medical School, University of Aberdeen, Foresterhill, Aberdeen, UK.,Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
198
|
Xu J, Sinclair KD. One-carbon metabolism and epigenetic regulation of embryo development. Reprod Fertil Dev 2017; 27:667-76. [PMID: 25710200 DOI: 10.1071/rd14377] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 01/08/2015] [Indexed: 12/15/2022] Open
Abstract
One-carbon (1C) metabolism consists of an integrated series of metabolic pathways that include the folate cycle and methionine remethylation and trans-sulfuration pathways. Most, but not all, 1C metabolic enzymes are expressed in somatic cells of the ovary, mammalian oocytes and in preimplantation embryos. The metabolic implications of this, with regard to the provision of methyl donors (e.g. betaine) and 1C cofactors (e.g. vitamin B12), together with consequences of polymorphic variances in genes encoding 1C enzymes, are not fully understood but are the subject of ongoing investigations at the authors' laboratory. However, deficiencies in 1C-related substrates and/or cofactors during the periconception period are known to lead to epigenetic alterations in DNA and histone methylation in genes that regulate key developmental processes in the embryo. Such epigenetic modifications have been demonstrated to negatively impact on the subsequent health and metabolism of offspring. For this reason, parental nutrition around the time of conception has become a focal point of investigation in many laboratories with the aim of providing improved nutritional advice to couples. These issues are considered in detail in this article, which offers a contemporary overview of the effects of 1C metabolism on epigenetic programming in mammalian gametes and the early embryo.
Collapse
Affiliation(s)
- Juan Xu
- School of Bioscience, University of Nottingham, Sutton Bonington, Leicestershire LE12 5RD, UK
| | - Kevin D Sinclair
- School of Bioscience, University of Nottingham, Sutton Bonington, Leicestershire LE12 5RD, UK
| |
Collapse
|
199
|
Annalora AJ, Marcus CB, Iversen PL. Alternative Splicing in the Cytochrome P450 Superfamily Expands Protein Diversity to Augment Gene Function and Redirect Human Drug Metabolism. Drug Metab Dispos 2017; 45:375-389. [PMID: 28188297 DOI: 10.1124/dmd.116.073254] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 02/06/2017] [Indexed: 02/13/2025] Open
Abstract
The human genome encodes 57 cytochrome P450 genes, whose enzyme products metabolize hundreds of drugs, thousands of xenobiotics, and unknown numbers of endogenous compounds, including steroids, retinoids, and eicosanoids. Indeed, P450 genes are the first line of defense against daily environmental chemical challenges in a manner that parallels the immune system. Several National Institutes of Health databases, including PubMed, AceView, and Ensembl, were queried to establish a comprehensive analysis of the full human P450 transcriptome. This review describes a remarkable diversification of the 57 human P450 genes, which may be alternatively processed into nearly 1000 distinct mRNA transcripts to shape an individual's P450 proteome. Important P450 splice variants from families 1A, 1B, 2C, 2D, 3A, 4F, 19A, and 24A have now been documented, with some displaying alternative subcellular distribution or catalytic function directly linked to a disease pathology. The expansion of P450 transcript diversity involves tissue-specific splicing factors, transformation-sensitive alternate splicing, trans-splicing between gene transcripts, single-nucleotide polymorphisms, and epigenetic regulation of alternate splicing. Homeostatic regulation of variant P450 expression is influenced also by nuclear receptor signaling, suppression of nonsense-mediated decay or premature termination codons, mitochondrial dysfunction, or host infection. This review focuses on emergent aspects of the adaptive gene-splicing process, which when viewed through the lens of P450-nuclear receptor gene interactions, resembles a primitive immune-like system that can rapidly monitor, respond, and diversify to acclimate to fluctuations in endo-xenobiotic exposure. Insights gained from this review should aid future drug discovery and improve therapeutic management of personalized drug regimens.
Collapse
Affiliation(s)
- Andrew J Annalora
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon
| | - Craig B Marcus
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon
| | - Patrick L Iversen
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon
| |
Collapse
|
200
|
Mela V, Piscitelli F, Berzal AL, Chowen J, Silvestri C, Viveros MP, Di Marzo V. Sex-dependent effects of neonatal maternal deprivation on endocannabinoid levels in the adipose tissue: influence of diet. J Physiol Biochem 2017; 73:349-357. [DOI: 10.1007/s13105-017-0558-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 03/08/2017] [Indexed: 12/27/2022]
|