101
|
Kasparian NA. Heart care before birth: A psychobiological perspective on fetal cardiac diagnosis. PROGRESS IN PEDIATRIC CARDIOLOGY 2019. [DOI: 10.1016/j.ppedcard.2019.101142] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
102
|
Şanlı E, Kabaran S. Maternal Obesity, Maternal Overnutrition and Fetal Programming: Effects of Epigenetic Mechanisms on the Development of Metabolic Disorders. Curr Genomics 2019; 20:419-427. [PMID: 32476999 PMCID: PMC7235386 DOI: 10.2174/1389202920666191030092225] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 10/12/2019] [Accepted: 10/21/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Maternal obesity and maternal overnutrition, can lead to epigenetic alterations during pregnancy and these alterations can influence fetal and neonatal phenotype which increase the risk of metabolic disorders in later stages of life. OBJECTIVE The effects of maternal obesity on fetal programming and potential mechanisms of maternal epigenetic regulation of gene expression which have persistent effects on fetal health and development were investigated. METHODS Review of the literature was carried out in order to discuss the effects of maternal obesity and epigenetic mechanisms in fetal programming of metabolic disorders. All abstracts and full-text articles were examined and the most relevant articles were included in this review. RESULTS Maternal obesity and maternal overnutrition during fetal period has important overall effects on long-term health. Maternal metabolic alterations during early stages of fetal development can lead to permanent changes in organ structures, cell numbers and metabolism. Epigenetic modifications (DNA methylation, histone modifications, microRNAs) play an important role in disease susceptibility in the later stages of human life. Maternal nutrition alter expression of hypothalamic genes which can increase fetal and neonatal energy intake. Epigenetic modifications may affect the increasing rate of obesity and other metabolic disorders worldwide since the impact of these changes can be passed through generations. CONCLUSION Weight management before and during pregnancy, together with healthy nutritional intakes may improve the maternal metabolic environment, which can reduce the risks of fetal programming of metabolic diseases. Further evidence from long-term follow-up studies are needed in order to determine the role of maternal obesity on epigenetic mechanisms.
Collapse
Affiliation(s)
- Ezgi Şanlı
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Eastern Mediterranean University, Famagusta, T.R. North Cyprus via Mersin 10, Turkey
| | - Seray Kabaran
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Eastern Mediterranean University, Famagusta, T.R. North Cyprus via Mersin 10, Turkey
| |
Collapse
|
103
|
Wróblewski A, Strycharz J, Świderska E, Drewniak K, Drzewoski J, Szemraj J, Kasznicki J, Śliwińska A. Molecular Insight into the Interaction between Epigenetics and Leptin in Metabolic Disorders. Nutrients 2019; 11:nu11081872. [PMID: 31408957 PMCID: PMC6723573 DOI: 10.3390/nu11081872] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/16/2022] Open
Abstract
Nowadays, it is well-known that the deregulation of epigenetic machinery is a common biological event leading to the development and progression of metabolic disorders. Moreover, the expression level and actions of leptin, a vast adipocytokine regulating energy metabolism, appear to be strongly associated with epigenetics. Therefore, the aim of this review was to summarize the current knowledge of the epigenetic regulation of leptin as well as the leptin-induced epigenetic modifications in metabolic disorders and associated phenomena. The collected data indicated that the deregulation of leptin expression and secretion that occurs during the course of metabolic diseases is underlain by a variation in the level of promoter methylation, the occurrence of histone modifications, along with miRNA interference. Furthermore, leptin was proven to epigenetically regulate several miRNAs and affect the activity of the histone deacetylases. These epigenetic modifications were observed in obesity, gestational diabetes, metabolic syndrome and concerned various molecular processes like glucose metabolism, insulin sensitivity, liver fibrosis, obesity-related carcinogenesis, adipogenesis or fetal/early postnatal programming. Moreover, the circulating miRNA profiles were associated with the plasma leptin level in metabolic syndrome, and miRNAs were found to be involved in hypothalamic leptin sensitivity. In summary, the evidence suggests that leptin is both a target and a mediator of epigenetic changes that develop in numerous tissues during metabolic disorders.
Collapse
Affiliation(s)
- Adam Wróblewski
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland.
| | - Justyna Strycharz
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland
| | - Ewa Świderska
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland
| | - Karolina Drewniak
- Student Scientific Society of the Civilization Diseases, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland
| | - Józef Drzewoski
- Central Teaching Hospital of the Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland
| | - Jacek Kasznicki
- Department of Internal Diseases, Diabetology and Clinical Pharmacology, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland
| | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland.
| |
Collapse
|
104
|
Post-translational histone modifications and their interaction with sex influence normal brain development and elaboration of neuropsychiatric disorders. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1968-1981. [DOI: 10.1016/j.bbadis.2018.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023]
|
105
|
Gogou M, Kolios G. Nutritional Supplements During Gestation and Autism Spectrum Disorder: What Do We Really Know and How Far Have We Gone? J Am Coll Nutr 2019; 39:261-271. [PMID: 31318329 DOI: 10.1080/07315724.2019.1635920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Nutritional interventions are gaining remarkable attention as complementary management options for autism. Our aim is to provide literature data about the impact of the administration of dietary supplements during pregnancy on the risk of autism spectrum disorder in the offspring. A comprehensive search was undertaken by 2 reviewers independently using PubMed as the medical database source. Prospective clinical and experimental studies were considered and no year-of-publication restriction was placed. We were able to identify 4 basic (conducted in rodents) and 3 clinical research papers fulfilling our selection criteria. Supplements studied included folic acid, iron, multivitamins, choline, vitamin D, and docosahexaenoic acid. Choline and folic acid had a significant impact on the expression of autism-related genes. However, from a clinical point of view, prenatal folate administration did not reduce the risk of autism. Similarly, iron had no significant impact, while the use of multivitamins in moderate frequency had a protective effect. The use of vitamin D and docosahexaenoic acid during gestation decreased the incidence of autism in animal models. In conclusion, available data are controversial and cannot change current routine practice. More large-scale prospective studies are needed to identify the real effect of nutritional supplements and also optimize their administration.Key teaching pointsMultivitamins use during pregnancy can exert a protective effect on the risk of autism, although depending on the frequency of use. Nevertheless, prenatal iron and folate were not shown to have any significant impact.Research based on animal models showed that choline and folic acid can have a significant impact on the expression of autism-related genes in a sex-specific manner.Furthermore, the use of vitamin D and docosahexaenoic acid during gestation seem to decrease the incidence of autism in animal offspring.In the future, more clinical, large-scale prospective and methodologically homogenous clinical studies are needed to further investigate the effect of the periconceptional use of nutritional supplements on autism risk.
Collapse
Affiliation(s)
- Maria Gogou
- 2nd Department of Pediatrics, School of Medicine, Aristotle University of Thessaloniki, University General Hospital AHEPA, Thessaloniki, Greece
| | - George Kolios
- Laboratory of Pharmacology, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
106
|
Soares MJ, Varberg KM, Iqbal K. Hemochorial placentation: development, function, and adaptations. Biol Reprod 2019; 99:196-211. [PMID: 29481584 DOI: 10.1093/biolre/ioy049] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/21/2018] [Indexed: 11/12/2022] Open
Abstract
Placentation is a reproductive adaptation that permits fetal growth and development within the protected confines of the female reproductive tract. Through this important role, the placenta also determines postnatal health and susceptibility to disease. The hemochorial placenta is a prominent feature in primate and rodent development. This manuscript provides an overview of the basics of hemochorial placental development and function, provides perspectives on major discoveries that have shaped placental research, and thoughts on strategies for future investigation.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas, USA and the Center for Perinatal Research, Children΄s Research Institute, Children΄s Mercy, Kansas City, Missouri, USA
| | - Kaela M Varberg
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
107
|
Kim EN, Lee JY, Shim JY, Hwang D, Kim KC, Kim SR, Kim CJ. Clinicopathological characteristics of miscarriages featuring placental massive perivillous fibrin deposition. Placenta 2019; 86:45-51. [PMID: 31326089 DOI: 10.1016/j.placenta.2019.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/19/2019] [Accepted: 07/11/2019] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Massive perivillous fibrin deposition (MPFD) is frequently associated with detrimental pregnancy outcomes, and extensive perivillous fibrin deposition results in severe placental dysfunction and loss of maternofetal interface. Unfortunately, the fundamental pathogenesis of MPFD remains unknown, and systematic analyses of MPFD in miscarriage is lacking. We analyzed the frequency and clinicopathological characteristics of MPFD in first trimester miscarriages. METHODS We analyzed a consecutive series of miscarriages (n = 582) gathered between March 2012 and June 2016. MPFD was classified as fibrin-type (f-MPFD) and matrix-type (m-MPFD) by immunostaining for fibrin and collagen type IV. The control group consisted of miscarriage cases (MC, n = 18) that were matched to f-MPFD with normal chromosome (f-MPFD-nc) for number of previous miscarriages and placental chromosomal status. RESULTS MPFD was identified in 2.7% of miscarriages. f-MPFD was associated with recurrent abortions. Compared with miscarriages without fibrin deposition, MPFD cases had higher proportion of those with normal placental chromosome (69.2% vs. 27.4%, P < 0.005) and higher frequency of villous syncytiotrophoblast C4d deposition (73.3% vs. 33.9%, P < 0.005). All C4d(+) f-MPFD patients had more than three recurrent miscarriages, whereas C4d(-) f-MPFD patients had no history of recurrent miscarriage (P < 0.05). Patients with f-MPFD-nc had significantly higher HLA PRA immunopositivity rate than did MC patients (P = 0.005). DISCUSSION MPFD was more common in miscarriages than in preterm and term pregnancies. Placental massive fibrin-type fibrinoid deposition and villous C4d immunoreactivity were associated with recurrent miscarriage.
Collapse
Affiliation(s)
- Eun Na Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Asan Laboratory of Perinatal Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | - Jae-Yoon Shim
- Asan Laboratory of Perinatal Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Obstetrics and Gynecology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | - Ki Chul Kim
- Hamchoon Women's Clinic, Seoul, Republic of Korea
| | - So Ra Kim
- Asan Laboratory of Perinatal Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chong Jai Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Asan Laboratory of Perinatal Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
108
|
Barrientos RM, Brunton PJ, Lenz KM, Pyter L, Spencer SJ. Neuroimmunology of the female brain across the lifespan: Plasticity to psychopathology. Brain Behav Immun 2019; 79:39-55. [PMID: 30872093 PMCID: PMC6591071 DOI: 10.1016/j.bbi.2019.03.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/25/2019] [Accepted: 03/09/2019] [Indexed: 02/06/2023] Open
Abstract
The female brain is highly dynamic and can fundamentally remodel throughout the normal ovarian cycle as well as in critical life stages including perinatal development, pregnancy and old-age. As such, females are particularly vulnerable to infections, psychological disorders, certain cancers, and cognitive impairments. We will present the latest evidence on the female brain; how it develops through the neonatal period; how it changes through the ovarian cycle in normal individuals; how it adapts to pregnancy and postpartum; how it responds to illness and disease, particularly cancer; and, finally, how it is shaped by old age. Throughout, we will highlight female vulnerability to and resilience against disease and dysfunction in the face of environmental challenges.
Collapse
Affiliation(s)
- R M Barrientos
- Institute for Behavioral Medicine Research, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Department of Psychiatry and Behavioral Health, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Chronic Brain Injury Program, Discovery Themes Initiative, The Ohio State University, Columbus, OH 43210, United States
| | - P J Brunton
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, Scotland, UK; Zhejiang University-University of Edinburgh Joint Institute, Zhejiang University School of Medicine, International Campus, Haining, Zhejiang 314400, PR China
| | - K M Lenz
- Institute for Behavioral Medicine Research, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Department of Psychology, Department of Neuroscience, The Ohio State University, Columbus, OH 43210, United States
| | - L Pyter
- Institute for Behavioral Medicine Research, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Department of Psychiatry and Behavioral Health, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States
| | - S J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic. 3083, Australia.
| |
Collapse
|
109
|
Adverse Maternal Metabolic Intrauterine Environment and Placental Epigenetics: Implications for Fetal Metabolic Programming. Curr Environ Health Rep 2019; 5:531-543. [PMID: 30267228 DOI: 10.1007/s40572-018-0217-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE OF REVIEW Herein, we summarize existent epidemiological studies relating adverse maternal metabolic environments of maternal obesity and gestational diabetes and placental DNA methylation. RECENT FINDINGS Multiple studies have evaluated associations between intrauterine exposure to gestational diabetes and/or maternal glucose levels and DNA methylation at candidate metabolic genes as well as in epigenome-wide studies. Some of the genomic regions more consistently associated include lipid-related genes (LPL and PPARGC1A), the major histocompatibility complex (MHC), and imprinted genes. Studies solely focused on maternal obesity influences on the placental epigenome are scarce. Understanding the placental mechanisms involved in fetal metabolic programming could lead to discovery of placental biomarkers at birth that predict later-life metabolic risk. Moving forward is important to standardize methods utilized in epigenetics research; consistent methodology can help interpret disparate findings. Larger studies with longitudinal follow-up are needed to address future challenges in fetal programming research.
Collapse
|
110
|
Herrera-Van Oostdam AS, Salgado-Bustamante M, López JA, Herrera-Van Oostdam DA, López-Hernández Y. Placental exosomes viewed from an 'omics' perspective: implications for gestational diabetes biomarkers identification. Biomark Med 2019; 13:675-684. [PMID: 31157549 DOI: 10.2217/bmm-2018-0468] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Exosomes are defined as extracellular vesicles that are released from cells upon fusion of an intermediate endocytic compartment - the multivesicular body - with the plasma membrane. Recently, placenta-derived exosomes have gained special attention, since they play a crucial role in the communication between the mother and fetus. It is known that the concentration of placenta-derived exosomes in the maternal bloodstream is higher in patients with preeclampsia or gestational diabetes mellitus. However, their composition in terms of the content of proteins, nucleic acids or lipids is uncertain. In this work, we reviewed the recent advances in placental exosomes characterization through omics-based methods, and their potential to predict gestational diabetes mellitus.
Collapse
Affiliation(s)
- Ana S Herrera-Van Oostdam
- Department of Biochemistry, Medicine Faculty, Universidad Autónoma de San Luis Potosí, San Luis Potosí, PC 78210, Mexico
| | - Mariana Salgado-Bustamante
- Department of Biochemistry, Medicine Faculty, Universidad Autónoma de San Luis Potosí, San Luis Potosí, PC 78210, Mexico
| | - Jesús Adrián López
- MicroRNAs Laboratory, Unidad Académica de Ciencias Biológicas, Universidad Autónoma de Zacatecas, Zacatecas, PC 98610, Mexico
| | - David A Herrera-Van Oostdam
- Department of Rheumatology & Pathology, Hospital Central 'Dr. Ignacio Morones Prieto,' Universidad Autónoma de San Luis Potosí, San Luis Potosí, PC 78210, Mexico
| | - Yamilé López-Hernández
- CONACyT-Universidad Autónoma de Zacatecas, Unidad Académica de Ciencias Biológicas, Zacatecas, PC 98610, Mexico
| |
Collapse
|
111
|
Chen HJ, Gur TL. Intrauterine Microbiota: Missing, or the Missing Link? Trends Neurosci 2019; 42:402-413. [PMID: 31053242 PMCID: PMC6604064 DOI: 10.1016/j.tins.2019.03.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/21/2019] [Accepted: 03/19/2019] [Indexed: 12/29/2022]
Abstract
The intrauterine environment provides a key interface between the mother and the developing fetus during pregnancy, and is a target for investigating mechanisms of fetal programming. Studies have demonstrated an association between prenatal stress and neurodevelopmental disorders. The role of the intrauterine environment in mediating this effect is still being elucidated. In this review, we discuss emerging preclinical and clinical evidence suggesting the existence of microbial communities in utero. We also outline possible mechanisms of bacterial translocation to the intrauterine environment and immune responses to the presence of microbes or microbial components. Lastly, we overview the effects of intrauterine inflammation on neurodevelopment. We hypothesize that maternal gestational stress leads to disruptions in the maternal oral, gut, and vaginal microbiome that may lead to the translocation of bacteria to the intrauterine environment, eliciting an inflammatory response and resulting in deficits in neurodevelopment.
Collapse
Affiliation(s)
- Helen J Chen
- Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, OH, USA
| | - Tamar L Gur
- Department of Psychiatry and Behavioral Health, Wexner Medical Center at The Ohio State University, Columbus, OH, USA; Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, OH, USA; Department of Obstetrics and Gynecology, Wexner Medical Center at The Ohio State University, Columbus, OH, USA; Institute of Behavioral Medicine Research, Wexner Medical Center at The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
112
|
Appleton AA, Kiley K, Holdsworth EA, Schell LM. Social Support During Pregnancy Modifies the Association Between Maternal Adverse Childhood Experiences and Infant Birth Size. Matern Child Health J 2019; 23:408-415. [PMID: 30627949 DOI: 10.1007/s10995-018-02706-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Introduction Adverse childhood experiences (ACEs) can deleteriously affect health, including pregnancy and birth outcomes occurring later in life. Identification of modifiable factors during pregnancy that buffer the ill effects of adversity is warranted. Social support during pregnancy can promote better birth outcomes, yet it is unknown whether it could also mitigate perinatal risks stemming from ACEs. Thus, this study considers multiple forms of social support in pregnancy as modifiers of an ACEs and fetal growth association. Methods Data were collected from mother and infant pairs from an ongoing prospective birth cohort. Women enrolled around 27 weeks gestation and completed gold-standard assessments of ACEs and social support. Infant cephalization index scores [(head circumference /birthweight) × 100; a marker of asymmetric fetal growth] were derived. Multivariable regression models tested main effects and interaction between ACEs and social support in relation to infant cephalization. Results Higher levels of ACEs were associated with higher cephalization scores (β = 0.01, SE = 0.01, p < 0.05) whereas higher social support was associated with lower cephalization scores (β = - 0.03, SE = 0.01, p < 0.05). A significant interaction was observed showing a protective effect of social support among those with low (0 events) and moderate (1-3 events) ACEs but not among those with high ACEs (4 + events; p < 0.05). Tangible and emotional support, but not information support, contributed to the associations. Discussion Maternal ACEs can deleteriously affect birth size, yet social support during pregnancy provides some buffer from its enduring effects. Interventions designed to enhance pregnancy social support may not only improve maternal wellbeing, but may also safeguard infant health.
Collapse
Affiliation(s)
- Allison A Appleton
- Department of Epidemiology and Biostatistics, University at Albany School of Public Health, 1 University Place, Rensselaer, NY, 12144, USA.
| | - Kevin Kiley
- Department of Obstetrics and Gynecology, Albany Medical College, 391 Myrtle Avenue, Albany, NY, 12208, USA
| | - Elizabeth A Holdsworth
- Department of Anthropology, University at Albany College of Arts and Sciences, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Lawrence M Schell
- Department of Epidemiology and Biostatistics, University at Albany School of Public Health, 1 University Place, Rensselaer, NY, 12144, USA.,Department of Anthropology, University at Albany College of Arts and Sciences, 1400 Washington Avenue, Albany, NY, 12222, USA
| |
Collapse
|
113
|
Jones SL, Dufoix R, Laplante DP, Elgbeili G, Patel R, Chakravarty MM, King S, Pruessner JC. Larger Amygdala Volume Mediates the Association Between Prenatal Maternal Stress and Higher Levels of Externalizing Behaviors: Sex Specific Effects in Project Ice Storm. Front Hum Neurosci 2019; 13:144. [PMID: 31156408 PMCID: PMC6528106 DOI: 10.3389/fnhum.2019.00144] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 04/15/2019] [Indexed: 01/18/2023] Open
Abstract
Introduction: The amygdala is a brain structure involved in emotional regulation. Studies have shown that larger amygdala volumes are associated with behavioral disorders. Prenatal maternal depression is associated with structural changes in the amygdala, which in turn, is predictive of an increase in behavioral problems. Girls may be particularly vulnerable. However, it is not known whether disaster-related prenatal maternal stress (PNMS), or which aspect of the maternal stress experience (i.e., objective hardship, subjective distress, and cognitive appraisal), influences amygdala volumes. Nor is it known whether amygdala volumes mediate the effect of PNMS on behavioral problems in girls and boys. Aims: To assess whether aspects of PNMS are associated with amygdala volume, to determine whether timing of exposure moderates the effect, and to test whether amygdala volume mediates the association between PNMS and internalizing and externalizing problems in 11½ year old children exposed in utero, to varying levels of disaster-related PNMS. Methods: Bilateral amygdala volumes (AGV) and total brain volume (TBV) were acquired using magnetic resonance imaging, from 35 boys and 33 girls whose mothers were pregnant during the January 1998 Quebec Ice Storm. The mothers' disaster-related stress was assessed in June 1998. Child internalizing and externalizing problems were assessed at 11½ years using the Child Behavior Checklist (CBCL). Hierarchical regression analyses and mediation analyses were conducted on boys and girls separately, controlling for perinatal and postnatal factors. Results: In boys, subjective distress was associated with larger right AGV/TBV when mothers where exposed during late pregnancy, which in turn explained higher levels of externalizing behavior. However, when adjusting for postnatal factors, the effect was no longer significant. In girls, later gestational exposure to the ice storm was associated with larger AGV/TBV, but here, higher levels of objective PNMS were associated with more externalizing problems, which was, in part, mediated by larger AGV/TBV. No effects were detected on internalizing behaviors. Conclusion: These results suggest that the effects of PNMS on amygdala development and externalizing symptoms, as assessed in boys and girls in early adolescence, can be influenced by the timing of the stress in pregnancy, and the particular aspect of the mother's stress experience.
Collapse
Affiliation(s)
- Sherri Lee Jones
- Laboratory of Suzanne King, Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Laboratory of Suzanne King, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Romane Dufoix
- Laboratory of Suzanne King, Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Laboratory of Suzanne King, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - David P. Laplante
- Laboratory of Suzanne King, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Guillaume Elgbeili
- Laboratory of Suzanne King, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Raihaan Patel
- Computational Brain Anatomy Laboratory (CoBrA Lab), Douglas Mental Health University Institute, Montreal, QC, Canada
- Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - M. Mallar Chakravarty
- Computational Brain Anatomy Laboratory (CoBrA Lab), Douglas Mental Health University Institute, Montreal, QC, Canada
- Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Biological and Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Suzanne King
- Laboratory of Suzanne King, Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Laboratory of Suzanne King, Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Jens C. Pruessner
- Laboratory of Jens Pruessner, Department of Psychology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
114
|
Nelson LH, Saulsbery AI, Lenz KM. Small cells with big implications: Microglia and sex differences in brain development, plasticity and behavioral health. Prog Neurobiol 2019; 176:103-119. [PMID: 30193820 PMCID: PMC8008579 DOI: 10.1016/j.pneurobio.2018.09.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 07/17/2018] [Accepted: 09/01/2018] [Indexed: 12/20/2022]
Abstract
Brain sex differences are programmed largely by sex hormone secretions and direct sex chromosome effects in early life, and are subsequently modulated by early life experiences. The brain's resident immune cells, called microglia, actively contribute to brain development. Recent research has shown that microglia are sexually dimorphic, especially during early life, and may participate in sex-specific organization of the brain and behavior. Likewise, sex differences in immune cells and their signaling in the adult brain have been found, although in most cases their function remains unclear. Additionally, immune cells and their signaling have been implicated in many disorders in which brain development or plasticity is altered, including autism, schizophrenia, pain disorders, major depression, and postpartum depression. This review summarizes what is currently known about sex differences in neuroimmune function in development and during other major phases of brain plasticity, as well as the current state of knowledge regarding sex-specific neuroimmune function in psychiatric disorders.
Collapse
Affiliation(s)
- Lars H Nelson
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Angela I Saulsbery
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
| | - Kathryn M Lenz
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
115
|
Duan C, Hare M, Staring M, Deligiannidis KM. Examining the relationship between perinatal depression and neurodevelopment in infants and children through structural and functional neuroimaging research. Int Rev Psychiatry 2019; 31:264-279. [PMID: 30701993 PMCID: PMC6594877 DOI: 10.1080/09540261.2018.1527759] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Depression is the most common perinatal psychiatric disorder, but little is known about how it may impact offspring neurodevelopment, as well as the mechanisms by which it may confer transgenerational psychiatric risk. This review presents imaging studies conducted to evaluate the relationship between perinatal depression (PND) and infant and child neurodevelopment. Altered structural and functional connectivity is implicated in children exposed to PND and anxiety. Overall, there are changes in connectivity between amygdala and the prefrontal cortex. Studies suggest decreased hippocampal growth in the first 6 months after birth, decreased cortical thickness in children, and increased amygdala volumes, that are more pronounced in female offspring. Future research is needed to understand the impact of PND on development so that early interventions which promote mother-infant bonding and cognitive development may improve developmental outcomes in children exposed to PND, reducing later risk of psychopathology.
Collapse
Affiliation(s)
- Christy Duan
- Department of Psychiatry, Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 11004, USA
| | - Megan Hare
- Department of Psychology, Florida International University, Miami, FL 33199, USA
| | - Morganne Staring
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Kristina M. Deligiannidis
- Department of Psychiatry, Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 11004, USA,Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA,Departments of Psychiatry and Obstetrics & Gynecology, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA,Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| |
Collapse
|
116
|
Schmitz J, Güntürkün O, Ocklenburg S. Building an Asymmetrical Brain: The Molecular Perspective. Front Psychol 2019; 10:982. [PMID: 31133928 PMCID: PMC6524718 DOI: 10.3389/fpsyg.2019.00982] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 04/15/2019] [Indexed: 12/17/2022] Open
Abstract
The brain is one of the most prominent examples for structural and functional differences between the left and right half of the body. For handedness and language lateralization, the most widely investigated behavioral phenotypes, only a small fraction of phenotypic variance has been explained by molecular genetic studies. Due to environmental factors presumably also playing a role in their ontogenesis and based on first molecular evidence, it has been suggested that functional hemispheric asymmetries are partly under epigenetic control. This review article aims to elucidate the molecular factors underlying hemispheric asymmetries and their association with inner organ asymmetries. While we previously suggested that epigenetic mechanisms might partly account for the missing heritability of handedness, this article extends this idea by suggesting possible alternatives for transgenerational transmission of epigenetic states that do not require germ line epigenetic transmission. This is in line with a multifactorial model of hemispheric asymmetries, integrating genetic, environmental, and epigenetic influencing factors in their ontogenesis.
Collapse
Affiliation(s)
- Judith Schmitz
- Biopsychology, Department of Psychology, Institute of Cognitive Neuroscience, Ruhr University Bochum, Bochum, Germany
| | | | | |
Collapse
|
117
|
Addo KA, Bulka C, Dhingra R, Santos HP, Smeester L, O’Shea TM, Fry RC. Acetaminophen use during pregnancy and DNA methylation in the placenta of the extremely low gestational age newborn (ELGAN) cohort. ENVIRONMENTAL EPIGENETICS 2019; 5:dvz010. [PMID: 31404209 PMCID: PMC6682751 DOI: 10.1093/eep/dvz010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 05/09/2023]
Abstract
Acetaminophen is considered the safest antipyretic and analgesic medication for pregnant women. However, studies have reported that acetaminophen has endocrine disrupting properties and prenatal exposure has been associated with early life epigenetic changes and later life health outcomes. As the placenta is the central mediator of maternal and fetal interactions, exposure to acetaminophen during pregnancy could manifest as perturbations in the placenta epigenome. Here, we evaluated epigenome-wide cytosine-guanine dinucleotide (CpG) methylation in placental tissue in relation to maternal acetaminophen use during pregnancy in a cohort of 286 newborns born prior to 28 weeks gestation. According to maternal self-report, more than half (166 of 286) of the newborns were exposed to acetaminophen in utero. After adjustment for potential confounders, a total of 42 CpGs were identified to be differentially methylated at a false discovery rate < 0.05, with most displaying increased methylation as it relates to acetaminophen exposure. A notable gene that was significantly associated with acetaminophen is the prostaglandin receptor (PTGDR) which plays an essential role in mediating placental blood flow and fetal growth. Moreover, for 6 of the 42 CpGs, associations of acetaminophen use with methylation were significantly different between male and female placentas; 3 CpG sites were associated with acetaminophen use in the male placenta and 3 different sites were associated with acetaminophen use in the female placenta (P interaction < 0.2). These findings highlight a relationship between maternal acetaminophen use during pregnancy and the placental epigenome and suggest that the responses for some CpG sites are sex dependent.
Collapse
Affiliation(s)
- Kezia A Addo
- Curriculum in Toxicology and Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Catherine Bulka
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Radhika Dhingra
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
- Institute for Environmental Health Solutions, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Hudson P Santos
- Institute for Environmental Health Solutions, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
- School of Nursing, University of North Carolina, Chapel Hill, NC, USA
| | - Lisa Smeester
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
- Institute for Environmental Health Solutions, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - T Michael O’Shea
- Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Rebecca C Fry
- Curriculum in Toxicology and Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
- Institute for Environmental Health Solutions, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
- Correspondence address. Curriculum in Toxicology and Environmental Medicine, School of Medicine, University of North Carolina, Rosenau Hall, Rm 166, Campus Box 7431, 135 Dauer Drive, Chapel Hill, NC 27599, USA. Tel: +1-919-966-1171; Fax: +1-919-966-7911; E-mail:
| |
Collapse
|
118
|
Histone Acetylation of Immune Regulatory Genes in Human Placenta in Association with Maternal Intake of Olive Oil and Fish Consumption. Int J Mol Sci 2019; 20:ijms20051060. [PMID: 30823645 PMCID: PMC6429118 DOI: 10.3390/ijms20051060] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 02/26/2019] [Indexed: 12/13/2022] Open
Abstract
Maternal diet modifies epigenetic programming in offspring, a potentially critical factor in the immune dysregulation of modern societies. We previously found that prenatal fish oil supplementation affects neonatal T-cell histone acetylation of genes implicated in adaptive immunity including PRKCZ, IL13, and TBX21. In this study, we measured H3 and H4 histone acetylation levels by chromatin immunoprecipitation in 173 term placentas collected in the prospective birth cohort, ALADDIN, in which information on lifestyle and diet is thoroughly recorded. In anthroposophic families, regular olive oil usage during pregnancy was associated with increased H3 acetylation at FOXP3 (p = 0.004), IL10RA (p = 0.008), and IL7R (p = 0.007) promoters, which remained significant after adjustment by offspring gender. Furthermore, maternal fish consumption was associated with increased H4 acetylation at the CD14 gene in placentas of female offspring (p = 0.009). In conclusion, prenatal olive oil intake can affect placental histone acetylation in immune regulatory genes, confirming previously observed pro-acetylation effects of olive oil polyphenols. The association with fish consumption may implicate ω-3 polyunsaturated fatty acids present in fish oil. Altered histone acetylation in placentas from mothers who regularly include fish or olive oil in their diets could influence immune priming in the newborn.
Collapse
|
119
|
Abbott DH, Dumesic DA, Levine JE. Hyperandrogenic origins of polycystic ovary syndrome - implications for pathophysiology and therapy. Expert Rev Endocrinol Metab 2019; 14:131-143. [PMID: 30767580 PMCID: PMC6992448 DOI: 10.1080/17446651.2019.1576522] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 01/28/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) diagnosis comprises combinations of female hyperandrogenism, menstrual irregularity and polycystic ovaries. While it is a familial and highly prevalent endocrine disorder, progress towards a cure is hindered by absence of a definitive pathogenic mechanism and lack of an animal model of naturally occurring PCOS. AREAS COVERED These include an overview of PCOS and its potential etiology, and an examination of insights gained into its pathogenic origins. Animal models derived from experimentally-induced hyperandrogenism during gestation, or from naturally-occurring PCOS-like traits, most reliably demonstrate reproductive, neuroendocrine and metabolic pathogenesis. EXPERT OPINION Genetic studies, while identifying at least 17 PCOS risk genes, account for <10% of women with PCOS. A number of PCOS risk genes involve regulation of gonadotropin secretion or action, suggesting a reproductive neuroendocrine basis for PCOS pathogenesis. Consistent with this notion, a number of animal models employing fetal androgen excess demonstrate epigenetic induction of PCOS-like traits, including reproductive neuroendocrine and metabolic dysfunction. Monkey models are most comprehensive, while mouse models provide molecular insight, including identifying the androgen receptor, particularly in neurons, as mediating androgen-induced PCOS-like programming. Naturally-occurring female hyperandrogenism is also demonstrated in monkeys. Animal models are poised to delineate molecular gateways to PCOS pathogenesis.
Collapse
Affiliation(s)
- David H Abbott
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI, USA
- Department of Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, USA
| | - Daniel A Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Jon E Levine
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI, USA
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
120
|
Graham AM, Rasmussen JM, Entringer S, Ben Ward E, Rudolph MD, Gilmore JH, Styner M, Wadhwa PD, Fair DA, Buss C. Maternal Cortisol Concentrations During Pregnancy and Sex-Specific Associations With Neonatal Amygdala Connectivity and Emerging Internalizing Behaviors. Biol Psychiatry 2019; 85:172-181. [PMID: 30122286 PMCID: PMC6632079 DOI: 10.1016/j.biopsych.2018.06.023] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/06/2018] [Accepted: 06/22/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND Maternal cortisol during pregnancy has the potential to influence rapidly developing fetal brain systems that are commonly altered in neurodevelopmental and psychiatric disorders. Research examining maternal cortisol concentrations across pregnancy and offspring neurodevelopment proximal to birth is needed to advance understanding in this area and lead to insight into the etiology of these disorders. METHODS Participants were 70 adult women recruited during early pregnancy and their infants born after 34 weeks gestation. Maternal cortisol concentrations were assessed serially over 4 days in early, mid, and late gestation. Resting state functional connectivity magnetic resonance imaging of the neonatal amygdala was examined. Mothers reported on children's internalizing behavior problems at 24 months of age. RESULTS Maternal cortisol concentrations during pregnancy were significantly associated with neonatal amygdala connectivity in a sex-specific manner. Elevated maternal cortisol was associated with stronger amygdala connectivity to brain regions involved in sensory processing and integration, as well as the default mode network in girls, and with weaker connectivity to these brain regions in boys. Elevated maternal cortisol was associated with higher internalizing symptoms in girls only, and this association was mediated by stronger neonatal amygdala connectivity. CONCLUSIONS Normative variation in maternal cortisol during pregnancy is associated with the coordinated functioning of the amygdala soon after birth in a sex-specific manner. The identified pathway from maternal cortisol to higher internalizing symptoms in girls via alterations in neonatal amygdala connectivity may be relevant for the etiology of sex differences in internalizing psychiatric disorders, which are more prevalent in women.
Collapse
Affiliation(s)
- Alice M Graham
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| | - Jerod M Rasmussen
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, California
| | - Sonja Entringer
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, California; Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Medical Psychology, Berlin, Germany
| | - Elizabeth Ben Ward
- Department of Computer Science, University of California, Irvine, Irvine, California
| | - Marc D Rudolph
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, North Carolina
| | - John H Gilmore
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - Martin Styner
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - Pathik D Wadhwa
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, California.
| | - Damien A Fair
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon; Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon
| | - Claudia Buss
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, California; Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Medical Psychology, Berlin, Germany
| |
Collapse
|
121
|
Becker JB, Chartoff E. Sex differences in neural mechanisms mediating reward and addiction. Neuropsychopharmacology 2019; 44:166-183. [PMID: 29946108 PMCID: PMC6235836 DOI: 10.1038/s41386-018-0125-6] [Citation(s) in RCA: 298] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/27/2018] [Accepted: 06/11/2018] [Indexed: 12/18/2022]
Abstract
There is increasing evidence in humans and laboratory animals for biologically based sex differences in every phase of drug addiction: acute reinforcing effects, transition from occasional to compulsive use, withdrawal-associated negative affective states, craving, and relapse. There is also evidence that many qualitative aspects of the addiction phases do not differ significantly between males and females, but one sex may be more likely to exhibit a trait than the other, resulting in population differences. The conceptual framework of this review is to focus on hormonal, chromosomal, and epigenetic organizational and contingent, sex-dependent mechanisms of four neural systems that are known-primarily in males-to be key players in addiction: dopamine, mu-opioid receptors (MOR), kappa opioid receptors (KOR), and brain-derived neurotrophic factor (BDNF). We highlight data demonstrating sex differences in development, expression, and function of these neural systems as they relate-directly or indirectly-to processes of reward and addictive behavior, with a focus on psychostimulants and opioids. We identify gaps in knowledge about how these neural systems interact with sex to influence addictive behavior, emphasizing throughout that the impact of sex can be highly nuanced and male/female data should be reported regardless of the outcome.
Collapse
Affiliation(s)
- Jill B Becker
- Department of Psychology and the Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Elena Chartoff
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
122
|
Goldstein JM, Hale T, Foster SL, Tobet SA, Handa RJ. Sex differences in major depression and comorbidity of cardiometabolic disorders: impact of prenatal stress and immune exposures. Neuropsychopharmacology 2019; 44:59-70. [PMID: 30030541 PMCID: PMC6235859 DOI: 10.1038/s41386-018-0146-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/15/2018] [Accepted: 06/21/2018] [Indexed: 12/11/2022]
Abstract
Major depressive disorder topped ischemic heart disease as the number one cause of disability worldwide in 2012, and women have twice the risk of men. Further, the comorbidity of depression and cardiometabolic disorders will be one of the primary causes of disability worldwide by 2020, with women at twice the risk. Thus, understanding the sex-dependent comorbidities has public health consequences worldwide. We propose here that sex differences in MDD-cardiometabolic comorbidity originate, in part, from pathogenic processes initiated in fetal development that involve sex differences in shared pathophysiology between the brain, the vascular system, the CNS control of the heart and associated hormonal, immune, and metabolic physiology. Pathways implicate neurotrophic and angiogenic growth factors, gonadal hormone receptors, and neurotransmitters such as gamma amino butyric acid (GABA) on neuronal and vascular development of HPA axis regions, such as the paraventricular nucleus (PVN), in addition to blood pressure, in part through the renin-angiotensin system, and insulin and glucose metabolism. We show that the same prenatal exposures have consequences for sex differences across multiple organ systems that, in part, share common pathophysiology. Thus, we believe that applying a sex differences lens to understanding shared biologic substrates underlying these comorbidities will provide novel insights into the development of sex-dependent therapeutics. Further, taking a lifespan perspective beginning in fetal development provides the opportunity to target abnormalities early in the natural history of these disorders in a sex-dependent way.
Collapse
Affiliation(s)
- Jill M Goldstein
- Departments of Psychiatry and Obstetrics and Gynecology, Massachusetts General Hospital (MGH), Boston, MA, 02120, USA.
- Departments of Psychiatry and Medicine, Harvard Medical School, Boston, MA, USA.
| | - Taben Hale
- Department of Basic Medical Science, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, 85004, USA
| | - Simmie L Foster
- Department of Psychiatry, Harvard Medical School, at Massachusetts General Hospital, Boston, MA, USA
| | - Stuart A Tobet
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
| | - Robert J Handa
- Department of Basic Medical Science, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, 85004, USA
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| |
Collapse
|
123
|
Diz-Chaves Y, Toba L, Fandiño J, González-Matías LC, Garcia-Segura LM, Mallo F. The GLP-1 analog, liraglutide prevents the increase of proinflammatory mediators in the hippocampus of male rat pups submitted to maternal perinatal food restriction. J Neuroinflammation 2018; 15:337. [PMID: 30518432 PMCID: PMC6282252 DOI: 10.1186/s12974-018-1370-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/18/2018] [Indexed: 12/17/2022] Open
Abstract
Background Perinatal maternal malnutrition is related to altered growth of tissues and organs. The nervous system development is very sensitive to environmental insults, being the hippocampus a vulnerable structure, in which altered number of neurons and granular cells has been observed. Moreover, glial cells are also affected, and increased expression of proinflammatory mediators has been observed. We studied the effect of Glucagon-like peptide-1 receptor (GLP-1R) agonists, liraglutide, which have very potent metabolic and neuroprotective effects, in order to ameliorate/prevent the glial alterations present in the hippocampus of the pups from mothers with food restriction during pregnancy and lactation (maternal perinatal food restriction—MPFR). Methods Pregnant Sprague-Dawley rats were randomly assigned to 50% food restriction (FR; n = 12) or ad libitum controls (CT, n = 12) groups at day of pregnancy 12 (GD12). From GD14 to parturition, pregnant FR and CT rats were treated with liraglutide (100 μg/kg) or vehicle. At postnatal day 21 and before weaning, 48 males and 45 females (CT and MPFR) were sacrificed. mRNA expression levels of interleukin-1β (IL1β), interleukin-6 (IL-6), nuclear factor-κβ, major histocompatibility complex-II (MHCII), interleukin 10 (IL10), arginase 1 (Arg1), and transforming growth factor (TGFβ) were assessed in the hippocampus by quantitative real-time polymerase chain reaction. Iba1 and GFAP-immunoreactivity were assessed by immunocytochemistry. Results The mRNA expression IL1β, IL6, NF-κB, and MHCII increased in the hippocampus of male but not in female pups from MPFR. In addition, there was an increase in the percentage of GFAP and Iba1-immupositive cells in the dentate gyrus compared to controls, indicating an inflammatory response in the brain. On the other hand, liraglutide treatment prevented the neuroinflammatory process, promoting the production of anti-inflammatory molecules such as IL10, TGFβ, and arginase 1, and decreasing the number and reactivity of microglial cells and astrocytes in the hippocampus of male pups. Conclusion Therefore, the GLP-1 analog, liraglutide, emerges as neuroprotective drug that minimizes the harmful effects of maternal food restriction, decreasing neuroinflammation in the hippocampus in a very early stage.
Collapse
Affiliation(s)
- Y Diz-Chaves
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain.
| | - L Toba
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| | - J Fandiño
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| | - L C González-Matías
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| | - L M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, E-28002, Madrid, Spain.,Centro de Investigación en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - F Mallo
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| |
Collapse
|
124
|
Placenta-specific 9, a putative secretory protein, induces G2/M arrest and inhibits the proliferation of human embryonic hepatic cells. Biosci Rep 2018; 38:BSR20180820. [PMID: 30291214 PMCID: PMC6239258 DOI: 10.1042/bsr20180820] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/21/2018] [Accepted: 10/04/2018] [Indexed: 01/10/2023] Open
Abstract
Background: Placenta-specific 9 (Plac9) is a putative secreted protein that was first discovered in the context of embryogenesis. The expression pattern of Plac9 during embryogenesis, together with the results of recent reports, suggest that Plac9 may play a role in the liver development. The present study was conducted to investigate the secretory characteristics of Plac9 and its potential role in liver cell physiology. Methods: Immunofluorescence was employed to identify the subcellular distribution of Plac9. Cellular proliferative activity was analyzed by MTT assay and cell colony formation. The cell cycle distribution of Plac9 was analyzed by flow cytometry, and a functional analysis was performed using L02 cells following their stable infection with a lentivirus over-expressing Plac9. Results:Plac9 is a novel protein that is localized to the cytoplasm and may be secreted through the classic endoplasmic reticulum-Golgi route. The overexpression of Plac9 inhibits cell growth and induces G2/M phase arrest. Conclusion: Our findings reveal a novel role for Plac9 in regulating cell growth.
Collapse
|
125
|
Schroeder M, Jakovcevski M, Polacheck T, Drori Y, Ben-Dor S, Röh S, Chen A. Sex dependent impact of gestational stress on predisposition to eating disorders and metabolic disease. Mol Metab 2018; 17:1-16. [PMID: 30174229 PMCID: PMC6197785 DOI: 10.1016/j.molmet.2018.08.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/08/2018] [Accepted: 08/14/2018] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Vulnerability to eating disorders (EDs) is broadly assumed to be associated with early life stress. However, a careful examination of the literature shows that susceptibility to EDs may depend on the type, severity and timing of the stressor and the sex of the individual. We aimed at exploring the link between chronic prenatal stress and predisposition to EDs and metabolic disease. METHODS We used a chronic variable stress protocol during gestation to explore the metabolic response of male and female offspring to food restriction (FR), activity-based anorexia (ABA), binge eating (BE) and exposure to high fat (HF) diet. RESULTS Contrary to controls, prenatally stressed (PNS) female offspring showed resistance to ABA and BE and displayed a lower metabolic rate leading to hyperadiposity and obesity on HF diet. Male PNS offspring showed healthy responses to FR and ABA, increased propensity to binge and improved coping with HF compared to controls. We found that long-lasting abnormal responses to metabolic challenge are linked to fetal programming and adult hypothalamic dysregulation in PNS females, resulting from sexually dimorphic adaptations in placental methylation and gene expression. CONCLUSIONS Our results show that maternal stress may have variable and even opposing effects on ED risk, depending on the ED and the sex of the offspring.
Collapse
Affiliation(s)
- Mariana Schroeder
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, 80804, Germany.
| | - Mira Jakovcevski
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, 80804, Germany
| | - Tamar Polacheck
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Yonat Drori
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Shifra Ben-Dor
- Bioinformatics and Biological Computing Unit, Biological Services, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Simone Röh
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, 80804, Germany
| | - Alon Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, 80804, Germany.
| |
Collapse
|
126
|
Signorile PG, Severino A, Santoro M, Spyrou M, Viceconte R, Baldi A. Methylation analysis of HOXA10 regulatory elements in patients with endometriosis. BMC Res Notes 2018; 11:722. [PMID: 30309386 PMCID: PMC6182800 DOI: 10.1186/s13104-018-3836-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/09/2018] [Indexed: 11/30/2022] Open
Abstract
Objective The pathogenesis of endometriosis is still mysterious, being retrograde menstruation and coelomic metaplasia the most accepted hypotheses. Recently, it has been proposed that endometriosis is caused by fine-tuning alterations of the female genital system development during the foetal life and that in utero exposition to endocrine disruptors can be one of the factors causing the disease, possibly acting on the methylation status of the genome. In this study, we have evaluated the methylation status of HOXA10 gene regulation regions in a cohort of 22 endometriosis patients respect to a control group of 6 healthy women. Results The methylation study was carried out on three CpG islands, previously described hypermethylated in the endometrium of endometriosis patients and include 22 CpG sites, 21 CpG sites and 10 CpG sites, respectively identified through the online platform MethPrimer. The analysis did not find significant differences between patients with endometriosis and healthy control individuals. These results confirm previous studies on genome wide methylation analysis in endometriosis patients. Therefore, other epigenetically altered genes should be considered more related to the pathogenesis of endometriosis.
Collapse
Affiliation(s)
| | - Anna Severino
- Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | | | | | | | - Alfonso Baldi
- Fondazione Italiana Endometriosi, Rome, Italy. .,Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy.
| |
Collapse
|
127
|
Yehuda R, Lehrner A. Intergenerational transmission of trauma effects: putative role of epigenetic mechanisms. World Psychiatry 2018; 17:243-257. [PMID: 30192087 PMCID: PMC6127768 DOI: 10.1002/wps.20568] [Citation(s) in RCA: 231] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 06/13/2018] [Accepted: 06/13/2018] [Indexed: 12/18/2022] Open
Abstract
This paper reviews the research evidence concerning the intergenerational transmission of trauma effects and the possible role of epigenetic mechanisms in this transmission. Two broad categories of epigenetically mediated effects are highlighted. The first involves developmentally programmed effects. These can result from the influence of the offspring's early environmental exposures, including postnatal maternal care as well as in utero exposure reflecting maternal stress during pregnancy. The second includes epigenetic changes associated with a preconception trauma in parents that may affect the germline, and impact fetoplacental interactions. Several factors, such as sex-specific epigenetic effects following trauma exposure and parental developmental stage at the time of exposure, explain different effects of maternal and paternal trauma. The most compelling work to date has been done in animal models, where the opportunity for controlled designs enables clear interpretations of transmissible effects. Given the paucity of human studies and the methodological challenges in conducting such studies, it is not possible to attribute intergenerational effects in humans to a single set of biological or other determinants at this time. Elucidating the role of epigenetic mechanisms in intergenerational effects through prospective, multi-generational studies may ultimately yield a cogent understanding of how individual, cultural and societal experiences permeate our biology.
Collapse
Affiliation(s)
- Rachel Yehuda
- James J. Peters Bronx Veterans Affairs Hospital, Bronx, NY, USA
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amy Lehrner
- James J. Peters Bronx Veterans Affairs Hospital, Bronx, NY, USA
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
128
|
Abstract
AbstractThe question of whether and how the effects of cultural trauma can be transmitted intergenerationally from parents to offspring, or even to later generations, has evoked interest and controversy in academic and popular forums. Recent methodological advances have spurred investigations of potential epigenetic mechanisms for this inheritance, representing an exciting area of emergent research. Epigenetics has been described as the means through which environmental influences “get under the skin,” directing transcriptional activity and influencing the expression or suppression of genes. Over the past decade, this complex environment–biology interface has shown increasing promise as a potential pathway for the intergenerational transmission of the effects of trauma. This article reviews challenges facing research on cultural trauma, biological findings in trauma and posttraumatic stress disorder, and putative epigenetic mechanisms for transmission of trauma effects, including through social, intrauterine, and gametic pathways. Implications for transmission of cultural trauma effects are discussed, focused on the relevance of cultural narratives and the possibilities of resilience and adaptivity.
Collapse
|
129
|
Prenatal exposure to TiO 2 nanoparticles in mice causes behavioral deficits with relevance to autism spectrum disorder and beyond. Transl Psychiatry 2018; 8:193. [PMID: 30237468 PMCID: PMC6148221 DOI: 10.1038/s41398-018-0251-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/23/2018] [Accepted: 08/07/2018] [Indexed: 02/07/2023] Open
Abstract
Environmental factors are involved in the etiology of autism spectrum disorder (ASD) and may contribute to the raise in its incidence rate. It is currently unknown whether the increasing use of nanoparticles such as titanium dioxide (TiO2 NPs) in consumer products and biomedical applications may play a role in these associations. While nano-sized TiO2 is generally regarded as safe and non-toxic, excessive exposure to TiO2 NPs may be associated with negative health consequences especially when occurring during sensitive developmental periods. To test if prenatal exposure to TiO2 NPs alters fetal development and behavioral functions relevant to ASD, C57Bl6/N dams were subjected to a single intravenous injection of a low (100 µg) or high (1000 µg) dose of TiO2 NPs or vehicle solution on gestation day 9. ASD-related behavioral functions were assessed in the offspring using paradigms that index murine versions of ASD symptoms. Maternal exposure to TiO2 NPs led to subtle and dose-dependent impairments in neonatal vocal communication and juvenile sociability, as well as a dose-dependent increase in prepulse inhibition of the acoustic startle reflex of both sexes. These behavioral alterations emerged in the absence of pregnancy complications. Prenatal exposure to TiO2 NPs did not cause overt fetal malformations or changes in pregnancy outcomes, nor did it affect postnatal growth of the offspring. Taken together, our study provides a first set of preliminary data suggesting that prenatal exposure to nano-sized TiO2 can induce behavioral deficits relevant to ASD and related neurodevelopmental disorders without inducing major changes in physiological development. If extended further, our preclinical findings may provide an incentive for epidemiological studies examining the role of prenatal TiO2 NPs exposure in the etiology of ASD and other neurodevelopmental disorders.
Collapse
|
130
|
Brunst KJ, Tignor N, Just A, Liu Z, Lin X, Hacker MR, Bosquet Enlow M, Wright RO, Wang P, Baccarelli AA, Wright RJ. Cumulative lifetime maternal stress and epigenome-wide placental DNA methylation in the PRISM cohort. Epigenetics 2018; 13:665-681. [PMID: 30001177 DOI: 10.1080/15592294.2018.1497387] [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] [Indexed: 12/21/2022] Open
Abstract
Evolving evidence links maternal stress exposure to changes in placental DNA methylation of specific genes regulating placental function that may have implications for the programming of a host of chronic disorders. Few studies have implemented an epigenome-wide approach. Using the Infinium HumanMethylation450 BeadChip (450K), we investigated epigenome-wide placental DNA methylation in relation to maternal experiences of traumatic and non-traumatic stressors over her lifetime assessed using the Life Stressor Checklist-Revised (LSC-R) survey (n = 207). We found differential DNA methylation at epigenome-wide statistical significance (FDR = 0.05) for 112 CpGs. Additionally, we observed three clusters that exhibited differential methylation in response to high maternal lifetime stress. Enrichment analyses, conducted at an FDR = 0.20, revealed lysine degradation to be the most significant pathway associated with maternal lifetimes stress exposure. Targeted enrichment analyses of the three largest clusters of probes, identified using the gap statistic, were enriched for genes associated with endocytosis (i.e., SMAP1, ANKFY1), tight junctions (i.e., EPB41L4B), and metabolic pathways (i.e., INPP5E, EEF1B2). These pathways, also identified in the top 10 KEGG pathways associated with maternal lifetime stress exposure, play important roles in multiple physiological functions necessary for proper fetal development. Further, two genes were identified to exhibit multiple probes associated with maternal lifetime stress (i.e., ANKFY1, TM6SF1). The methylation status of the probes belonging to each cluster and/or genes exhibiting multiple hits, may play a role in the pathogenesis of adverse health outcomes in children born to mothers with increased lifetime stress exposure.
Collapse
Affiliation(s)
- Kelly J Brunst
- a Department of Environmental Health , University of Cincinnati College of Medicine , Cincinnati , OH , USA
| | - Nicole Tignor
- b Icahn Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences , Icahn School of Medicine at Mount Sinai One Gustave L. Levy Place , New York , NY , USA
| | - Allan Just
- c Department of Environmental Medicine and Public Health , Icahn School of Medicine at Mount Sinai , New York , NY , USA
| | - Zhonghua Liu
- d Department of Biostatistics , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Xihong Lin
- d Department of Biostatistics , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Michele R Hacker
- e Department of Obstetrics and Gynecology , Beth Israel Deaconess Medical Center , Boston , MA , USA.,f Department of Obstetrics , Gynecology and Reproductive Biology, Harvard Medical School , Boston , MA , USA
| | - Michelle Bosquet Enlow
- g Department of Psychiatry, Program for Behavioral Science, Boston Children's Hospital and Department of Psychiatry , Harvard Medical School , Boston , MA , USA
| | - Robert O Wright
- c Department of Environmental Medicine and Public Health , Icahn School of Medicine at Mount Sinai , New York , NY , USA
| | - Pei Wang
- b Icahn Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences , Icahn School of Medicine at Mount Sinai One Gustave L. Levy Place , New York , NY , USA
| | - Andrea A Baccarelli
- h Department of Environmental Health Sciences , Mailman School of Public Health, Columbia University , New York , NY , USA
| | - Rosalind J Wright
- c Department of Environmental Medicine and Public Health , Icahn School of Medicine at Mount Sinai , New York , NY , USA.,i Department of Pediatrics , Kravis Children's Hospital, Icahn School of Medicine at Mount Sinai , New York , NY , USA
| |
Collapse
|
131
|
Stroud LR, McCallum M, Salisbury AL. Impact of maternal prenatal smoking on fetal to infant neurobehavioral development. Dev Psychopathol 2018; 30:1087-1105. [PMID: 30068428 PMCID: PMC6541397 DOI: 10.1017/s0954579418000676] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite recent emphasis on the profound importance of the fetal environment in "programming" postnatal development, measurement of offspring development typically begins after birth. Using a novel coding strategy combining direct fetal observation via ultrasound and actocardiography, we investigated the impact of maternal smoking during pregnancy (MSDP) on fetal neurobehavior; we also investigated links between fetal and infant neurobehavior. Participants were 90 pregnant mothers and their infants (52 MSDP-exposed; 51% minorities; ages 18-40). Fetal neurobehavior at baseline and in response to vibro-acoustic stimulus was assessed via ultrasound and actocardiography at M = 35 weeks gestation and coded via the Fetal Neurobehavioral Assessment System (FENS). After delivery, the NICU Network Neurobehavioral Scale was administered up to seven times over the first postnatal month. MSDP was associated with increased fetal activity and fetal limb movements. Fetal activity, complex body movements, and cardiac-somatic coupling were associated with infants' ability to attend to stimuli and to self-regulate over the first postnatal month. Furthermore, differential associations emerged by MSDP group between fetal activity, complex body movements, quality of movement, and coupling, and infant attention and self-regulation. The present study adds to a growing literature establishing the validity of fetal neurobehavioral measures in elucidating fetal programming pathways.
Collapse
|
132
|
Erbas O, Erdogan MA, Khalilnezhad A, Gürkan FT, Yiğittürk G, Meral A, Taskiran D. Neurobehavioral effects of long‐term maternal fructose intake in rat offspring. Int J Dev Neurosci 2018; 69:68-79. [DOI: 10.1016/j.ijdevneu.2018.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 07/03/2018] [Accepted: 07/04/2018] [Indexed: 01/29/2023] Open
Affiliation(s)
- Oytun Erbas
- Istanbul Bilim University School of MedicineDepartment of PhysiologyIstanbulTurkey
| | | | | | | | - Gürkan Yiğittürk
- Ege University School of MedicineDepartment of Histology and EmbryologyIzmirTurkey
| | - Ayfer Meral
- Dumlupinar University School of MedicineDepartment of BiochemistryKütahyaTurkey
| | - Dilek Taskiran
- Ege University School of MedicineDepartment of PhysiologyIzmirTurkey
| |
Collapse
|
133
|
Placental H3K27me3 establishes female resilience to prenatal insults. Nat Commun 2018; 9:2555. [PMID: 29967448 PMCID: PMC6028627 DOI: 10.1038/s41467-018-04992-1] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 06/07/2018] [Indexed: 12/13/2022] Open
Abstract
Although sex biases in disease presentation are well documented, the mechanisms mediating vulnerability or resilience to diseases are unknown. In utero insults are more likely to produce detrimental health outcomes for males versus females. In our mouse model of prenatal stress, male offspring experience long-term dysregulation of body weight and hypothalamic pituitary adrenal stress axis dysfunction, endophenotypes of male-biased neurodevelopmental disorders. Placental function is critical for healthy fetal development, and we previously showed that sex differences in placental O-linked N-acetylglucosamine transferase (OGT) mediate the effects of prenatal stress on neurodevelopmental programming. Here we show that one mechanism whereby sex differences in OGT confer variation in vulnerability to prenatal insults is by establishing sex-specific trophoblast gene expression patterns and via regulation of the canonically repressive epigenetic modification, H3K27me3. We demonstrate that high levels of H3K27me3 in the female placenta create resilience to the altered hypothalamic programming associated with prenatal stress exposure. Sex differences in placental O-linked N-acetylglucosamine transferase (OGT) activity mediate the effects of prenatal stress on neurodevelopmental programming. Here authors provide evidence that OGT confers variation in vulnerability to prenatal insults by establishing sex-specific trophoblast gene expression via regulation of H3K27me3.
Collapse
|
134
|
Chan JC, Nugent BM, Bale TL. Parental Advisory: Maternal and Paternal Stress Can Impact Offspring Neurodevelopment. Biol Psychiatry 2018; 83:886-894. [PMID: 29198470 PMCID: PMC5899063 DOI: 10.1016/j.biopsych.2017.10.005] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 09/07/2017] [Accepted: 10/03/2017] [Indexed: 12/16/2022]
Abstract
Parental stress exposures are implicated in the risk for offspring neurodevelopmental and neuropsychiatric disorders, prompting critical examination of preconception and prenatal periods as vulnerable to environmental insults such as stress. Evidence from human studies and animal models demonstrates the influence that both maternal and paternal stress exposures have in changing the course of offspring brain development. Mechanistic examination of modes of intergenerational transmission of exposure during pregnancy has pointed to alterations in placental signaling, including changes in inflammatory, nutrient-sensing, and epigenetic pathways. Transmission of preconception paternal stress exposure is associated with changes in epigenetic marks in sperm, with a primary focus on the reprogramming of DNA methylation, histone posttranslational modifications, and small noncoding RNAs. In this review, we discuss evidence supporting the important contribution of intergenerational parental stress in offspring neurodevelopment and disease risk, and the currently known epigenetic mechanisms underlying this transmission.
Collapse
Affiliation(s)
- Jennifer C Chan
- Department of Biomedical Sciences, School of Veterinary Medicine and Perelman School of Medicine University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bridget M Nugent
- Department of Pharmacology, University of Maryland School of Medicine, University of Maryland, Baltimore, Maryland
| | - Tracy L Bale
- Department of Pharmacology, University of Maryland School of Medicine, University of Maryland, Baltimore, Maryland.
| |
Collapse
|
135
|
Placental miR-340 mediates vulnerability to activity based anorexia in mice. Nat Commun 2018; 9:1596. [PMID: 29686286 PMCID: PMC5913294 DOI: 10.1038/s41467-018-03836-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 03/15/2018] [Indexed: 12/20/2022] Open
Abstract
Anorexia nervosa (AN) is a devastating eating disorder characterized by self-starvation that mainly affects women. Its etiology is unknown, which impedes successful treatment options leading to a limited chance of full recovery. Here, we show that gestation is a vulnerable window that can influence the predisposition to AN. By screening placental microRNA expression of naive and prenatally stressed (PNS) fetuses and assessing vulnerability to activity-based anorexia (ABA), we identify miR-340 as a sexually dimorphic regulator involved in prenatal programming of ABA. PNS caused gene-body hypermethylation of placental miR-340, which is associated with reduced miR-340 expression and increased protein levels of several target transcripts, GR, Cry2 and H3F3b. MiR-340 is linked to the expression of several nutrient transporters both in mice and human placentas. Using placenta-specific lentiviral transgenes and embryo transfer, we demonstrate the key role miR-340 plays in the mechanism involved in early life programming of ABA. Anorexia nervosa is characterised by self-starvation but its etiology is not completely understood. Here the authors describe how prenatal stress can induce activity-based anorexia in the offspring during early adulthood by upregulating miR-340 expression in the placenta that affects expression of nutrient transporters.
Collapse
|
136
|
Yehuda R, Lehrner A, Bierer LM. The public reception of putative epigenetic mechanisms in the transgenerational effects of trauma. ENVIRONMENTAL EPIGENETICS 2018; 4:dvy018. [PMID: 30038801 PMCID: PMC6051458 DOI: 10.1093/eep/dvy018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/23/2018] [Indexed: 06/08/2023]
Abstract
There has been great interest in the possibility that effects of trauma might be passed from parent to offspring through epigenetic mechanisms. This topic has stimulated discussion and controversy in the scientific literature, the popular press, and culture at large. This article describes the initial observations that have led to recent examinations of epigenetic mechanisms in association with effects of parental trauma exposure on offspring. Epigenetic research in animals has provided models for how such effects might be transmitted. However, the attribution of any specific epigenetic mechanisms in human studies of offspring of trauma survivors is premature at this time. The article describes some of the ways in which initial epigenetic findings in the offspring of trauma survivors have been represented in the popular media. Reports have ranged from overly simplistic and sensationalistic claims to global dismissals. The authors discuss the importance of clarity in language when describing epigenetic findings for lay audiences, the need to emphasize the limitations as well as the promise of research on intergenerational transmission of trauma effects, and the importance of countering popular interpretations that imply a reductionist biological determinism. Scientists have an obligation to assist in translating important research findings and nascent avenues of research to the public. It is important to recognize the ways in which this research may unintentionally be received as supporting a narrative of permanent and significant damage in offspring, rather than contributing to discussions of potential resilience, adaptability, and mutability in biological systems affected by stress.
Collapse
Affiliation(s)
- Rachel Yehuda
- James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amy Lehrner
- James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Linda M Bierer
- James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
137
|
O-GlcNAc in cancer: An Oncometabolism-fueled vicious cycle. J Bioenerg Biomembr 2018; 50:155-173. [PMID: 29594839 DOI: 10.1007/s10863-018-9751-2] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 03/15/2018] [Indexed: 12/17/2022]
Abstract
Cancer cells exhibit unregulated growth, altered metabolism, enhanced metastatic potential and altered cell surface glycans. Fueled by oncometabolism and elevated uptake of glucose and glutamine, the hexosamine biosynthetic pathway (HBP) sustains glycosylation in the endomembrane system. In addition, the elevated pools of UDP-GlcNAc drives the O-GlcNAc modification of key targets in the cytoplasm, nucleus and mitochondrion. These targets include transcription factors, kinases, key cytoplasmic enzymes of intermediary metabolism, and electron transport chain complexes. O-GlcNAcylation can thereby alter epigenetics, transcription, signaling, proteostasis, and bioenergetics, key 'hallmarks of cancer'. In this review, we summarize accumulating evidence that many cancer hallmarks are linked to dysregulation of O-GlcNAc cycling on cancer-relevant targets. We argue that onconutrient and oncometabolite-fueled elevation increases HBP flux and triggers O-GlcNAcylation of key regulatory enzymes in glycolysis, Kreb's cycle, pentose-phosphate pathway, and the HBP itself. The resulting rerouting of glucose metabolites leads to elevated O-GlcNAcylation of oncogenes and tumor suppressors further escalating elevation in HBP flux creating a 'vicious cycle'. Downstream, elevated O-GlcNAcylation alters DNA repair and cellular stress pathways which influence oncogenesis. The elevated steady-state levels of O-GlcNAcylated targets found in many cancers may also provide these cells with a selective advantage for sustained growth, enhanced metastatic potential, and immune evasion in the tumor microenvironment.
Collapse
|
138
|
Rezaei M, Eskandari F, Mohammadpour-Gharehbagh A, Harati-Sadegh M, Teimoori B, Salimi S. Hypomethylation of the miRNA-34a gene promoter is associated with Severe Preeclampsia. Clin Exp Hypertens 2018; 41:1-5. [PMID: 29557690 DOI: 10.1080/10641963.2018.1451534] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 02/26/2018] [Accepted: 02/26/2018] [Indexed: 01/08/2023]
Abstract
PURPOSE PE is a pregnancy-specific complication, which genetic and epigenetic factors play key roles in its pathogenesis. DNA methylation is a main epigenetic alteration with important roles in gene regulation. Micro RNAs (miRNAs) as another member of epigenetic machinery regulate the gene expression and involve in different biological pathways including apoptosis and placental development. Therefore, the present study performed to assess the association between miRNA-34a promoter methylation and PE susceptibility. METHODS The placenta of 104 PE pregnant women and 119 normotensive pregnant women were collected after delivery. The methylation status of the miRNA-34a promoter was assessed using Methylation Specific PCR (MSP). RESULTS The frequency of the hemi-methylated (MU) miR-34a promoter was significantly lower in PE women compared to the controls (17.3 vs. 29.4%) (OR, 0.45 [95% CI, 0.2-0.9], P = 0.016). The overall methylation rate was 23.1% in PE women and 41.2% in the control group and was significantly lower in PE women (OR, 0.4 [95% CI, 0.2-0.8], P = 0.004). The frequency of hemi-methylated (MU) and overall methylated (MU+MM) promoter of miR-34a gene was significantly lower in severe PE but not in mild PE women compared to the controls [(OR, 0.3 [95% CI, 0.1-0.8], P = 0.02) and (OR, 0.3 [95% CI, 0.1-0.7], P = 0.009), respectively]. There was an association between hemi-methylated (MU) and overall methylated (MU+MM) promoter and late onset PE [(OR, 0.4 [95% CI, 0.2-0.9], P = 0.03) and (OR, 0.4 [95% CI, 0.2-0.8], P = 0.01), respectively]. CONCLUSIONS An association was found between hypo-methylation of the miR-34a promoter and PE and PE severity.
Collapse
Affiliation(s)
- Mahnaz Rezaei
- a Department of Clinical Biochemistry, School of Medicine , Zahedan University of Medical Sciences , Zahedan , Iran
- b Cellular and Molecular Research Center , Zahedan University of Medical Sciences , Zahedan , Iran
| | - Fatemeh Eskandari
- a Department of Clinical Biochemistry, School of Medicine , Zahedan University of Medical Sciences , Zahedan , Iran
- b Cellular and Molecular Research Center , Zahedan University of Medical Sciences , Zahedan , Iran
| | - Abbas Mohammadpour-Gharehbagh
- a Department of Clinical Biochemistry, School of Medicine , Zahedan University of Medical Sciences , Zahedan , Iran
- b Cellular and Molecular Research Center , Zahedan University of Medical Sciences , Zahedan , Iran
| | - Mahdiyeh Harati-Sadegh
- c Department of Genetics, Fars Science and Research Branch , Islamic Azad University , Marvdasht , Iran
- d Department of Genetics, Marvdasht Branch , Islamic Azad University , Marvdasht , Iran
| | - Batool Teimoori
- e Department of Obstetrics and Gynecology, School of Medicine , Zahedan University of Medical Sciences , Zahedan , Iran
| | - Saeedeh Salimi
- a Department of Clinical Biochemistry, School of Medicine , Zahedan University of Medical Sciences , Zahedan , Iran
- b Cellular and Molecular Research Center , Zahedan University of Medical Sciences , Zahedan , Iran
| |
Collapse
|
139
|
Eskandari F, Teimoori B, Rezaei M, Mohammadpour‐Gharehbagh A, Narooei‐Nejad M, Mehrabani M, Salimi S. Relationships between Dicer 1 polymorphism and expression levels in the etiopathogenesis of preeclampsia. J Cell Biochem 2018; 119:5563-5570. [DOI: 10.1002/jcb.26725] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 01/24/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Fatemeh Eskandari
- Department of Clinical BiochemistrySchool of MedicineZahedan University of Medical SciencesZahedanIran
- Cellular and Molecular Research CenterZahedan University of Medical SciencesZahedanIran
| | - Batool Teimoori
- Department of Obstetrics and GynecologySchool of MedicineZahedan University of Medical SciencesZahedanIran
| | - Mahnaz Rezaei
- Department of Clinical BiochemistrySchool of MedicineZahedan University of Medical SciencesZahedanIran
- Cellular and Molecular Research CenterZahedan University of Medical SciencesZahedanIran
| | - Abbas Mohammadpour‐Gharehbagh
- Department of Clinical BiochemistrySchool of MedicineZahedan University of Medical SciencesZahedanIran
- Cellular and Molecular Research CenterZahedan University of Medical SciencesZahedanIran
| | - Mehrnaz Narooei‐Nejad
- Department of Medical GeneticsSchool of MedicineZahedan University of Medical SciencesZahedanIran
| | - Mehrnaz Mehrabani
- Physiology Research CenterInstitute of Basic and Clinical Physiology SciencesKerman University of Medical SciencesKermanIran
| | - Saeedeh Salimi
- Department of Clinical BiochemistrySchool of MedicineZahedan University of Medical SciencesZahedanIran
- Cellular and Molecular Research CenterZahedan University of Medical SciencesZahedanIran
| |
Collapse
|
140
|
Abstract
Background The placenta is the central regulator of maternal and fetal interactions. Perturbations of placental structure and function have been associated with adverse neurodevelopmental outcomes later in life. Placental CpG methylation represents an epigenetic modification with the potential to impact placental function, fetal development and child health later in life. Study design Genome-wide placental CpG methylation levels were compared between spontaneous versus indicated deliveries from extremely preterm births (EPTBs) (n = 84). The association between the identified differentially methylated CpG sites and neurocognitive outcome at ten years of age was then evaluated. Results Spontaneous EPTB was associated with differential CpG methylation levels in 250 CpG sites (217 unique genes) with the majority displaying hypermethylation. The identified genes are known to play a role in neurodevelopment and are enriched for basic helix-loop-helix transcription factor binding sites. The placental CpG methylation levels for 17 of these sites predicted cognitive function at ten years of age. Conclusion A hypermethylation signature is present in DNA from placentas in infants with spontaneous EPTB. CpG methylation levels of critical neurodevelopment genes in the placenta predicted later life cognitive function, supporting the developmental origins of health and disease hypothesis (DOHaD).
Collapse
|
141
|
Brunst KJ, Sanchez-Guerra M, Chiu YHM, Wilson A, Coull BA, Kloog I, Schwartz J, Brennan KJ, Bosquet Enlow M, Wright RO, Baccarelli AA, Wright RJ. Prenatal particulate matter exposure and mitochondrial dysfunction at the maternal-fetal interface: Effect modification by maternal lifetime trauma and child sex. ENVIRONMENT INTERNATIONAL 2018; 112:49-58. [PMID: 29248865 PMCID: PMC6094933 DOI: 10.1016/j.envint.2017.12.020] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 05/23/2023]
Abstract
BACKGROUND Prenatal ambient fine particulate matter (PM2.5) and maternal chronic psychosocial stress have independently been linked to changes in mithochondrial DNA copy number (mtDNAcn), a marker of mitochondrial response and dysfunction. Further, overlapping research shows sex-specific effects of PM2.5 and stress on developmental outcomes. Interactions among PM2.5, maternal stress, and child sex have not been examined in this context. METHODS We examined associations among exposure to prenatal PM2.5, maternal lifetime traumatic stressors, and mtDNAcn at birth in a sociodemographically diverse pregnancy cohort (N=167). Mothers' daily exposure to PM2.5 over gestation was estimated using a satellite-based spatio-temporally resolved prediction model. Lifetime exposure to traumatic stressors was ascertained using the Life Stressor Checklist-Revised; exposure was categorized as high vs. low based on a median split. Quantitative real-time polymerase chain reaction (qPCR) was used to determine mtDNAcn in placenta and cord blood leukocytes. Bayesian Distributed Lag Interaction regression models (BDLIMs) were used to statistically model and visualize the PM2.5 timing-dependent pattern of associations with mtDNAcn and explore effect modification by maternal lifetime trauma and child sex. RESULTS Increased PM2.5 exposure across pregnancy was associated with decreased mtDNAcn in cord blood (cumulative effect estimate=-0.78; 95%CI -1.41, -0.16). Higher maternal lifetime trauma was associated with reduced mtDNAcn in placenta (β=-0.33; 95%CI -0.63, -0.02). Among women reporting low trauma, increased PM2.5 exposure late in pregnancy (30-38weeks gestation) was significantly associated with decreased mtDNAcn in placenta; no significant association was found in the high trauma group. BDLIMs identified a significant 3-way interaction between PM2.5, maternal trauma, and child sex. Specifically, PM2.5 exposure between 25 and 40weeks gestation was significantly associated with increased placental mtDNAcn among boys of mothers reporting high trauma. In contrast, PM2.5 exposure in this same window was significantly associated with decreased placental mtDNAcn among girls of mothers reporting low trauma. Similar 3-way interactive effects were observed in cord blood. CONCLUSIONS These results indicate that joint exposure to PM2.5 in late pregnancy and maternal lifetime trauma influence mtDNAcn at the maternal-fetal interface in a sex-specific manner. Additional studies will assist in understanding if the sex-specific patterns reflect distinct pathophysiological processes in addition to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Kelly J Brunst
- Department of Environmental Health, University of Cincinnati College of Medicine, 160 Panzeca Way, Cincinnati, OH 45267, United States.
| | - Marco Sanchez-Guerra
- Department of Developmental Neurobiology, National Institute of Perinatology, Montes Urales 800, Lomas Virreyes, Mexico City 11000, Mexico.
| | - Yueh-Hsiu Mathilda Chiu
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States.
| | - Ander Wilson
- Department of Statistics, Colorado State University, Fort Collins, CO, United States.
| | - Brent A Coull
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 655 Huntington Ave., Boston, MA 02115, United States.
| | - Itai Kloog
- Department of Geography and Environmental Development, Ben-Gurion University of the Negev, P.O.B 653, Beer Sheva, Israel.
| | - Joel Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, United States.
| | - Kasey J Brennan
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University Medical Center, 722 W 168th St., New York, NY 10032, United States.
| | - Michelle Bosquet Enlow
- Department of Psychiatry, Boston Children's Hospital, 300 Longwood Ave., Boston, MA 02215, United States; Department of Psychiatry, Harvard Medical School, 401 Park Dr., Boston, MA 02215, United States.
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, 17 East 102nd St., New York, NY 10029, United States; Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, 17 East 102nd St., New York, NY 10029, United States.
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University Medical Center, 722 W 168th St., New York, NY 10032, United States.
| | - Rosalind J Wright
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States; Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, 17 East 102nd St., New York, NY 10029, United States.
| |
Collapse
|
142
|
Lobato G, Reichenheim ME, Moraes CL, Peixoto-Filho FM, Migowski LS. Psychologic intimate partner violence and the risk of intrauterine growth restriction in Rio de Janeiro. Int J Gynaecol Obstet 2017; 143:77-83. [DOI: 10.1002/ijgo.12393] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/19/2017] [Accepted: 11/16/2017] [Indexed: 01/07/2023]
Affiliation(s)
- Gustavo Lobato
- Fernandes Figueira Institute; Oswaldo Cruz Foundation (FIOCRUZ); Rio de Janeiro Brazil
| | - Michael E. Reichenheim
- Institute of Social Medicine; State University of Rio de Janeiro (UERJ); Rio de Janeiro Brazil
| | - Claudia L. Moraes
- Institute of Social Medicine; State University of Rio de Janeiro (UERJ); Rio de Janeiro Brazil
| | | | - Lívia S. Migowski
- Fernandes Figueira Institute; Oswaldo Cruz Foundation (FIOCRUZ); Rio de Janeiro Brazil
| |
Collapse
|
143
|
Brunst KJ, Sanchez Guerra M, Gennings C, Hacker M, Jara C, Bosquet Enlow M, Wright RO, Baccarelli A, Wright RJ. Maternal Lifetime Stress and Prenatal Psychological Functioning and Decreased Placental Mitochondrial DNA Copy Number in the PRISM Study. Am J Epidemiol 2017; 186:1227-1236. [PMID: 28595325 PMCID: PMC5859981 DOI: 10.1093/aje/kwx183] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 01/12/2017] [Accepted: 01/26/2017] [Indexed: 12/17/2022] Open
Abstract
Psychosocial stress contributes to placental oxidative stress. Mitochondria are vulnerable to oxidative stress, which can lead to changes in mitochondrial DNA copy number (mtDNAcn). We examined associations of maternal lifetime stress, current negative life events, and depressive and posttraumatic-stress-disorder symptom scores with placental mtDNAcn in a racially/ethnically diverse sample (n = 147) from the Programming of Intergenerational Stress Mechanisms (PRISM) study (Massachusetts, March 2011 to August 2012). In linear regression analyses adjusted for maternal age, race/ethnicity, education, prenatal fine particulate matter exposure, prenatal smoking exposure, and the sex of the child, all measures of stress were associated with decreased placental mtDNAcn (all P values < 0.05). Weighted-quantile-sum (WQS) regression showed that higher lifetime stress and depressive symptoms accounted for most of the effect on mtDNAcn (WQS weights: 0.25 and 0.39, respectively). However, among white individuals, increased lifetime stress and posttraumatic stress disorder symptoms explained the majority of the effect (WQS weights: 0.20 and 0.62, respectively) while among nonwhite individuals, lifetime stress and depressive symptoms accounted for most of the effect (WQS weights: 0.27 and 0.55, respectively). These analyses are first to link increased maternal psychosocial stress with reduced placental mtDNAcn and add to literature documenting racial/ethnic differences in the psychological sequelae of chronic stress that may contribute to maternal-fetal health.
Collapse
Affiliation(s)
- Kelly J Brunst
- Department of Pediatrics, Kravis Children’s Hospital, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Marco Sanchez Guerra
- Laboratory of Environmental Epigenetics, Exposure Epidemiology and Risk Program, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Department of Developmental Neurobiology, National Institute of Perinatology, Mexico City, Mexico
| | - Chris Gennings
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Michele Hacker
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Calvin Jara
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Michelle Bosquet Enlow
- Program for Behavioral Science, Department of Psychiatry, Boston Children’s Hospital, Boston, Massachusetts
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Andrea Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University Medical Center, New York, New York
| | - Rosalind J Wright
- Department of Pediatrics, Kravis Children’s Hospital, Icahn School of Medicine at Mount Sinai, New York, New York
- Institute for Exposomics Research, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
144
|
Akan I, Olivier-Van Stichelen S, Bond MR, Hanover JA. Nutrient-driven O-GlcNAc in proteostasis and neurodegeneration. J Neurochem 2017; 144:7-34. [PMID: 29049853 DOI: 10.1111/jnc.14242] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 09/28/2017] [Accepted: 10/11/2017] [Indexed: 12/14/2022]
Abstract
Proteostasis is essential in the mammalian brain where post-mitotic cells must function for decades to maintain synaptic contacts and memory. The brain is dependent on glucose and other metabolites for proper function and is spared from metabolic deficits even during starvation. In this review, we outline how the nutrient-sensitive nucleocytoplasmic post-translational modification O-linked N-acetylglucosamine (O-GlcNAc) regulates protein homeostasis. The O-GlcNAc modification is highly abundant in the mammalian brain and has been linked to proteopathies, including neurodegenerative diseases such as Alzheimer's, Parkinson's, and Huntington's. C. elegans, Drosophila, and mouse models harboring O-GlcNAc transferase- and O-GlcNAcase-knockout alleles have helped define the role O-GlcNAc plays in development as well as age-associated neurodegenerative disease. These enzymes add and remove the single monosaccharide from protein serine and threonine residues, respectively. Blocking O-GlcNAc cycling is detrimental to mammalian brain development and interferes with neurogenesis, neural migration, and proteostasis. Findings in C. elegans and Drosophila model systems indicate that the dynamic turnover of O-GlcNAc is critical for maintaining levels of key transcriptional regulators responsible for neurodevelopment cell fate decisions. In addition, pathways of autophagy and proteasomal degradation depend on a transcriptional network that is also reliant on O-GlcNAc cycling. Like the quality control system in the endoplasmic reticulum which uses a 'mannose timer' to monitor protein folding, we propose that cytoplasmic proteostasis relies on an 'O-GlcNAc timer' to help regulate the lifetime and fate of nuclear and cytoplasmic proteins. O-GlcNAc-dependent developmental alterations impact metabolism and growth of the developing mouse embryo and persist into adulthood. Brain-selective knockout mouse models will be an important tool for understanding the role of O-GlcNAc in the physiology of the brain and its susceptibility to neurodegenerative injury.
Collapse
Affiliation(s)
- Ilhan Akan
- Laboratory of Cell and Molecular Biology, NIDDK, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Michelle R Bond
- Laboratory of Cell and Molecular Biology, NIDDK, National Institutes of Health, Bethesda, Maryland, USA
| | - John A Hanover
- Laboratory of Cell and Molecular Biology, NIDDK, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
145
|
Goeden N, Notarangelo FM, Pocivavsek A, Beggiato S, Bonnin A, Schwarcz R. Prenatal Dynamics of Kynurenine Pathway Metabolism in Mice: Focus on Kynurenic Acid. Dev Neurosci 2017; 39:519-528. [PMID: 29080891 DOI: 10.1159/000481168] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/31/2017] [Indexed: 01/09/2023] Open
Abstract
The kynurenine pathway (KP), the major catabolic route of tryptophan in mammals, contains several neuroactive metabolites, including kynurenic acid (KYNA) and 3-hydroxykynurenine (3-HK). KP metabolism, and especially the fate of KYNA, during pregnancy is poorly understood, yet it may play a significant role in the development of psychiatric disorders later in life. The present study was designed to investigate the prenatal features of KP metabolism in vivo, with special focus on KYNA. To this end, pregnant CD-1 mice were treated systemically with kynurenine (100 mg/kg), KYNA (10 mg/kg), or saline on embryonic day 18. As expected, administration of either kynurenine or KYNA increased KYNA levels in the maternal plasma and placenta. Maternal kynurenine treatment also raised kynurenine levels in the fetal plasma and brain, demonstrating the ability of this pivotal KP metabolite to cross the placenta and increase the levels of both KYNA and 3-HK in the fetal brain. In contrast, maternal administration of KYNA caused only a small, nonsignificant elevation in KYNA levels in fetal plasma and brain. Complementary experiments using an ex vivo placental perfusion procedure confirmed the significant transplacental transfer of kynurenine and demonstrated that only a very small fraction of maternal kynurenine is converted to KYNA in the placenta and released into the fetal compartment under physiological conditions. Jointly, these results help to clarify the contributions of the maternal circulation and the placenta to fetal KYNA in the late prenatal period.
Collapse
Affiliation(s)
- Nick Goeden
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
146
|
Mohammadpour‐Gharehbagh A, Teimoori B, Narooei‐nejad M, Mehrabani M, Saravani R, Salimi S. The association of the placental MTHFR 3′‐UTR polymorphisms, promoter methylation, and MTHFR expression with preeclampsia. J Cell Biochem 2017; 119:1346-1354. [DOI: 10.1002/jcb.26290] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 07/18/2017] [Indexed: 11/05/2022]
Affiliation(s)
- Abbas Mohammadpour‐Gharehbagh
- Cellular and Molecular Research CenterZahedan University of Medical SciencesZahedanIran
- Department of Clinical BiochemistrySchool of MedicineZahedan University of Medical SciencesZahedanIran
| | - Batool Teimoori
- Department of Obstetrics and GynecologySchool of MedicineZahedan University of Medical SciencesZahedanIran
- Pregnancy Health Research CenterZahedan University of Medical SciencesZahedanIran
| | - Mehrnaz Narooei‐nejad
- Department of Medical GeneticsSchool of MedicineZahedan University of Medical SciencesZahedanIran
| | - Mehrnaz Mehrabani
- Physiology Research Center, Institute of Basic and Clinical Physiology SciencesKerman University of Medical SciencesKermanIran
| | - Ramin Saravani
- Cellular and Molecular Research CenterZahedan University of Medical SciencesZahedanIran
- Department of Clinical BiochemistrySchool of MedicineZahedan University of Medical SciencesZahedanIran
| | - Saeedeh Salimi
- Cellular and Molecular Research CenterZahedan University of Medical SciencesZahedanIran
- Department of Clinical BiochemistrySchool of MedicineZahedan University of Medical SciencesZahedanIran
| |
Collapse
|
147
|
Goldstein JM, Holsen L, Huang G, Hammond BD, James-Todd T, Cherkerzian S, Hale TM, Handa RJ. Prenatal stress-immune programming of sex differences in comorbidity of depression and obesity/metabolic syndrome. DIALOGUES IN CLINICAL NEUROSCIENCE 2017. [PMID: 28179814 PMCID: PMC5286728 DOI: 10.31887/dcns.2016.18.4/jgoldstein] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Major depressive disorder (MDD) is the number one cause of disability worldwide and is comorbid with many chronic diseases, including obesity/metabolic syndrome (MetS). Women have twice as much risk for MDD and comorbidity with obesity/MetS as men, although pathways for understanding this association remain unclear. On the basis of clinical and preclinical studies, we argue that prenatal maternal stress (ie, excess glucocorticoid expression and associated immune responses) that occurs during the sexual differentiation of the fetal brain has sex-dependent effects on brain development within highly sexually dimorphic regions that regulate mood, stress, metabolic function, the autonomic nervous system, and the vasculature. Furthermore, these effects have lifelong consequences for shared sex-dependent risk of MDD and obesity/MetS. Thus, we propose that there are shared biologic substrates at the anatomical, molecular, and/or genetic levels that produce the comorbid risk for MDD-MetS through sex-dependent fetal origins.
Collapse
Affiliation(s)
- Jill M Goldstein
- Connors Center for Women's Health and Gender Biology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Departments of Psychiatry and Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Laura Holsen
- Connors Center for Women's Health and Gender Biology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Departments of Psychiatry and Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Grace Huang
- Division of Endocrinology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Bradley D Hammond
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA; Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Tamarra James-Todd
- Connors Center for Women's Health and Gender Biology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Sara Cherkerzian
- Connors Center for Women's Health and Gender Biology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Departments of Psychiatry and Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Taben M Hale
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA
| | - Robert J Handa
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA; Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
148
|
Abstract
Prenatal insults, such as maternal stress, are associated with an increased neurodevelopmental disease risk and impact males significantly more than females, including increased rates of autism, mental retardation, stuttering, dyslexia, and attention deficit/hyperactivity disorder (ADHD). Sex differences in the placenta, which begin with sex chromosomes, are likely to produce sex-specific transplacental signals to the developing brain. Our studies and others have identified X-linked genes that are expressed at higher levels in the female placenta. Through a genome-wide screen after maternal stress in mice, we identified the X-linked gene O-linked N-acetylglucosamine transferase (OGT) and demonstrated its causality in neurodevelopmental programming producing a male-specific stress phenotype. Elucidating the sex-specific molecular mechanisms involved in transplacental signals that impact brain development is key to understanding the sex bias in neurodevelopmental disorders and is expected to yield novel insight into disease risk and resilience.
Collapse
Affiliation(s)
- Tracy L Bale
- Department of Biomedical Sciences, School of Veterinary Medicine and Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
149
|
Stroud LR, Papandonatos GD, Parade SH, Salisbury AL, Phipps MG, Lester B, Padbury JF, Marsit CJ. Prenatal Major Depressive Disorder, Placenta Glucocorticoid and Serotonergic Signaling, and Infant Cortisol Response. Psychosom Med 2017; 78:979-990. [PMID: 27763986 PMCID: PMC6541396 DOI: 10.1097/psy.0000000000000410] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Extending prior studies of prenatal adversity and depressive symptoms, we tested associations between maternal prenatal major depressive disorder (MDD) and infant cortisol regulation. Based on prior findings by our group, we also tested placenta glucocorticoid (HSD11B2 methylation) and serotonin (SLC6A4 gene expression) signaling as moderators of links between prenatal MDD and infant cortisol. METHODS Participants were 153 mother-infant pairs from a low-income, diverse sample (M [SD] age = 26 [6] years). Repeated structured diagnostic interviews were used to identify mothers with (a) prenatal MDD, (b) preconception-only MDD, and (c) controls. Placenta samples were assayed for HSD11B2 methylation and SLC6A4 gene expression. Infant salivary cortisol response to a neurobehavioral examination was assessed at 1 month. RESULTS Daughters of prenatal MDD mothers had 51% higher baseline (ratio = 1.51; 95% confidence interval [CI] = 1.01-2.27; p = .045) and 64% higher stress responsive cortisol (ratio = 1.64; 95% CI = 1.05-2.56; p = .03) than daughters of controls and 75% higher stress-responsive cortisol (ratio = 1.75; 95% CI = 1.04-2.94; p = .04) than daughters of preconception-only MDD mothers. HSD11B2 methylation moderated links between prenatal MDD and baseline cortisol (p = .02), with 1% methylation decreases associated with 9% increased baseline cortisol in infants of prenatal MDD mothers (ratio = 1.09; 95% CI = 1.01-1.16). SLC6A4 expression moderated links between prenatal MDD and cortisol response among boys alone (p = .007), with 10-fold increases in expression associated with threefold increases in stress-responsive cortisol (ratio = 2.87; 95% CI = 1.39-5.93) in sons of control mothers. CONCLUSIONS Results highlight specificity of associations between prenatal versus preconception MDD and cortisol regulation and the importance and complexity of placenta glucocorticoid and serotonergic pathways underlying the intergenerational transmission of risk from maternal adversity.
Collapse
Affiliation(s)
- Laura R. Stroud
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University
- Centers for Behavioral and Preventive Medicine, The Miriam Hospital
| | | | - Stephanie H. Parade
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University
- Bradley/Hasbro Children’s Research Center, Department of Psychology
| | - Amy L. Salisbury
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University
- Department of Pediatrics, Warren Alpert Medical School, Brown University
- Women & Infants’ Hospital of Rhode Island
| | - Maureen G. Phipps
- Women & Infants’ Hospital of Rhode Island
- Department of Obstetrics and Gynecology, Warren Alpert Medical School, Brown University
| | - Barry Lester
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University
- Department of Pediatrics, Warren Alpert Medical School, Brown University
- Women & Infants’ Hospital of Rhode Island
| | - James F. Padbury
- Department of Pediatrics, Warren Alpert Medical School, Brown University
- Women & Infants’ Hospital of Rhode Island
| | | |
Collapse
|
150
|
Association of prenatal and early childhood stress with reduced lung function in 7-year-olds. Ann Allergy Asthma Immunol 2017; 119:153-159. [PMID: 28668548 DOI: 10.1016/j.anai.2017.05.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/03/2017] [Accepted: 05/30/2017] [Indexed: 11/23/2022]
Abstract
BACKGROUND No prior study has examined associations between prenatal and early-life stress on childhood lung function or identified critical windows of exposure. OBJECTIVE To prospectively examine associations between prenatal and early-life stress and childhood lung function. METHODS Stress was indexed by a maternal negative life events (NLEs) score ascertained during pregnancy and between 1 and 2 years post partum. Spirometry was performed when children were a mean (SD) of 6.99 (0.89) years old. Associations of prenatal and early postnatal stress with spirometry z scores were examined in 199 children using linear regression. Effect modification by child sex was explored. RESULTS Most mothers were minorities (65% Hispanic, 21% African American), had 12 years or less of education (67%), and did not smoke prenatally (78%). The highest level of prenatal stress (≥5 NLEs) was associated with lower levels of forced expiratory volume in 1 second (FEV1) (z score = -0.53, P = .03), forced vital capacity (FVC) (z score = -0.49, P = .04), and forced expiratory flow between 25% and 75% (FEF25%-75%) (z score = -0.68, P = .01) after covariate adjustment; effects were similar for postnatal stress considered separately. In sex-stratified analyses, high postnatal stress (≥5 NLEs) was associated with lower FEV1 (z score = -0.76, P = .01), FVC (z score = -0.77, P = .01), and FEF25%-75% (z score = -0.67, P = .02) in boys but not girls, although the interaction term was not significant (P for interaction >.10). CONCLUSION These are the first prospective data that link perinatal stress with reduced child lung function. High levels of stress in the prenatal and postnatal periods were associated with symmetric reductions in FEV1 and FVC consistent with impaired lung growth. Given that lung function growth patterns are established by 7 years of age, these findings have lifelong implications.
Collapse
|