1
|
Torres DB, Lopes A, Rodrigues AJ, Ventura-Silva AP, Sousa N, Gontijo JAR, Boer PA, Lopes MG. Early morphological and neurochemical changes of the bed nucleus of stria terminalis (BNST) in gestational protein-restricted male offspring. Nutr Neurosci 2024; 27:1250-1268. [PMID: 38576309 DOI: 10.1080/1028415x.2024.2320498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
BACKGROUND The bed nucleus of the stria terminalis (BNST) is a structure with a peculiar neurochemical composition involved in modulating anxietylike behavior and fear. AIM The present study investigated the effects on the BNST neurochemical composition and neuronal structure in critical moments of the postnatal period in gestational protein-restricted male rats' offspring. METHODS Dams were maintained during the pregnancy on isocaloric rodent laboratory chow with standard protein content [NP, 17%] or low protein content [LP, 6%]. BNST from male NP and age-matched LP offspring was studied using the isotropic fractionator method, Neuronal 3D reconstruction, dendritic-tree analysis, blotting analysis, and high-performance liquid chromatography. RESULTS Serum corticosterone levels were higher in male LP offspring than NP rats in 14-day-old offspring, without any difference in 7-day-old progeny. The BNST total cell number and anterodorsal BNST division volume in LP progeny were significantly reduced on the 14th postnatal day compared with NP offspring. The BNST HPLC analysis from 7 days-old LP revealed increased norepinephrine levels compared to NP progeny. The BNST blot analysis from 7-day-old LP revealed reduced levels of GR and BDNF associated with enhanced CRF1 expression compared to NP offspring. 14-day-old LP offspring showed reduced expression of MR and 5HT1A associated with decreased DOPAC and DOPA turnover levels relative to NP rats. In Conclusion, the BNST cellular and neurochemical changes may represent adaptation during development in response to elevated fetal exposure to maternal corticosteroid levels. In this way, gestational malnutrition alters the BNST content and structure and contributes to already-known behavioral changes.
Collapse
Affiliation(s)
- D B Torres
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | - A Lopes
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | - A J Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - A P Ventura-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - N Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - J A R Gontijo
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | - P A Boer
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | | |
Collapse
|
2
|
Cheon BK, Bittner JMP, Pink AE. Contributions of subjective status to eating behaviors, obesity, and metabolic health across development. Appetite 2024:107735. [PMID: 39481682 DOI: 10.1016/j.appet.2024.107735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 09/20/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
Subjective status is the evaluation of one's social or socioeconomic status relative to others. Lower subjective status has been associated with risk of overweight/obesity, poorer metabolic health, and obesogenic food preferences and eating behaviors. However, these findings are predominantly based on studies of adolescents and young adults. This indicates major gaps in knowledge and application of this social determinant of obesity and metabolic health, given that perceived status develops throughout the life course along with food environments and eating habits. Here, we review the relationships that subjective status shares with the outcomes of eating behaviors, obesity, and metabolic health across milestones and periods of development: during the prenatal period, as caregivers who feed children, during childhood (prior to age 10) and from adolescence into emerging adulthood (until mid-20's). For each developmental period, we explore why the period critically contributes to these outcomes and how subjective status may affect eating behaviors and metabolic health. We propose that subjective status contributes to eating/feeding behaviors and metabolic health both within and across developmental periods, such that the effect of low subjective status at an earlier period may contribute to obesogenic eating behaviors and metabolic health in later developmental periods and intergenerationally. The influence of low subjective status on higher body weight may also threaten subjective status later in development through heightened vulnerability to social stressors, such as weight-based stigma. Overall, subjective status may be a broadly influential factor to consider when examining social determinants of obesity and metabolic health across development.
Collapse
Affiliation(s)
- Bobby K Cheon
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, 6710B Rockledge Drive, Bethesda, Maryland, USA.
| | - Julia M P Bittner
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, 6710B Rockledge Drive, Bethesda, Maryland, USA
| | - Aimee E Pink
- Institute of High Performance Computing (IHPC), Agency for Science, Technology and Research (A*STAR), 1 Fusionopolis Way, #16-16 Connexis, 138632, Republic of Singapore; Institute of Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Brenner Centre for Molecular Medicine, 117609, Republic of Singapore
| |
Collapse
|
3
|
Ivanova N, Atanasova M, Terzieva D, Georgieva K, Tchekalarova J. The Role of Piromelatine on Peripheral and Hippocampal Insulin Resistance in Rat Offspring Exposed to Chronic Maternal Stress. Int J Mol Sci 2024; 25:7022. [PMID: 39000130 PMCID: PMC11241293 DOI: 10.3390/ijms25137022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Prenatal stress (PNS), which alters the hypothalamic-pituitary-adrenal axis function in the offspring, predisposes to insulin resistance (IR) in later life and is associated with numerous disorders, including cognitive and memory impairments. At present, our main goal is to assess the effects of chronic piromelatine (Pir) administration, a melatonin analogue, on PNS-provoked IR in the periphery and the hippocampus in male and female offspring. Pregnant Sprague-Dawley rats were exposed to chronic stress (one short-term stressor on a daily basis and one long-term stressor on a nightly basis) from the first gestation week until birth. Vehicle or Pir 20 mg/kg were administered intraperitoneally for 21 days. Plasma glucose, serum insulin levels, and the homeostasis model assessment of insulin resistance (HOMA-IR) were determined as markers of peripheral IR. For the hippocampal IR assessment, insulin receptors (IRs) and glucose transporter 4 (GLUT4) were examined. Prenatally stressed offspring of both sexes indicated enhanced plasma glucose and serum insulin concentrations, increased HOMA-IR, and decreased hippocampal GLUT4 only in male rats. The PNS-induced changes were corrected by chronic treatment with Pir. The present results suggest that the melatoninergic compound Pir exerts beneficial effects on altered glucose/insulin homeostasis in PNS-exposed offspring.
Collapse
Affiliation(s)
- Natasha Ivanova
- Institute of Neurobiology, Bulgarian Academy of Sciences (BAS), 1113 Sofia, Bulgaria
| | - Milena Atanasova
- Department of Biology, Medical University of Pleven, 5800 Pleven, Bulgaria;
| | - Dora Terzieva
- Department of Clinical Laboratory, Medical Faculty, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Katerina Georgieva
- Department of Physiology, Medical University of Plovdiv, 5800 Pleven, Bulgaria;
| | - Jana Tchekalarova
- Institute of Neurobiology, Bulgarian Academy of Sciences (BAS), 1113 Sofia, Bulgaria
| |
Collapse
|
4
|
Ngema M, Xulu ND, Ngubane PS, Khathi A. A Review of Fetal Development in Pregnancies with Maternal Type 2 Diabetes Mellitus (T2DM)-Associated Hypothalamic-Pituitary-Adrenal (HPA) Axis Dysregulation: Possible Links to Pregestational Prediabetes. Biomedicines 2024; 12:1372. [PMID: 38927579 PMCID: PMC11201628 DOI: 10.3390/biomedicines12061372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Research has identified fetal risk factors for adult diseases, forming the basis for the Developmental Origins of Health and Disease (DOHaD) hypothesis. DOHaD suggests that maternal insults during pregnancy cause structural and functional changes in fetal organs, increasing the risk of chronic diseases like type 2 diabetes mellitus (T2DM) in adulthood. It is proposed that altered maternal physiology, such as increased glucocorticoid (GC) levels associated with a dysregulated hypothalamic-pituitary-adrenal (HPA) axis in maternal stress and T2DM during pregnancy, exposes the fetus to excess GC. Prenatal glucocorticoid exposure reduces fetal growth and programs the fetal HPA axis, permanently altering its activity into adulthood. This programmed HPA axis is linked to increased risks of hypertension, cardiovascular diseases, and mental disorders in adulthood. With the global rise in T2DM, particularly among young adults of reproductive age, it is crucial to prevent its onset. T2DM is often preceded by a prediabetic state, a condition that does not show any symptoms, causing many to unknowingly progress to T2DM. Studying prediabetes is essential, as it is a reversible stage that may help prevent T2DM-related pregnancy complications. The existing literature focuses on HPA axis dysregulation in T2DM pregnancies and its link to fetal programming. However, the effects of prediabetes on HPA axis function, specifically glucocorticoid in pregnancy and fetal outcomes, are not well understood. This review consolidates research on T2DM during pregnancy, its impact on fetal programming via the HPA axis, and possible links with pregestational prediabetes.
Collapse
Affiliation(s)
| | | | | | - Andile Khathi
- School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4001, South Africa; (M.N.); (N.D.X.); (P.S.N.)
| |
Collapse
|
5
|
Ngema M, Xulu ND, Ngubane PS, Khathi A. Pregestational Prediabetes Induces Maternal Hypothalamic-Pituitary-Adrenal (HPA) Axis Dysregulation and Results in Adverse Foetal Outcomes. Int J Mol Sci 2024; 25:5431. [PMID: 38791468 PMCID: PMC11122116 DOI: 10.3390/ijms25105431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Maternal type 2 diabetes mellitus (T2DM) has been shown to result in foetal programming of the hypothalamic-pituitary-adrenal (HPA) axis, leading to adverse foetal outcomes. T2DM is preceded by prediabetes and shares similar pathophysiological complications. However, no studies have investigated the effects of maternal prediabetes on foetal HPA axis function and postnatal offspring development. Hence, this study investigated the effects of pregestational prediabetes on maternal HPA axis function and postnatal offspring development. Pre-diabetic (PD) and non-pre-diabetic (NPD) female Sprague Dawley rats were mated with non-prediabetic males. After gestation, male pups born from the PD and NPD groups were collected. Markers of HPA axis function, adrenocorticotropin hormone (ACTH) and corticosterone, were measured in all dams and pups. Glucose tolerance, insulin and gene expressions of mineralocorticoid (MR) and glucocorticoid (GR) receptors were further measured in all pups at birth and their developmental milestones. The results demonstrated increased basal concentrations of ACTH and corticosterone in the dams from the PD group by comparison to NPD. Furthermore, the results show an increase basal ACTH and corticosterone concentrations, disturbed MR and GR gene expression, glucose intolerance and insulin resistance assessed via the Homeostasis Model Assessment (HOMA) indices in the pups born from the PD group compared to NPD group at all developmental milestones. These observations reveal that pregestational prediabetes is associated with maternal dysregulation of the HPA axis, impacting offspring HPA axis development along with impaired glucose handling.
Collapse
Affiliation(s)
| | | | | | - Andile Khathi
- School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Westville, Private Bag X54001, Durban 4041, KwaZulu Natal, South Africa; (M.N.); (N.D.X.); (P.S.N.)
| |
Collapse
|
6
|
Xu J, Zhang Q, Jiang T, Liu L, Gu H, Tan Y, Wang H. Dose- and stage-dependent toxic effects of prenatal prednisone exposure on fetal articular cartilage development. Toxicol Lett 2024; 393:14-23. [PMID: 38211732 DOI: 10.1016/j.toxlet.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/16/2023] [Accepted: 01/06/2024] [Indexed: 01/13/2024]
Abstract
Prednisone is frequently used to treat rheumatoid diseases in pregnant women because of its high degree of safety. Whether prenatal prednisone exposure (PPE) negatively impacts fetal articular cartilage development is unclear. In this study, we simulated a clinical prednisone treatment regimen to examine the effects of different timings and doses of PPE on cartilage development in female and male fetal mice. Prednisone doses (0.25, 0.5, and 1 mg/kg/d) was administered to Kunming mice at different gestational stages (0-9 gestational days, GD0-9), mid-late gestation (GD10-18), or during the entire gestation (GD0-18) by oral gavage. The amount of matrix aggrecan (ACAN) and collagen type II a1(COL2a1), and expression of transforming growth factor β1 (TGFβ1) signaling pathway also demonstrated that the chondrocyte count and ACAN and COL2a1 expression reduced in fetal mice with early and mid-late PPE, with the reduction being more significant in the mice with early PPE than that in those with PPE at other stages. Prenatal exposure to different prednisone doses prevented the reduction of TGFβ signaling pathway-related genes [TGFβR1, SMAD family member 3 (Smad3), SRY-box9 (SOX9)] as well as ACAN and COL2a1 mRNA expression levels in fetal mouse cartilage, with the most significant decrease after 1 mg/kg·d PPE. In conclusion, PPE can inhibit/restrain fetal cartilage development, with the greatest effect at higher clinical dose (1 mg/kg·d) and early stage of pregnancy (GD0-9), and the mechanism may be related to TGFβ signaling pathway inhibition. The result of this study provide a theoretical and experimental foundation for the rational clinical use of prednisone.
Collapse
Affiliation(s)
- Junmiao Xu
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qi Zhang
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Tao Jiang
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Liang Liu
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hanwen Gu
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yang Tan
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
7
|
Nomura Y, Newcorn JH, Ginalis C, Heitz C, Zaki J, Khan F, Nasrin M, Sie K, DeIngeniis D, Hurd YL. Prenatal exposure to a natural disaster and early development of psychiatric disorders during the preschool years: stress in pregnancy study. J Child Psychol Psychiatry 2023; 64:1080-1091. [PMID: 36129196 PMCID: PMC10027622 DOI: 10.1111/jcpp.13698] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/28/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Growing evidence shows an association between in utero exposure to natural disasters and child behavioral problems, but we still know little about the development of specific psychopathology in preschool-aged children. METHODS Preschool children (n = 163, mean age = 3.19, 85.5% racial and ethnic minorities) and their parents (n = 151) were evaluated annually at ages 2-5 to assess the emergence of psychopathology using the Preschool Age Psychopathological Assessment (PAPA), a parent-report structured diagnostic interview developed for preschool-age children. Sixty-six (40.5%) children were exposed to Sandy Storm (SS) in utero and 97 (59.5%) were not. Survival analysis evaluated patterns of onset and estimated cumulative risks of psychopathology among exposed and unexposed children, in total and by sex. Analyses were controlled for the severity of objective and subjective SS-related stress, concurrent family stress, and demographic and psychosocial confounders, such as maternal age, race, SES, maternal substance use, and normative prenatal stress. RESULTS Exposure to SS in utero was associated with a substantial increase in depressive disorders (Hazard Ratio (HR) = 16.9, p = .030), anxiety disorders (HR = 5.1, p < .0001), and attention-deficit/disruptive behavioral disorders (HR = 3.4, p = .02). Diagnostic rates were elevated for generalized anxiety disorder (GAD; HR = 8.5, p = .004), attention-deficit/hyperactivity disorder (ADHD; HR = 5.5, p = .01), oppositional-defiant disorder (ODD; HR = 3.8, p = .05), and separation-anxiety disorder (SAD; HR = 3.5, p = .001). Males had distinctively elevated risks for attention-deficit/disruptive behavioral disorders (HR = 7.8, p = .02), including ADHD, CD, and ODD, whereas females had elevated risks for anxiety disorders (HR = 10.0, p < .0001), phobia (HR = 2.8, p = .02) and depressive disorders (HR = 30.0, p = .03), including SAD, GAD, and dysthymia. CONCLUSIONS The findings demonstrate that in utero exposure to a major weather-related disaster (SS) was associated with increased risk for psychopathology in children and provided evidence of distinct psychopathological outcomes as a function of sex. More attention is needed to understand specific parent, child, and environmental factors which account for this increased risk, and to develop mitigation strategies.
Collapse
Affiliation(s)
- Yoko Nomura
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA
- CUNY Graduate Center, Department of Psychology, 365 5th Avenue, New York, NY 10016, USA
- Icahn School of Medicine at Mount Sinai, One Gustave Levy Pl, Box 1230, New York, NY 10029, USA
| | - Jeffrey H. Newcorn
- Icahn School of Medicine at Mount Sinai, One Gustave Levy Pl, Box 1230, New York, NY 10029, USA
| | - Christine Ginalis
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA
- CUNY Graduate Center, Department of Psychology, 365 5th Avenue, New York, NY 10016, USA
| | - Catherine Heitz
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA
| | - Jeenia Zaki
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA
| | - Farzana Khan
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA
- New York Medical College, School of Medicine, 40 Sunshine Cottage Rd, Valhalla, NY 10595, USA
| | - Mardia Nasrin
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA
- St. John’s University, Department of Clinical Health Professions, 8000 Utopia Pkwy, Queens, NY 11439, USA
| | - Kathryn Sie
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA
| | - Donato DeIngeniis
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA
| | - Yasmin L. Hurd
- Icahn School of Medicine at Mount Sinai, One Gustave Levy Pl, Box 1230, New York, NY 10029, USA
| |
Collapse
|
8
|
Mascioli I, Iapadre G, Ingrosso D, Donato GD, Giannini C, Salpietro V, Chiarelli F, Farello G. Brain and eye involvement in McCune-Albright Syndrome: clinical and translational insights. Front Endocrinol (Lausanne) 2023; 14:1092252. [PMID: 37274327 PMCID: PMC10235602 DOI: 10.3389/fendo.2023.1092252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/10/2023] [Indexed: 06/06/2023] Open
Abstract
McCune-Albright Syndrome (MAS) is a rare mosaic (post-zygotic) genetic disorder presenting with a broad continuum clinical spectrum. MAS arises from somatic, activating mutations in the GNAS gene, which induces a dysregulated Gsα-protein signaling in several tissues and an increased production of intracellular cyclic adenosine monophosphate (cAMP). Overall, MAS is a rare disorder affecting less than 1/100,000 children and, for this reason, data establishing genotype-phenotype correlations remain limited. Affected individuals clinically present with a variable combination of fibrous dysplasia of bone (FD), extra-skeletal manifestations (including cafeí-au-lait spots) and precocious puberty which might also be associated to broad hyperfunctioning endocrinopathies, and also gastrointestinal and cardiological involvement. Central nervous system (CNS) and eye involvement in MAS are among the less frequently described complications and remain largely uncharacterized. These rare complications mainly include neurodevelopmental abnormalities (e.g., delayed motor development, cognitive and language impairment), CNS anomalies (e.g., Chiari malformation type I) and a wide array of ophthalmological abnormalities often associated with vision loss. The pathophysiological mechanisms underlying abnormal neurological development have not been yet fully elucidated. The proposed mechanisms include a deleterious impact of chronically dysregulated Gsα-protein signaling on neurological function, or a secondary (damaging) effect of (antenatal and/or early postnatal) hypercortisolism on early pre- and post-natal CNS development. In this Review, we summarize the main neurological and ophthalmological features eventually associated with the MAS spectrum, also providing a detailed overview of the potential pathophysiological mechanisms underlying these clinical complications.
Collapse
Affiliation(s)
- Ilaria Mascioli
- Department of Pediatrics, University of Chieti, Chieti, Italy
| | - Giulia Iapadre
- Department of Pediatrics, University of L’Aquila, L’Aquila, Italy
| | | | - Giulio Di Donato
- Department of Pediatrics, University of L’Aquila, L’Aquila, Italy
| | - Cosimo Giannini
- Department of Pediatrics, University of Chieti, Chieti, Italy
| | | | | | - Giovanni Farello
- Department of Pediatrics, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
9
|
Gao LT, Yuan JQ, Zhang ZY, Zhao HM, Gao L. Hypermethylation of the Bmp4 promoter dampens binding of HIF-1α and impairs its cardiac protective effects from oxidative stress in prenatally GC-exposed offspring. Cell Mol Life Sci 2023; 80:58. [PMID: 36746787 PMCID: PMC9902418 DOI: 10.1007/s00018-023-04703-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/30/2022] [Accepted: 01/22/2023] [Indexed: 02/08/2023]
Abstract
The exposure to an unhealthy environment in utero can lead to the occurrence of cardiovascular diseases in the offspring. Glucocorticoids (GC) are essential for normal development and maturation of fetal organs and is a first-line treatment for pregnant women affected by autoimmune diseases. However, excess prenatal GC exposure might program the development of fetal organs and cause a number of chronic diseases in later life. Our previous studies indicated that cardiac functions were significantly compromised in rat offspring prenatally exposed to the synthetic glucocorticoid dexamethasone (DEX), only after ischemia-reperfusion. In the present study, we further observed that DNA hypermethylation of bone morphogenetic protein 4 (Bmp4) promoter in cardiomyocytes caused by prenatal DEX exposure substantially dampened the binding activity of transcription factor HIF-1α induced by cardiac ischemia. Therefore, prenatal DEX exposure inhibits the induction of BMP4 upon I/R and attenuates the protective effects of BMP4 in cardiomyocytes, which eventually manifests as malfunction of the adult heart. Moreover, we employed two cardiac-specific Bmp4 knock-in mouse models and found that in vivo BMP4 overexpression could rescue the cardiac dysfunction caused by prenatal GC exposure. In depth mechanistic research revealed that BMP4 protects the cardiomyocytes from mitophagy and apoptosis by attenuating mitochondrial PGC-1α expression in a p-Smad and Parkin-dependent manner. These findings suggest that prenatal GC exposure increases the susceptibility of the offspring's heart to a "second strike" after birth, due to the failure of hypoxia-induced HIF-1α transactivation of the hypermethylated Bmp4 promoter in cardiomyocytes. Pretreatment with the DNA methylation inhibitor, 5-Aza-2'-deoxycytidine, could be a potential therapeutic method for this programming effect of GC exposure during pregnancy on neonatal cardiac dysfunction.
Collapse
Affiliation(s)
- Ling-Tong Gao
- Department of Physiology, Naval Medical University, 800 Xiangyin Rd., Shanghai, 200433, People's Republic of China
| | - Jian-Qiang Yuan
- Department of Physiology, Naval Medical University, 800 Xiangyin Rd., Shanghai, 200433, People's Republic of China
| | - Zhi-Yu Zhang
- Department of Health Management, Changzheng Hospital, Naval Medical University, Shanghai, 200003, People's Republic of China
| | - Hou-Ming Zhao
- Department of Physiology, Naval Medical University, 800 Xiangyin Rd., Shanghai, 200433, People's Republic of China
| | - Lu Gao
- Department of Physiology, Naval Medical University, 800 Xiangyin Rd., Shanghai, 200433, People's Republic of China. .,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China. .,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, People's Republic of China.
| |
Collapse
|
10
|
Li X, Hu W, Li L, Chen Z, Jiang T, Zhang D, Liu K, Wang H. MiR-133a-3p/Sirt1 epigenetic programming mediates hypercholesterolemia susceptibility in female offspring induced by prenatal dexamethasone exposure. Biochem Pharmacol 2022; 206:115306. [PMID: 36326533 DOI: 10.1016/j.bcp.2022.115306] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 12/14/2022]
Abstract
Mounting evidence indicates that adverse intrauterine conditions increase offspring's hypercholesterolemia susceptibility in adulthood. This study aimed to confirm prenatal dexamethasone exposure (PDE)-induced hypercholesterolemia susceptibility in female adult offspring rats, and elucidate its intrauterine programming mechanism. Pregnant Wistar rats were injected with dexamethasone subcutaneously (0, 0.1 and 0.2 mg/kg·d) from gestational day (GD) 9 to 20. Serum and liver of the female offspring were collected at GD21 and postnatal week (PW) 12 and 28. PDE offspring showed elevated serum total cholesterol (TCH) levels and a cholesterol phenotype of high cardiovascular disease risk at PW12 and PW28. The histone acetylation levels of 3-hydroxy-3-methylglutaryl-coenzyme A reductase (Hmgcr) and its expression were consistently increased in the PDE offspring both in utero and after birth. Moreover, PDE promoted glucocorticoid receptor (GR) nuclear translocation and miR-133a-3p expression and inhibited sirtuin-1 (Sirt1) expression in the fetal liver. In vitro, dexamethasone increased intracellular and supernatant TCH levels and miR-133a-3p expression, decreased SIRT1 expression, and promoted HMGCR histone acetylation and expression in bone marrow mesenchymal stem cells (BMSCs) hepatoid differentiated cells and HepG2 cell line. GR siRNA, miR-133a-3p inhibitor or SIRT1 overexpression reversed dexamethasone-induced downstream molecular and phenotypic changes. Furthermore, elevated TCH levels in umbilical cord blood and increased HMGCR expression in peripheral blood mononuclear cells (PBMCs) were observed in human female neonates who had received dexamethasone treatment during pregnancy. In conclusion, PDE can cause persistent enhancement of hepatic cholesterol synthesis function before and after birth through GR/miR-133a-3p/Sirt1 pathway, eventually leading to increased hypercholesterolemia susceptibility in female offspring rats.
Collapse
Affiliation(s)
- Xufeng Li
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Wen Hu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Li Li
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Ze Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Tao Jiang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Dingmei Zhang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Kexin Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
11
|
Messina V, van’t Westeinde A, Padilla N, Lajic S. First Trimester Dexamethasone Treatment Is Not Associated With Alteration in Resting-state Connectivity at Adolescent or Adult Age. J Clin Endocrinol Metab 2022; 107:2769-2776. [PMID: 35882216 PMCID: PMC9516042 DOI: 10.1210/clinem/dgac426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Indexed: 11/28/2022]
Abstract
CONTEXT Prenatal treatment with dexamethasone (DEX) has been used to prevent virilization in females at risk of congenital adrenal hyperplasia (CAH). Both affected and unaffected girls, as well boys, are treated until the genotype and sex of the fetus is known (gestational weeks 10-12). After that, only affected girls are treated until term. Exposure to a high synthetic glucocorticoid dosage may alter the developmental trajectory of the brain, with alterations in resting-state functional connectivity of the brain at adult age. OBJECTIVE To investigate resting-state functional connectivity in subjects at risk of having CAH, exposed to DEX treatment during the first trimester of fetal life, both in the whole brain and in 3 regions of interest (amygdala, hippocampus, and superior frontal gyrus). DESIGN, SETTING, AND PARTICIPANTS Eighteen participants (8 females) at risk of having CAH, exposed to DEX treatment, and 38 controls (24 females), age range 16 to 26 years, from a single research institute, underwent functional magnetic resonance imaging of the brain during rest. We used 2 different approaches: an exploratory whole-brain analysis and seed-based analysis. For seed-based analysis, we chose 3 different brain regions (amygdala, hippocampus, and superior frontal gyrus) based on our previous findings and literature evidence. RESULTS We did not observe any differences in functional connectivity during rest, either in the whole brain nor in seed-based connectivity analyses at this adolescent and young adult age. CONCLUSIONS Our results are reassuring; however, future studies on larger samples and with more sensitive methodologies are needed to confirm these findings.
Collapse
Affiliation(s)
- Valeria Messina
- Department of Women’s and Children’s Health, Karolinska Institutet, Pediatric Endocrinology Unit (QB83), Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Annelies van’t Westeinde
- Department of Women’s and Children’s Health, Karolinska Institutet, Pediatric Endocrinology Unit (QB83), Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Nelly Padilla
- Department of Women’s and Children’s Health, Karolinska Institutet, Karolinska vägen 8 (S3:03), Karolinska University Hospital, SE- 171 76 Stockholm, Sweden
| | - Svetlana Lajic
- Correspondence: Svetlana Lajic, MD, Department of Women’s and Children’s Health, Pediatric Endocrinology Unit (QB83), Karolinska vägen 37A, Karolinska University Hospital, SE-171 76 Stockholm, Sweden.
| |
Collapse
|
12
|
Xu T, Ji B, Li L, Lei J, Zhao M, Sun M, Xu Z, Gao Q. Antenatal Dexamethasone Exposure Impairs Vascular Contractile Functions via Upregulating IP3 Receptor 1 and Cav1.2 in Adult Male Offspring. Hypertension 2022; 79:1997-2007. [PMID: 35762340 DOI: 10.1161/hypertensionaha.122.19040] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Administration of antenatal glucocorticoids remains common practice for treating preterm delivery. Antenatal glucocorticoid exposure increased the risk of developing vascular diseases in later life, but the precise mechanisms remain unclear. This study aimed to explore the effects and mechanisms of antenatal exposure to clinically relevant doses of dexamethasone (synthetic glucocorticoids) on vascular functions in adult male offspring. METHODS Pregnant Sprague-Dawley rats received dexamethasone or vehicle during the last week of pregnancy. Male offspring were killed at gestational day 21 (Fetus) or postnatal day 120 (adult offspring). Mesenteric arteries were collected for vascular function, electrophysiology, target gene expression, and promotor methylation studies. RESULTS Antenatal dexamethasone exposure increased phenylephrine-mediated vascular contractility in offspring, which was resulted by the activated inositol 1,4,5-trisphosphate (IP3) receptor and L-type Ca2+ channels. Specifically, increases of IP3R1 (IP3 receptor 1) and Cav1.2 (L-type Ca2+ channels subunit alpha1 C) were responsible for an activated IP3-Ca2+ pathway in the vasculature, and eventually predisposed the antenatal dexamethasone offspring to vascular hypercontractility. In addition, IP3R1 and Cav1.2 was upregulated through transcriptional mechanism; the overall changes in promotor histone modifications were consistent with the corresponding changes in transcriptional levels of the 2 genes, suggesting that antenatal dexamethasone exposure activated the transcription of IP3R1 and Cav1.2 via altering promotor histone modifications. CONCLUSIONS Taken together, this study demonstrated that antenatal dexamethasone exposure resulted in vascular adverse outcomes in male offspring that is linked to the increases of IP3R1 and Cav1.2 mediated by epigenetic modifications in the vasculature.
Collapse
Affiliation(s)
- Ting Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China (T.X., B.J., L.L., J.L., M.Z., M.S., Z.X., Q.G.)
| | - Bingyu Ji
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China (T.X., B.J., L.L., J.L., M.Z., M.S., Z.X., Q.G.)
| | - Lingjun Li
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China (T.X., B.J., L.L., J.L., M.Z., M.S., Z.X., Q.G.)
| | - Jiahui Lei
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China (T.X., B.J., L.L., J.L., M.Z., M.S., Z.X., Q.G.)
| | - Meng Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China (T.X., B.J., L.L., J.L., M.Z., M.S., Z.X., Q.G.)
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China (T.X., B.J., L.L., J.L., M.Z., M.S., Z.X., Q.G.)
| | - Zhice Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China (T.X., B.J., L.L., J.L., M.Z., M.S., Z.X., Q.G.)
| | - Qinqin Gao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China (T.X., B.J., L.L., J.L., M.Z., M.S., Z.X., Q.G.)
| |
Collapse
|
13
|
Murray A, Tharmalingam S, Khurana S, Lalonde C, Nguyen P, Tai TC. Effect of Prenatal Glucocorticoid Exposure on Circadian Rhythm Gene Expression in the Brains of Adult Rat Offspring. Cells 2022; 11:cells11101613. [PMID: 35626652 PMCID: PMC9139626 DOI: 10.3390/cells11101613] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/03/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022] Open
Abstract
Circadian clocks control many vital aspects of physiology from the sleep-wake cycle to metabolism. The circadian clock operates through transcriptional-translational feedback loops. The normal circadian signaling relies on a ‘master clock’, located in the suprachiasmatic nucleus (SCN), which synchronizes peripheral oscillators. Glucocorticoid receptor (GR) signaling has the ability to reset the phase of peripheral clocks. It has been shown that maternal exposure to glucocorticoids (GCs) can lead to modification of hypothalamic-pituitary-adrenal (HPA) function, impact stress-related behaviors, and result in a hypertensive state via GR activation. We previously demonstrated altered circadian rhythm signaling in the adrenal glands of offspring exposed to the synthetic GC, dexamethasone (Dex). Results from the current study show that prenatal exposure to Dex affects circadian rhythm gene expression in a brain region-specific and a sex-specific manner within molecular oscillators of the amygdala, hippocampus, paraventricular nucleus, and prefrontal cortex, as well as the main oscillator in the SCN. Results also show that spontaneously hypertensive rats (SHR) exhibited dysregulated circadian rhythm gene expression in these same brain regions compared with normotensive Wistar-Kyoto rats (WKY), although the pattern of dysregulation was markedly different from that seen in adult offspring prenatally exposed to GCs.
Collapse
Affiliation(s)
- Alyssa Murray
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (A.M.); (S.T.); (S.K.); (C.L.)
- Department of Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada;
| | - Sujeenthar Tharmalingam
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (A.M.); (S.T.); (S.K.); (C.L.)
- Department of Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada;
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Sandhya Khurana
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (A.M.); (S.T.); (S.K.); (C.L.)
| | - Christine Lalonde
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (A.M.); (S.T.); (S.K.); (C.L.)
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Phong Nguyen
- Department of Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada;
| | - T. C. Tai
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (A.M.); (S.T.); (S.K.); (C.L.)
- Department of Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada;
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Correspondence: ; Tel.: +1-705-662-7239
| |
Collapse
|
14
|
In Silico Predictions on the Productive Life Span and Theory of Its Developmental Origin in Dairy Cows. Animals (Basel) 2022; 12:ani12060684. [PMID: 35327081 PMCID: PMC8944687 DOI: 10.3390/ani12060684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Dairy cows are susceptible to a range of welfare factors, which lead to worsening health problems and shorten their productive life span. The health and welfare status of dairy cows could be improved if unwanted abnormalities and risk factors are detected in a timely manner, i.e., before diseases start to occur. Therefore, in addition to veterinary monitoring, quantitative parameters are necessary to predict the risks of early culling of cows. In the study of the age dynamics of culling rate in dairy cow populations, it was found that the average productive life span can be predicted by registration of the reciprocal relative disposal rate (culling for sum of reasons + death). This indicator represents the viability index, which has a maximal value at the first lactation and decreases in subsequent lactations with an inverse exponential trend. According to available scientific information, the structural prerequisites for this index are laid down during prenatal development and in the early periods of postnatal life; therefore, it is necessary to create a system of continuous monitoring of the physiological status of mothers and young animals. Abstract Animal welfare includes health but also concerns the need for natural factors that contribute to the increase in viability. Therefore, quantitative parameters are necessary to predict the risks of early culling of cows. In the study of the age dynamics of the disposal rate (culling for sum of reasons + death) in dairy cow populations, it was found that the average productive life span can be predicted by the value of the reciprocal culling/death rate (reciprocal value of Gompertz function) at the first lactation. This means that this potential of viability is formed during the developmental periods preceding the onset of lactation activity. Therefore, taking into account current data in the field of developmental biology, it can be assumed that the structural prerequisites for viability potential are laid down during prenatal development and in the early periods of postnatal life. To prevent unfavorable deviations in these processes due to negative welfare effects, it is advisable to monitor the physiological status of mothers and young animals using biosensors and Big Data systems.
Collapse
|
15
|
Babovic IR, Dotlic JR, Jovandaric MZ, Sparic RM, Bila JS, Nejkovic LV, Stulic JM, Tinelli A. Neurological outcomes of antenatal corticosteroid therapy. Int J Clin Pract 2021; 75:e14936. [PMID: 34605116 DOI: 10.1111/ijcp.14936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 10/01/2021] [Indexed: 11/27/2022] Open
Abstract
AIM OF THE STUDY The aim of the study was to investigate whether antenatal corticosteroid therapy (ACST) could impact neurological condition, as assessed through muscular tone, of prematurely born infants. METHODS All 82 patients at risk of preterm delivery treated and delivered over 12 months were divided into two equal groups regarding the use of ACST. The investigated parameters were pregnancy complications, biophysical profile, Apgar score, gestational age of delivery and all postpartum complications. Neurological development and muscular tone were evaluated at the 1st, 3rd, 6th and 12th months of life using Vojta's method, which classifies muscular tone as good, hypotonic or hypertonic. RESULTS After therapy, infants from the treated and control groups differed in biophysical profile, Apgar score, length of intensive care, occurrence of respiratory distress syndrome and intraventricular haemorrhage. During the follow-up, significantly more infants from the ACST group had good muscular tone when compared with those from the control group. Regression analysis showed that ASCT can significantly impact an infant's muscular tone. Still, the week of delivery and the complications such as diabetes mellitus, intrauterine growth restriction and respiratory distress syndrome, could change the association of ACST and infants' muscular tone. CONCLUSION ACST was associated with the positive neurological outcomes of prematurely born infants when assessed through their muscular tone.
Collapse
Affiliation(s)
- Ivana R Babovic
- Medical Faculty, University of Belgrade, Belgrade, Serbia
- Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, Belgrade, Serbia
| | - Jelena R Dotlic
- Medical Faculty, University of Belgrade, Belgrade, Serbia
- Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, Belgrade, Serbia
| | - Miljana Z Jovandaric
- Department of Neonatology, Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, Belgrade, Serbia
| | - Radmila M Sparic
- Medical Faculty, University of Belgrade, Belgrade, Serbia
- Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, Belgrade, Serbia
| | - Jovan S Bila
- Medical Faculty, University of Belgrade, Belgrade, Serbia
- Clinic for Gynecology and Obstetrics, University Clinical Center of Serbia, Belgrade, Serbia
| | - Lazar V Nejkovic
- Medical Faculty, University of Belgrade, Belgrade, Serbia
- Clinic of Obstetrics and Gynecology "Narodni Front", Belgrade, Serbia
| | - Jelena M Stulic
- Clinic of Obstetrics and Gynecology "Narodni Front", Belgrade, Serbia
| | - Andrea Tinelli
- Department of Obstetrics and Gynecology, "Veris delli Ponti Hospital", Scorrano, Italy
- Division of Experimental Endoscopic Surgery, Imaging, Technology and Minimally Invasive Therapy, Vito Fazzi Hospital, Lecce, Italy
- Laboratory of Human Physiology, Phystech BioMed School, Faculty of Biological & Medical Physics, Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia
| |
Collapse
|
16
|
Untargeted metabolomics reveals sex-specific differences in lipid metabolism of adult rats exposed to dexamethasone in utero. Sci Rep 2021; 11:20342. [PMID: 34645877 PMCID: PMC8514544 DOI: 10.1038/s41598-021-99598-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 09/22/2021] [Indexed: 12/13/2022] Open
Abstract
Prenatal stress through glucocorticoid (GC) exposure leads to an increased risk of developing diseases such as cardiovascular disease, metabolic syndrome and hypertension in adulthood. We have previously shown that administration of the synthetic glucocorticoid, dexamethasone (Dex), to pregnant Wistar-Kyoto dams produces offspring with elevated blood pressures and disrupted circadian rhythm signaling. Given the link between stress, circadian rhythms and metabolism, we performed an untargeted metabolomic screen on the livers of offspring to assess potential changes induced by prenatal Dex exposure. This metabolomic analysis highlighted 18 significantly dysregulated metabolites in females and 12 in males. Pathway analysis using MetaboAnalyst 4.0 highlighted key pathway-level metabolic differences: glycerophospholipid metabolism, purine metabolism and glutathione metabolism. Gene expression analysis revealed significant upregulation of several lipid metabolism genes in females while males showed no dysregulation. Triglyceride concentrations were also found to be significantly elevated in female offspring exposed to Dex in utero, which may contribute to lipid metabolism activation. This study is the first to conduct an untargeted metabolic profile of liver from GC exposed offspring. Corroborating metabolic, gene expression and lipid profiling results demonstrates significant sex-specific lipid metabolic differences underlying the programming of hepatic metabolism.
Collapse
|
17
|
Sex-dependent associations between maternal prenatal stressful life events, BMI trajectories and obesity risk in offspring: The Raine Study. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2021; 7:100066. [PMID: 35757055 PMCID: PMC9216251 DOI: 10.1016/j.cpnec.2021.100066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/25/2021] [Accepted: 06/03/2021] [Indexed: 11/21/2022] Open
|
18
|
Kieffer TE, Chin PY, Green ES, Moldenhauer LM, Prins JR, Robertson SA. Prednisolone in early pregnancy inhibits regulatory T cell generation and alters fetal and placental development in mice. Mol Hum Reprod 2021; 26:340-352. [PMID: 32159777 DOI: 10.1093/molehr/gaaa019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/16/2020] [Indexed: 01/01/2023] Open
Abstract
Corticosteroids have been utilised in the assisted reproduction setting with the expectation of suppressing aberrant immune activation and improving fertility in women. However, the effects of corticosteroids on fertility, and on pregnancy and offspring outcomes, are unclear. In this study, mice were administered prednisolone (1 mg/kg) or PBS daily in the pre-implantation phase, and effects on the adaptive immune response, the implantation rate, fetal development and postnatal outcomes were investigated. Prednisolone disrupted the expected expansion of CD4+ T cells in early pregnancy, inhibiting generation of both regulatory T cells (Treg cells) and effector T cells and suppressing IFNG required for T cell functional competence. Prednisolone caused an 8-20% increase in the embryo implantation rate and increased the number of viable pups per litter. In late gestation, fetal and placental weights were reduced in a litter size-dependent manner, and the canonical inverse relationship between litter size and fetal weight was lost. The duration of pregnancy was extended by ~ 0.5 day and birth weight was reduced by ~ 5% after prednisolone treatment. Viability of prednisolone-exposed offspring was comparable to controls, but body weight was altered in adulthood, particularly in male offspring. Thus, while prednisolone given in the pre-implantation phase in mice increases maternal receptivity to implantation and resource investment in fetal growth, there is a trade-off in long-term consequences for fetal development, birth weight and offspring health. These effects are associated with, and likely caused by, prednisolone suppression of the adaptive immune response at the outset of pregnancy.
Collapse
Affiliation(s)
- Tom Ec Kieffer
- Robinson Research Institute & Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia.,Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Peck Y Chin
- Robinson Research Institute & Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Ella S Green
- Robinson Research Institute & Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Lachlan M Moldenhauer
- Robinson Research Institute & Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Jelmer R Prins
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sarah A Robertson
- Robinson Research Institute & Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
19
|
Jeje S, Akpan E, Kunle-Alabi O, Akindele O, Raji Y. Protective role of Allium cepa Linn (onion) juice on maternal dexamethasone induced alterations in reproductive functions of female offspring of Wistar rats. Curr Res Physiol 2021; 4:145-154. [PMID: 34746834 PMCID: PMC8562199 DOI: 10.1016/j.crphys.2021.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/21/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023] Open
Abstract
Maternal treatment with dexamethasone induces oxidative stress in the reproductive structures of offspring. Consumption of Allium cepa Linn improves antioxidant status. This study was designed to evaluate the protective role of Allium cepa Linn juice on maternal dexamethasone induced alterations in reproductive functions of the female offspring of Wistar rats. Twenty lactating dams (180-200 g) were randomly assigned into four groups (n = 5) on the day of parturition and treated as follows during lactation for 21 days: Control (5 ml/kg BW distilled water); Dexamethasone (60 μg/kg BW); Allium cepa (5 ml/kg BW); Dexamethasone + Allium cepa (60 μg/kg BW + 5 ml/kg BW). The female offspring were separated at birth. Days of vaginal opening and first oestrus cycle, length and frequency of estrous cycle as well as serum hormonal profiles were assessed as measure of reproductive functions. Ovarian superoxide dismutase (SOD) activity, catalase activity and malondialdehyde (MDA) level were measured as indices of oxidative stress. Oestrous cycle length, frequencies of diestrus as well as the Ovarian MDA were significantly increased (p < 0.05) in dexamethasone (DEX) group relative to control group. Serum 17β-oestradiol and corticosterone level in addition to SOD and catalase activities were significantly reduced (p < 0.05) in DEX group relative to control. Co-administration of Dex with Allium cepa Linn juice reduced the oestrous length, frequency of diestrous as well as ovarian MDA. There was also a significant increase in serum 17β-oestradiol, ovarian SOD and catalase activity. The results suggest that Allium cepa could protect against alterations in reproductive functions of offspring induced by maternal treatment with dexamethasone during lactation in Wistar rats. The flavonoid constituent of onion may also help in reducing oxidative stress in the offspring.
Collapse
Affiliation(s)
- S.O. Jeje
- Department of Physiology, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - E.E. Akpan
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| | - O.T. Kunle-Alabi
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| | - O.O. Akindele
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| | - Y. Raji
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
20
|
Tampakakis E, Mahmoud AI. The role of hormones and neurons in cardiomyocyte maturation. Semin Cell Dev Biol 2021; 118:136-143. [PMID: 33931308 DOI: 10.1016/j.semcdb.2021.03.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/19/2021] [Accepted: 03/29/2021] [Indexed: 12/28/2022]
Abstract
The heart undergoes profound morphological and functional changes as it continues to mature postnatally. However, this phase of cardiac development remains understudied. More recently, cardiac maturation research has attracted a lot of interest due to the need for more mature stem cell-derived cardiomyocytes for disease modeling, drug screening and heart regeneration. Additionally, neonatal heart injury models have been utilized to study heart regeneration, and factors regulating postnatal heart development have been associated with adult cardiac disease. Critical components of cardiac maturation are systemic and local biochemical cues. Specifically, cardiac innervation and the concentration of various metabolic hormones appear to increase perinatally and they have striking effects on cardiomyocytes. Here, we first report some of the key parameters of mature cardiomyocytes and then discuss the specific effects of neurons and hormonal cues on cardiomyocyte maturation. We focus primarily on the structural, electrophysiologic, metabolic, hypertrophic and hyperplastic effects of each factor. This review highlights the significance of underappreciated regulators of cardiac maturation and underscores the need for further research in this exciting field.
Collapse
Affiliation(s)
- Emmanouil Tampakakis
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Ahmed I Mahmoud
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA.
| |
Collapse
|
21
|
Kozieł S, Chakraborty R, Bose K, Ignasiak Z, Gomula A, Nowak-Szczepanska N. The effect of a natural disaster on handgrip strength in prepubertal Indian children exposed to a severe cyclone during the prenatal and early postnatal growth. Sci Rep 2021; 11:7473. [PMID: 33811238 PMCID: PMC8018953 DOI: 10.1038/s41598-021-86845-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 03/16/2021] [Indexed: 11/09/2022] Open
Abstract
Natural disasters (NDs) experienced by women and their children during prenatal and infant growth may have long-lasting effects on offspring’s development. Handgrip strength (HGS) is one of the measures of muscular strength and an indicator of health status. This study compared HGS in children exposed to cyclone Aila in India during their prenatal and infant growth compared to a control group from a non-affected, adjacent area. The total sample involved 444 boys and 423 girls aged 7–9 years, categorised into 3 groups: prenatally exposed to Aila, exposed to Aila in infancy, and the control group, non-exposed to Aila. Results revealed that prenatally exposed children of both sexes had significantly lower HGS than the controls (at least, p < 0.001 in boys; p < 0.05 in girls). On the other hand, the postnatally exposed boys, but not the girls, showed lower HGS than the controls. A significant effect of a group factor (ND exposure) on HGS was observed even after controlling for confounding variables (age, height, BMI, birth weight, gestational age; at least, p < 0.05). Our findings indicate that prenatal or early postnatal experience of a ND may have association with impaired HGS in prepubertal children.
Collapse
Affiliation(s)
- Sławomir Kozieł
- Department of Anthropology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Raja Chakraborty
- Department of Anthropology, Dinabandhu Mahavidyalaya, Bongaon, West Bengal, India.
| | - Kaushik Bose
- Department of Anthropology, Vidyasagar University, Midnapore, West Bengal, India
| | - Zofia Ignasiak
- Department of Biostructure, University School of Physical Education in Wroclaw, Wroclaw, Poland
| | - Aleksandra Gomula
- Department of Anthropology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Natalia Nowak-Szczepanska
- Department of Anthropology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
22
|
Abstract
In recent decades low- and middle-income countries (LMICs) have been witnessing a significant shift toward raised blood pressure; yet in LMICs, only 1 in 3 are aware of their hypertension status, and ≈8% have their blood pressure controlled. This rising burden widens the inequality gap, contributes to massive economic hardships of patients and carers, and increases costs to the health system, facing challenges such as low physician-to-patient ratios and lack of access to medicines. Established risk factors include unhealthy diet (high salt and low fruit and vegetable intake), physical inactivity, tobacco and alcohol use, and obesity. Emerging risk factors include pollution (air, water, noise, and light), urbanization, and a loss of green space. Risk factors that require further in-depth research are low birth weight and social and commercial determinants of health. Global actions include the HEARTS technical package and the push for universal health care. Promising research efforts highlight that successful interventions are feasible in LMICs. These include creation of health-promoting environments by introducing salt-reduction policies and sugar and alcohol tax; implementing cost-effective screening and simplified treatment protocols to mitigate treatment inertia; pooled procurement of low-cost single-pill combination therapy to improve adherence; increasing access to telehealth and mHealth (mobile health); and training health care staff, including community health workers, to strengthen team-based care. As the blood pressure trajectory continues creeping upward in LMICs, contextual research on effective, safe, and cost-effective interventions is urgent. New emergent risk factors require novel solutions. Lowering blood pressure in LMICs requires urgent global political and scientific priority and action.
Collapse
Affiliation(s)
- Aletta E Schutte
- School of Population Health, University of New South Wales, Sydney, Australia (A.E.S.)
- George Institute for Global Health, Sydney, NSW, Australia (A.E.S.)
- Hypertension in Africa Research Team, MRC Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa (A.E.S.)
| | - Nikhil Srinivasapura Venkateshmurthy
- Centre for Chronic Conditions and Injuries, Public Health Foundation of India, Gurgaon (N.S.V., S.M., D.P.)
- Centre for Chronic Disease Control, New Delhi, India (N.S.V., S.M., D.P.)
- School of Exercise and Nutrition Sciences (N.S.V.), Deakin University, Burwood, VIC, Australia
| | - Sailesh Mohan
- Centre for Chronic Conditions and Injuries, Public Health Foundation of India, Gurgaon (N.S.V., S.M., D.P.)
- Centre for Chronic Disease Control, New Delhi, India (N.S.V., S.M., D.P.)
- Faculty of Health (S.M.), Deakin University, Burwood, VIC, Australia
| | - Dorairaj Prabhakaran
- Centre for Chronic Conditions and Injuries, Public Health Foundation of India, Gurgaon (N.S.V., S.M., D.P.)
- Centre for Chronic Disease Control, New Delhi, India (N.S.V., S.M., D.P.)
- Department of Epidemiology, London School of Hygiene and Tropical Medicine, United Kingdom (D.P.)
| |
Collapse
|
23
|
Eberle C, Fasig T, Brüseke F, Stichling S. Impact of maternal prenatal stress by glucocorticoids on metabolic and cardiovascular outcomes in their offspring: A systematic scoping review. PLoS One 2021; 16:e0245386. [PMID: 33481865 PMCID: PMC7822275 DOI: 10.1371/journal.pone.0245386] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022] Open
Abstract
Background “Stress” is an emerging problem in our society, health care system as well as patient care, worldwide. Especially by focusing on pre-gestational, gestational but also lactation phases “stress” is to be considered as an own trans-generational risk factor which is associated with adverse metabolic as well cardiovascular outcomes in mothers and their children. Hence, the maternal hypothalamic-pituitary-adrenotrophic (HPA) axis may be stimulated by various “stress” mechanisms as well as risk factors leading to an adverse in utero environment, e.g. by excess exposure of glucocorticoids, contributing to cardio-metabolic disorders in mothers and their offspring. Objective To review the evidence of in utero programming by focusing on the impact of maternal “stress”, on adverse cardio-metabolic outcomes on their offspring later in life, by identifying underlying (patho-) physiological mechanisms (1) as well as adverse short and long-term cardio-metabolic outcomes (2). Methods We conducted a systematic scoping review to identify publications systematically including reviews, interventional, observational, experimental studies as well as human and animal model studies. MEDLINE (PubMed) and EMBASE databases and reference lists were searched. Peer-reviewed articles from January 2000 until August 2020 were included. Results Overall, n = 2.634 citations were identified, n = 45 eligible studies were included and synthesized according to their key findings. In brief, maternal hypothalamic-pituitary-adrenotrophic (HPA) axis might play a key role modifying in utero milieu leading to cardio-metabolic diseases in the offspring later in life. However, maternal risk factor “stress”, is clearly linked to adverse cardio-metabolic offspring outcomes, postnatally, such as obesity, hyperglycemia, insulin resistance, diabetes mellitus (DM), Metabolic Syndrome (MetS), cardiovascular disease (CD), hypertension, restricted fetal growth as well as reduced birth, adrenal, and pancreas weights. Conclusions Women who experienced “stress” as risk factor, as well as their offspring, clearly have a higher risk of adverse short- as well as long-term cardio-metabolic outcomes. Future research work is needed to understand complex transgenerational mechanisms.
Collapse
Affiliation(s)
- Claudia Eberle
- Medicine with Specialization in Internal Medicine and General Medicine, Hochschule Fulda-University of Applied Sciences, Fulda, Germany
| | - Teresa Fasig
- Medicine with Specialization in Internal Medicine and General Medicine, Hochschule Fulda-University of Applied Sciences, Fulda, Germany
| | - Franziska Brüseke
- Medicine with Specialization in Internal Medicine and General Medicine, Hochschule Fulda-University of Applied Sciences, Fulda, Germany
| | - Stefanie Stichling
- Medicine with Specialization in Internal Medicine and General Medicine, Hochschule Fulda-University of Applied Sciences, Fulda, Germany
| |
Collapse
|
24
|
Hlisníková H, Petrovičová I, Kolena B, Šidlovská M, Sirotkin A. Effects and mechanisms of phthalates’ action on neurological processes and neural health: a literature review. Pharmacol Rep 2021; 73:386-404. [DOI: 10.1007/s43440-021-00215-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/15/2020] [Accepted: 01/05/2021] [Indexed: 12/17/2022]
|
25
|
Sivasangari K, Rajan KE. Standardized Bacopa monnieri Extract Ameliorates Learning and Memory Impairments through Synaptic Protein, Neurogranin, Pro-and Mature BDNF Signaling, and HPA Axis in Prenatally Stressed Rat Offspring. Antioxidants (Basel) 2020; 9:antiox9121229. [PMID: 33291595 PMCID: PMC7761874 DOI: 10.3390/antiox9121229] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/19/2020] [Accepted: 11/24/2020] [Indexed: 01/10/2023] Open
Abstract
Prenatal stress (PNS) influences offspring neurodevelopment, inducing anxiety-like behavior and memory deficits. We investigated whether pretreatment of Bacopa monnieri extract (CDRI-08/BME) ameliorates PNS-induced changes in signaling molecules, and changes in the behavior of Wistar rat offspring. Pregnant rats were randomly assigned into control (CON)/prenatal stress (PNS)/PNS and exposed to BME treatment (PNS + BME). Dams were exposed to stress by placing them in a social defeat cage, where they observed social defeat from gestational day (GD)-16–18. Pregnant rats in the PNS + BME group were given BME treatment from GD-10 to their offspring’s postnatal day (PND)-23, and to their offspring from PND-15 to -30. PNS led to anxiety-like behavior; impaired memory; increased the level of corticosterone (CORT), adrenocorticotropic hormone, glucocorticoid receptor, pro-apoptotic Casepase-3, and 5-HT2C receptor; decreased anti-apoptotic Bcl-2, synaptic proteins (synaptophysin, synaptotagmin-1), 5-HT1A, receptor, phosphorylation of calmodulin-dependent protein kinase II/neurogranin, N-methyl-D-aspartate receptors (2A,2B), postsynaptic density protein 95; and conversion of pro and mature brain derived neurotropic factor in their offspring. The antioxidant property of BME possibly inhibiting the PNS-induced changes in observed molecules, anxiety-like behavior, and memory deficits. The observed results suggest that pretreatment of BME could be an effective coping strategy to prevent PNS-induced behavioral impairments in their offspring.
Collapse
|
26
|
Pham A, Zhang J, Feng L. Exposure to perfluorobutane sulfonate and perfluorooctanesulfonic acid disrupts the production of angiogenesis factors and stress responses in human placental syncytiotrophoblast. Reprod Toxicol 2020; 98:269-277. [PMID: 33144174 DOI: 10.1016/j.reprotox.2020.10.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 10/14/2020] [Accepted: 10/25/2020] [Indexed: 12/18/2022]
Abstract
Poly- and per-fluoroalkyl substances (PFAS) have attracted widespread attention in recent years due to their bioaccumulation, toxicity, and ubiquitous nature. We and others have reported that maternal exposure to PFAS is associated with adverse birth outcomes due to altered placental functions. In this study, we investigated the effects of two major PFAS compounds, perfluorobutane sulfonate (PFBS) and perfluorooctanesulfonic acid (PFOS), on the regulation of the production of angiogenic factors and stress response in placental multinucleated syncytial BeWo cells using qRT-PCR and ELISA. Using this in vitro model, we showed that 1) PFOS or PFBS treatment did not seem to interrupt BeWo cell fusion through syncytins; 2) Exposure to PFOS at 10 μM decreased a potent angiogenic factor PlGF gene expression, which is implicated in preeclampsia; 3) Exposure to either PFOS or PFBS significantly decreased the production of CGB7 and hCG except hCG secretion in PFOS (10 nM) and PFBS (100 nM) treatment groups; 4) Exposure to PFOS (10 μM) increased the gene expression of the stress response molecules CRH while neither PFOS nor PFBS treatment affected a stress mitigation factor 11β-HSD2 expression. Our results demonstrate that exposure to PFOS or PFBS impacts several key pathways involved in placental cell functions. PFOS seems more potent than PFBS. These novel findings provide a potential explanation for the adverse reproductive complications associated with prenatal exposure to PFOS or PFBS, including preeclampsia and contribute to our knowledge of the reproductive toxicity of PFAS, specifically PFOS and PFBS.
Collapse
Affiliation(s)
- Angela Pham
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, United States
| | - Jun Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Liping Feng
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, United States; MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
27
|
Crisafulli A, Bassareo PP, Kelleher S, Calcaterra G, Mercuro G. Factors Predisposing to Hypertension in Subjects Formerly Born Preterm: Renal Impairment, Arterial Stiffness, Endothelial Dysfunction or Something Else? Curr Hypertens Rev 2020; 16:82-90. [PMID: 31244440 PMCID: PMC7499358 DOI: 10.2174/1573402115666190627140523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/21/2019] [Accepted: 05/26/2019] [Indexed: 12/23/2022]
Abstract
Subjects formerly born preterm subsequently develop arterial - particularly isolated systolic- hypertension more frequently than their peers born at term. Numerous factors may influence this predisposition, including an incomplete nephrogenesis, implying the presence of kidneys with a reduced number of nephrons and consequent reduction in haematic filtration, increased sodium absorption and activation of renin-angiotensin-aldosterone system, increased arterial rigidity produced by an elastin deficiency previously observed in anatomic specimens of human immature aorta, and reduced endothelial nitric oxide excretion, due to high blood levels of ADMA, a strong direct inhibitor of nitric oxide that exerts a vasoconstrictor effect. Other possible factors (i.e. excretion of neuroendocrine compounds) may also be implicated. The aim of this paper was to review all possible mechanisms involved in the observed increase in blood pressure in individuals who had been born preterm and/or with intrauterine growth restriction. The outlook for new and promising laboratory techniques capable of identifying alterations in the metabolic pathways regulating blood pressure levels, such as metabolomics, is also provided.
Collapse
Affiliation(s)
- Antonio Crisafulli
- Department of Medical Sciences and Public Health, Sports Physiology Laboratory, University of Cagliari, Cagliari, Italy
| | | | - Sean Kelleher
- Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | | | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
28
|
Ward SA, Kirkwood RN, Plush KJ. Administering dexamethasone to prepartum sows: Effects on sow and piglet performance. Livest Sci 2020. [DOI: 10.1016/j.livsci.2020.104171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
29
|
Xiao H, Wu Z, Li B, Shangguan Y, Stoltz JF, Magdalou J, Chen L, Wang H. The low-expression programming of 11β-HSD2 mediates osteoporosis susceptibility induced by prenatal caffeine exposure in male offspring rats. Br J Pharmacol 2020; 177:4683-4700. [PMID: 32757222 DOI: 10.1111/bph.15225] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 02/12/2020] [Accepted: 07/23/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Prenatal caffeine exposure (PCE) can cause developmental toxicity of long bones in offspring, but the long-term effects and the underlying mechanism have not been fully clarified. Here, we investigated the effects of PCE peak bone mass accumulation and osteoporosis susceptibility in offspring and its intrauterine programming mechanism. EXPERIMENTAL APPROACH Pregnant Wistar rats were administrated intragastrically with saline or caffeine (120 mg·kg-1 ·day-1 ) on gestational days 9-20. The serum and bone samples were collected from the fetal and postnatal offspring for bone mass, genes expression and corticosterone analysis. Then, rat bone marrow mesenchymal stem cells (BMSCs) were treated with corticosterone in vitro to confirm the molecular mechanism. KEY RESULTS PCE caused fetal bone dysplasia in male and female offspring. In adulthood, PCE reduced peak bone mass and increased osteoporosis susceptibility in male offspring but not in females. Meanwhile, PCE only decreased the H3K9ac and expression levels of 11β-hydroxysteroid dehydrogenase 2 (11β-HSD2) before and after birth in the male offspring but not in the females. Moreover, the high level of corticosterone induced by PCE down-regulated the H3K9ac and expression levels of 11β-HSD2 through promoting glucocorticoid receptor (GR; NR3C1) into the nucleus of bone marrow mesenchymal stem cells (BMSCs) and recruiting histone deacetylase 11 (HDAC11) binding to 11β-HSD2 promoter region, which further enhanced the effect of corticosterone on suppressing osteogenic function of BMSCs. CONCLUSION AND IMPLICATIONS PCE caused osteoporosis susceptibility in male adult offspring, which attributed to the low-functional programming of 11β-HSD2 induced by corticosterone via GR/HDAC11 signalling.
Collapse
Affiliation(s)
- Hao Xiao
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhixin Wu
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bin Li
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yangfan Shangguan
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | | | | | - Liaobin Chen
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| |
Collapse
|
30
|
Skeffington KL, Beck C, Itani N, Niu Y, Shaw CJ, Giussani DA. Hypertension Programmed in Adult Hens by Isolated Effects of Developmental Hypoxia In Ovo. Hypertension 2020; 76:533-544. [PMID: 32536277 PMCID: PMC7340221 DOI: 10.1161/hypertensionaha.120.15045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In mammals, pregnancy complicated by chronic hypoxia can program hypertension in the adult offspring. However, mechanisms remain uncertain because the partial contributions of the challenge on the placenta, mother, and fetus are difficult to disentangle. Here, we used chronic hypoxia in the chicken embryo-an established model system that permits isolation of the direct effects of developmental hypoxia on the cardiovascular system of the offspring, independent of additional effects on the mother or the placenta. Fertilized chicken eggs were exposed to normoxia (N; 21% O2) or hypoxia (H; 13.5%-14% O2) from the start of incubation (day 0) until day 19 (hatching, ≈day 21). Following hatching, all birds were maintained under normoxic conditions until ≈6 months of adulthood. Hypoxic incubation increased hematocrit (+27%) in the chicken embryo and induced asymmetrical growth restriction (body weight, -8.6%; biparietal diameter/body weight ratio, +7.5%) in the hatchlings (all P<0.05). At adulthood (181±4 days), chickens from hypoxic incubations remained smaller (body weight, -7.5%) and showed reduced basal and stimulated in vivo NO bioavailability (pressor response to NG-nitro-L-arginine methyl ester, -43%; phenylephrine pressor response during NO blockade, -61%) with significant hypertension (mean arterial blood pressure, +18%), increased cardiac work (ejection fraction, +12%; fractional shortening, +25%; enhanced baroreflex gain, +456%), and left ventricular wall thickening (left ventricular wall volume, +36%; all P<0.05). Therefore, we show that chronic hypoxia can act directly on a developing embryo to program hypertension, cardiovascular dysfunction, and cardiac wall remodeling in adulthood in the absence of any maternal or placental effects.
Collapse
Affiliation(s)
- Katie L. Skeffington
- From the Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (K.L.S., C.B., N.I., Y.N., C.J.S., D.A.G.)
| | - Christian Beck
- From the Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (K.L.S., C.B., N.I., Y.N., C.J.S., D.A.G.)
| | - Nozomi Itani
- From the Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (K.L.S., C.B., N.I., Y.N., C.J.S., D.A.G.)
| | - Youguo Niu
- From the Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (K.L.S., C.B., N.I., Y.N., C.J.S., D.A.G.)
| | - Caroline J. Shaw
- From the Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (K.L.S., C.B., N.I., Y.N., C.J.S., D.A.G.),Department of Metabolism, Digestion and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, United Kingdom (C.J.S.)
| | - Dino A. Giussani
- From the Department of Physiology, Development and Neuroscience, University of Cambridge, United Kingdom (K.L.S., C.B., N.I., Y.N., C.J.S., D.A.G.)
| |
Collapse
|
31
|
Wang WS, Guo CM, Sun K. Cortisol Regeneration in the Fetal Membranes, A Coincidental or Requisite Event in Human Parturition? Front Physiol 2020; 11:462. [PMID: 32523541 PMCID: PMC7261858 DOI: 10.3389/fphys.2020.00462] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/16/2020] [Indexed: 12/20/2022] Open
Abstract
The fetal membranes are equipped with high capacity of cortisol regeneration through the reductase activity of 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1). The expression of 11β-HSD1 in the fetal membranes is under the feedforward induction by cortisol, which is potentiated by proinflammatory cytokines. As a result, the abundance of 11β-HSD1 increases with gestational age and furthermore at parturition with an escalation of cortisol concentration in the fetal membranes. Accumulated cortisol takes parts in a number of crucial events pertinent to the onset of labor in the fetal membranes, including extracellular matrix (ECM) remodeling and stimulation of prostaglandin output. Cortisol remodels the ECM through multiple approaches including induction of collagen I, III, and IV degradation, as well as inhibition of their cross-linking. These effects of cortisol are executed through activation of the autophagy, proteasome, and matrix metalloprotease 7 pathways, as well as inhibition of the expression of cross-linking enzyme lysyl oxidase in mesenchymal cells of the membranes. With regard to prostaglandin output, cortisol not only increases prostaglandin E2 and F2α syntheses through induction of their synthesizing enzymes such as cytosolic phospholipase A2, cyclooxygenase 2, and carbonyl reductase 1 in the amnion, but also decreases their degradation through inhibition of their metabolizing enzyme 15-hydroxyprostaglandin dehydrogenase in the chorion. Taking all together, data accumulated so far denote that the feedforward cortisol regeneration by 11β-HSD1 in the fetal membranes is a requisite event in the onset of parturition, and the effects of cortisol on prostaglandin synthesis and ECM remodeling may be enhanced by proinflammatory cytokines in chorioamnionitis.
Collapse
Affiliation(s)
- Wang-Sheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Chun-Ming Guo
- School of Life Sciences, Yunnan University, Kunming, China
| | - Kang Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
32
|
Jeje SO, Adegbite LO, Akindele OO, Kunle-Alabi OT, Raji Y. Allium cepa Linn juice protect against alterations in reproductive functions induced by maternal dexamethsone treatment during lactation in male offspring of Wistar rats. Heliyon 2020; 6:e03872. [PMID: 32395653 PMCID: PMC7205748 DOI: 10.1016/j.heliyon.2020.e03872] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/02/2019] [Accepted: 04/23/2020] [Indexed: 11/13/2022] Open
Abstract
Treatment with dams with dexamethasone during lactation has been reported to induce oxidative stress in the testis of the offspring. Allium cepa L (Red Onion) is known to be a potent free radical scavenger. The protective role of Allium cepa against oxidative stress induced in testis following treatment with dexamehasone during lactation in Wistar rats was assessed. Twenty female rats were assigned into four groups (n = 5) during lactation and they were treated as follows: Group 1 serve as Control (distilled water), Group 2, 3, and four were admistered dexamethasone (60 μg/kg), Allium cepa (5 ml/kg) and dexamethasone + Allium cepa respectively. Testicular descent, pubertal age, sperm quality indices, and serum hormonal profile were assessed as indices of reproductive function. Testicular malondialdehyde (MDA) reduced glutathione (GSH) as well as superoxide dismutase (SOD) and catalase activities were assessed as measures of oxidative stress. Results obtained showed that dexamethasone caused significant (P < 0.05) reduction in testes weights, indices of sperm quality, serum testosterone, FSH, LH levels and testicular antioxidant enzyme activities. There was significant delay (P < 0.05) in days of testes descent, preputial separation and increase in testicular MDA. However, maternal treatment with Allium cepa Linn juice significantly (P < 0.05) improved both indices of reproductive function and testicular antioxidant enzymes. These findings suggest that Allium cepa Linn has a protective effect against testicular oxidative stress and reproductive dysfunction following treatment of dams with dexamethasone during lactation.
Collapse
Affiliation(s)
- S O Jeje
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria.,Department of Physiology, School of Health and Health Technology, Federal University of Technology, AKure, Nigeria
| | - L O Adegbite
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| | - O O Akindele
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| | - O T Kunle-Alabi
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| | - Y Raji
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
33
|
Li L, Hu W, Liu K, Zhang D, Liu M, Li X, Wang H. miR-148a/LDLR mediates hypercholesterolemia induced by prenatal dexamethasone exposure in male offspring rats. Toxicol Appl Pharmacol 2020; 395:114979. [PMID: 32234517 DOI: 10.1016/j.taap.2020.114979] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/11/2022]
Abstract
Epidemiology suggests that adverse environmental exposure during pregnancy may predispose children to hypercholesterolemia in adulthood. This study aimed to demonstrate hypercholesterolemia induced by prenatal dexamethasone exposure (PDE) in adult male offspring rats and explore the intrauterine programming mechanisms. Pregnant Wistar rats were injected subcutaneously with dexamethasone (0, 0.1, 0.2, and 0.4 mg/kg∙d) from gestational days (GD) 9 to 21, and the serum and liver of the male offsprings were collected at GD21, postnatal week (PW) 12 and 28. Furthermore, the effects of dexamethasone on the expression of low-density lipoprotein receptor (LDLR) and its epigenetic mechanism was confirmed in the bone marrow mesenchymal stem cells (BMSCs) hepatoid differentiated cells and continuous hepatocyte line. PDE could reduce the birth weight of male offsprings, increase the serum total cholesterol (TCH) level in adult rats, and decrease the liver low-density lipoprotein receptor (LDLR) expression. Serum TCH level and liver LDLR expression were decreased in PDE male fetuses in utero (GD21). Moreover, PDE increased the translocation of the glucocorticoid receptor (GR) in the fetal liver, the expression of DiGeorge syndrome critical region 8 gene (DGCR8), the pre- and post-natal expression of miR-148a. The results of PDE offspring in vivo and in vitro exhibited similar changes. These changes could be reversed by overexpressing LDLR, inhibiting miR-148a or GR. PDE caused hypercholesterolemia in male adult offspring rats, which was mediated via dexamethasone activated intrauterine hepatic GR, enhanced the expression of DGCR8 and miR-148a, thereby reducing the expression of LDLR, leading to impaired liver cholesterol reverse transport function, and finally causing hypercholesterolemia in adult rats.
Collapse
Affiliation(s)
- Li Li
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Wen Hu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Kexin Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Dingmei Zhang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Min Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Xufeng Li
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China.
| |
Collapse
|
34
|
Li J, Xiao H, Luo H, Tan Y, Ni Q, He C, Magdalou J, Chen L, Wang H. GR/HDAC2/TGFβR1 pathway contributes to prenatal caffeine induced-osteoarthritis susceptibility in male adult offspring rats. Food Chem Toxicol 2020; 140:111279. [PMID: 32199975 DOI: 10.1016/j.fct.2020.111279] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 03/05/2020] [Accepted: 03/15/2020] [Indexed: 12/25/2022]
Abstract
Prenatal caffeine exposure (PCE) induces developmental toxicity of multi-organ and susceptibility to multi-disease in offspring. However, the effects of PCE on osteoarthritis susceptibility in adult offspring and its intrauterine programming mechanism remain to be further investigated. Here, we found that PCE induced susceptibility to osteoarthritis in male adult offspring rats, which was related to the inhibited function of cartilage matrix synthesis from fetuses to adults. Meanwhile, PCE consistently downregulated the H3K9ac and expression levels of transforming growth factor β receptor 1 (TGFβR1), and then blocked TGFβ signaling pathway, which contributed to the suppressed cartilage matrix synthesis. Moreover, the high level of corticosterone caused by PCE reduced the H3K9ac level on TGFβR1 promoter region through acting on glucocorticoids receptor (GR) and recruiting histone deacetylase 2 (HDAC2) into the nucleus of fetal chondrocytes. Taken together, PCE induced osteoarthritis susceptibility in male adult offspring rats, which was attributed to the low-functional programming of TGFβR1 induced by corticosterone via GR/HDAC2 signaling.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Hao Xiao
- Department of Orthopedic Surgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China
| | - Hanwen Luo
- Department of Orthopedic Surgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China
| | - Yang Tan
- Department of Orthopedic Surgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Qubo Ni
- Department of Orthopedic Surgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Chunjiang He
- Department of Medical Genetics, Basic Medical School of Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | | | - Liaobin Chen
- Department of Orthopedic Surgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
35
|
Hollanders J, Dijkstra LR, van der Voorn B, Kouwenhoven SM, Toorop AA, van Goudoever JB, Rotteveel J, Finken MJ. No Association between Glucocorticoid Diurnal Rhythm in Breastmilk and Infant Body Composition at 3 Months. Nutrients 2019; 11:nu11102351. [PMID: 31581748 PMCID: PMC6835896 DOI: 10.3390/nu11102351] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/08/2023] Open
Abstract
Objective: Glucocorticoids (GCs) in breastmilk have previously been associated with infant body growth and body composition. However, the diurnal rhythm of breastmilk GCs was not taken into account, and we therefore aimed to assess the associations between breastmilk GC rhythmicity at 1 month and growth and body composition at 3 months in infants. Methods: At 1 month postpartum, breastmilk GCs were collected over a 24-h period and analyzed by LC-MS/MS. Body composition was measured using air-displacement plethysmography at 3 months. Length and weight were collected at 1, 2, and 3 months. Results: In total, 42 healthy mother–infant pairs were included. No associations were found between breastmilk GC rhythmicity (area-under-the-curve increase and ground, maximum, and delta) and infant growth trajectories or body composition (fat and fat free mass index, fat%) at 3 months. Conclusions: This study did not find an association between breastmilk GC rhythmicity at 1 month and infant’s growth or body composition at 3 months. Therefore, this study suggests that previous observations linking breastmilk cortisol to changes in infant weight might be flawed by the lack of serial cortisol measurements and detailed information on body composition.
Collapse
Affiliation(s)
- Jonneke Hollanders
- Emma Children’s Hospital, Amsterdam UMC, Pediatric Endocrinology, Vrije Universiteit Amsterdam, 1000-1183 Amsterdam, The Netherlands; (L.R.D.); (A.A.T.); (J.R.)
- Correspondence: ; Tel.: +31-(0)20-4443137
| | - Lisette R. Dijkstra
- Emma Children’s Hospital, Amsterdam UMC, Pediatric Endocrinology, Vrije Universiteit Amsterdam, 1000-1183 Amsterdam, The Netherlands; (L.R.D.); (A.A.T.); (J.R.)
| | - Bibian van der Voorn
- Department of Paediatric Endocrinology, Obesity Center CGG, Sophia Children’s Hospital, 3000-3099 Rotterdam, The Netherlands;
| | - Stefanie M.P. Kouwenhoven
- Emma Children’s Hospital, Amsterdam UMC, Department of Pediatrics, Vrije Universiteit Amsterdam, 1000-1183 Amsterdam, The Netherlands (J.B.v.G.)
| | - Alyssa A. Toorop
- Emma Children’s Hospital, Amsterdam UMC, Pediatric Endocrinology, Vrije Universiteit Amsterdam, 1000-1183 Amsterdam, The Netherlands; (L.R.D.); (A.A.T.); (J.R.)
| | - Johannes B. van Goudoever
- Emma Children’s Hospital, Amsterdam UMC, Department of Pediatrics, Vrije Universiteit Amsterdam, 1000-1183 Amsterdam, The Netherlands (J.B.v.G.)
| | - Joost Rotteveel
- Emma Children’s Hospital, Amsterdam UMC, Pediatric Endocrinology, Vrije Universiteit Amsterdam, 1000-1183 Amsterdam, The Netherlands; (L.R.D.); (A.A.T.); (J.R.)
| | - Martijn J.J. Finken
- Emma Children’s Hospital, Amsterdam UMC, Pediatric Endocrinology, Vrije Universiteit Amsterdam, 1000-1183 Amsterdam, The Netherlands; (L.R.D.); (A.A.T.); (J.R.)
| |
Collapse
|
36
|
Early postnatal hypothalamic-pituitary-adrenal axis activity and reduced insulin sensitivity in adult rats. Endocr Regul 2019; 53:213-220. [PMID: 31734655 DOI: 10.2478/enr-2019-0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE Early life stress influences the development of metabolic disorders, including functional changes in the developing of pancreas mediated hypothalamic-pituitary-adrenal (HPA) axis. In the present study, the role of an early postnatal stress on corticosterone, glucose, and insulin levels was investigated during young adulthood. METHODS Two groups of pups were studied, including control group (pups not receiving foot shock by communication box), and early stress group (pups receiving foot shock by communication box 2 times/day for 5 consecutive days). In rats, concentration of plasma corticosterone, glucose, and insulin was detected before and after placing them into the communication box at 2 weeks of age. At 8-10 weeks of age, concentrations of plasma corticosterone, glucose, and insulin and glucose tolerance were measured in young adult rats. RESULTS Our results showed that early postnatal foot shock stress increased the corticosterone, insulin, and glucose levels in the postnatal age (p<0.01) that did not last until young adult age, but it caused a significant increase in plasma glucose and insulin levels (p<0.05) following the intraperitoneal glucose tolerance test (IPGTT) in young adult rats. CONCLUSIONS These results suggest that impaired IPGTT in young adult rats who experienced early postnatal stress can indicate insulin resistance or reduced insulin sensitivity that make it at risk of the type 2 diabetes later in life.
Collapse
|
37
|
D’Agostino S, Testa M, Aliperti V, Venditti M, Minucci S, Aniello F, Donizetti A. Expression pattern dysregulation of stress- and neuronal activity-related genes in response to prenatal stress paradigm in zebrafish larvae. Cell Stress Chaperones 2019; 24:1005-1012. [PMID: 31209726 PMCID: PMC6717227 DOI: 10.1007/s12192-019-01017-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/06/2019] [Accepted: 06/10/2019] [Indexed: 12/19/2022] Open
Abstract
Maternal stress during pregnancy adversely affects developmental fetal programming. Glucocorticoid excess is one of those conditions that underlie the prenatal stress and can lead to many pathological disorders later in life. Beyond the obvious use of mammalian model organisms to uncover the different mechanisms at the basis of prenatal stress effects, zebrafish represents a complementary fruitful model for this research field. Here we demonstrated that the application of an experimental paradigm, which simulates prenatal stress by exposing embryos to cortisol excess, produced an alteration of gene expression pattern. In particular, the transcript level of hsd11b2, a gene involved in the cortisol catabolism, was affected in prenatally stressed larvae, even after many hours from the removal of cortisol excess. Interestingly, the expression pattern of c-fos, a marker gene of neural activity, was affected in prenatally stressed larvae even in response to a swirling and osmotic stress challenge. Our data corroborate the idea of zebrafish as a useful model organism to study prenatal stress effects on vertebrate development.
Collapse
Affiliation(s)
- Serena D’Agostino
- Department of Biology, University of Naples Federico II, via Cinthia, 80126 Naples, Italy
| | - Martino Testa
- Department of Biology, University of Naples Federico II, via Cinthia, 80126 Naples, Italy
| | - Vincenza Aliperti
- Department of Biology, University of Naples Federico II, via Cinthia, 80126 Naples, Italy
| | - Massimo Venditti
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, via Costantinopoli 16, 80138 Naples, Italy
| | - Sergio Minucci
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, via Costantinopoli 16, 80138 Naples, Italy
| | - Francesco Aniello
- Department of Biology, University of Naples Federico II, via Cinthia, 80126 Naples, Italy
| | - Aldo Donizetti
- Department of Biology, University of Naples Federico II, via Cinthia, 80126 Naples, Italy
| |
Collapse
|
38
|
Song R, Hu XQ, Zhang L. Glucocorticoids and programming of the microenvironment in heart. J Endocrinol 2019; 242:T121-T133. [PMID: 31018174 PMCID: PMC6602534 DOI: 10.1530/joe-18-0672] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 04/24/2019] [Indexed: 12/11/2022]
Abstract
Glucocorticoids are primary stress hormones and can improve neonatal survival when given to pregnant women threatened by preterm birth or to preterm infants. It has become increasingly apparent that glucocorticoids, primarily by interacting with glucocorticoid receptors, play a critical role in late gestational cardiac maturation. Altered glucocorticoid actions contribute to the development and progression of heart disease. The knowledge gained from studies in the mature heart or cardiac damage is insufficient but a necessary starting point for understanding cardiac programming including programming of the cardiac microenvironment by glucocorticoids in the fetal heart. This review aims to highlight the potential roles of glucocorticoids in programming of the cardiac microenvironment, especially the supporting cells including endothelial cells, immune cells and fibroblasts. The molecular mechanisms by which glucocorticoids regulate the various cellular and extracellular components and the clinical relevance of glucocorticoid functions in the heart are also discussed.
Collapse
Affiliation(s)
- Rui Song
- Correspondence to: Rui Song, PhD, , Lubo Zhang, PhD,
| | | | - Lubo Zhang
- Correspondence to: Rui Song, PhD, , Lubo Zhang, PhD,
| |
Collapse
|
39
|
Buthmann J, Finik J, Ventura G, Zhang W, Shereen AD, Nomura Y. The children of Superstorm Sandy: Maternal prenatal depression blunts offspring electrodermal activity. Biol Psychol 2019; 146:107716. [PMID: 31176750 DOI: 10.1016/j.biopsycho.2019.107716] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022]
Abstract
We set out to examine the relations between prenatal exposure to the natural disaster Superstorm Sandy, maternal depression, and offspring electrodermal activity (EDA). EDA was measured via skin conductance response (SCR) magnitude in 198 children (M = 42.54 months, SD = 12.76) during a startle paradigm. In keeping with prior research, we expected prenatal depression to be associated with hyporeactive EDA and prenatal stress to be associated with hyperreactive EDA. SCR magnitude was lower in children prenatally exposed to depression alone, when compared to Superstorm Sandy, and controls. SCR magnitude of children prenatally exposed to both maternal depression and the storm was lower than that of all other groups. Our results emphasize the influence of maternal prenatal mental health, support targeted risk assessment for children who experienced an adverse prenatal environment, and highlight the need for a deeper understanding of the interactions between maternal mood and stress on the developing child.
Collapse
Affiliation(s)
- J Buthmann
- CUNY Graduate Center, Department of Psychology, 365 5th Avenue, New York, NY 10016, USA; CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA.
| | - J Finik
- CUNY Graduate School of Public Health and Health Policy, Department of Epidemiology and Biostatistics, 55 W 125th St., New York, NY 10027, USA
| | - G Ventura
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA
| | - W Zhang
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA; New Jersey City University, Department of Psychology, 2039 John Fitzgerald Kennedy Blvd, Jersey City, NJ 07305, USA
| | - A D Shereen
- CUNY Advanced Science Research Center, Graduate Center, 85 St Nicholas Terrace, New York, NY 10031, USA; University of California, Department of Neurology, 680 California Ave, Irvine, CA 92697, USA
| | - Y Nomura
- CUNY Graduate Center, Department of Psychology, 365 5th Avenue, New York, NY 10016, USA; CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY 11367, USA; CUNY Graduate School of Public Health and Health Policy, Department of Epidemiology and Biostatistics, 55 W 125th St., New York, NY 10027, USA; CUNY Advanced Science Research Center, Graduate Center, 85 St Nicholas Terrace, New York, NY 10031, USA; Mount Sinai School of Medicine, Department of Psychiatry, 1 Gustave Levy Pl, New York, NY 10029, USA
| |
Collapse
|
40
|
Byrne L, Drake AJ. Paediatrician's guide to epigenetics. Arch Dis Child 2019; 104:297-301. [PMID: 30655261 DOI: 10.1136/archdischild-2018-316054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 11/03/2022]
Abstract
Epigenetic regulation of gene expression is critical for normal development. Dysregulation of the epigenome can lead to the development and progression of a number of diseases relevant to paediatricians, including disorders of genomic imprinting and malignancies. It has long been recognised that early life events have implications for future disease risk, and epigenetic modifications may play a role in this, although further high-quality research is needed to better understand the underlying mechanisms. Research in the field of epigenetics will contribute to a greater understanding of growth, development and disease; however, paediatricians need to be able to interpret such research critically, in order to use the potential advances brought about through epigenetic studies while appreciating their limitations.
Collapse
Affiliation(s)
- Lauren Byrne
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Amanda Jane Drake
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK.,Department of Paediatric Endocrinology, Royal Hospital for Sick Children, Edinburgh, UK
| |
Collapse
|
41
|
Buthmann J, Ham J, Davey K, Finik J, Dana K, Pehme P, Zhang W, Glover V, Nomura Y. Infant Temperament: Repercussions of Superstorm Sandy-Related Maternal Stress. Child Psychiatry Hum Dev 2019; 50:150-162. [PMID: 30030653 PMCID: PMC6339835 DOI: 10.1007/s10578-018-0828-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This study recruited a prospective cohort of 380 pregnant women before, during, or after Superstorm Sandy in 2012 to examine the association between disaster-related pre- and post-natal maternal stress and offspring temperament at 6 months-old. Mothers prospectively reported stressful experiences during the storm and rated their child's temperament 6 months postpartum. Results indicated that length of time without phone or electricity and financial loss was associated with offspring negative affect, whereas financial loss and threat of death or injury was associated with emotion dysregulation. Furthermore, offspring born before the storm had greater negative affect and lower emotion regulation than those born after the storm. Given the probable increase in the occurrence of natural disasters due to climate change in recent years (McCarthy, Intergovernmental Panel on Climate Change, Climate change 2001: impacts, adaptation, and vulnerability: contribution of Working Group II to the third assessment report of the Intergovernmental Panel on Climate Change, Cambridge University Press, Cambridge, 2001), our results highlight the necessity of education and planning to help ameliorate any potential consequences on the developing infant.
Collapse
Affiliation(s)
- Jessica Buthmann
- Psychology, Queens College, CUNY, 65-30 Kissena Blvd, Flushing, 11367, NY, USA.
- Psychology, The Graduate Center, CUNY, New York, NY, USA.
| | - Jacob Ham
- Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Child Trauma and Resilience, Mount Sinai Beth Israel, New York, NY, USA
| | - Katherine Davey
- Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Classics, Bryn Mawr College, Bryn Mawr, PA, USA
| | - Jackie Finik
- Psychology, Queens College, CUNY, 65-30 Kissena Blvd, Flushing, 11367, NY, USA
- Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Hunter College, CUNY, School of Public Health, New York, NY, USA
| | - Kathryn Dana
- Psychology, Queens College, CUNY, 65-30 Kissena Blvd, Flushing, 11367, NY, USA
- Psychology, The Graduate Center, CUNY, New York, NY, USA
- Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Patricia Pehme
- Psychology, Queens College, CUNY, 65-30 Kissena Blvd, Flushing, 11367, NY, USA
- Psychology, The Graduate Center, CUNY, New York, NY, USA
| | - Wei Zhang
- Psychology, Queens College, CUNY, 65-30 Kissena Blvd, Flushing, 11367, NY, USA
- Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Yoko Nomura
- Psychology, Queens College, CUNY, 65-30 Kissena Blvd, Flushing, 11367, NY, USA.
- Psychology, The Graduate Center, CUNY, New York, NY, USA.
- Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Hunter College, CUNY, School of Public Health, New York, NY, USA.
| |
Collapse
|
42
|
Zhu P, Wang W, Zuo R, Sun K. Mechanisms for establishment of the placental glucocorticoid barrier, a guard for life. Cell Mol Life Sci 2019; 76:13-26. [PMID: 30225585 PMCID: PMC11105584 DOI: 10.1007/s00018-018-2918-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/16/2018] [Accepted: 09/06/2018] [Indexed: 01/07/2023]
Abstract
The fetus is shielded from the adverse effects of excessive maternal glucocorticoids by 11β-HSD2, an enzyme which is expressed in the syncytial layer of the placental villi and is capable of converting biologically active cortisol into inactive cortisone. Impairment of this placental glucocorticoid barrier is associated with fetal intrauterine growth restriction (IUGR) and development of chronic diseases in later life. Ontogeny studies show that the expression of 11β-HSD2 is initiated at a very early stage after conception and increases with gestational age but declines around term. The promoter for HSD11B2, the gene encoding 11β-HSD2, has a highly GC-rich core. However, the pattern of methylation on HSD11B2 may have already been set up in the blastocyst when the trophoblast identity is committed. Instead, hCG-initiated signals appear to be responsible for the upsurge of 11β-HSD2 expression during trophoblast syncytialization. By activating the cAMP/PKA pathway, hCG not only alters the modification of histones but also increases the expression of Sp1 which activates the transcription of HSD11B2. Adverse conditions such as stress, hypoxia and nutritional restriction can cause IUGR of the fetus. It appears that different causes of IUGR may attenuate HSD11B2 expression differentially in the placenta. While stress and nutritional restriction may reduce HSD11B2 expression by increasing its methylation, hypoxia may decrease HSD11B2 expression via alternative mechanisms rather than by methylation. Herein, we summarize the advances in the study of mechanisms underlying the establishment of the placental glucocorticoid barrier and the attenuation of this barrier by adverse conditions during pregnancy.
Collapse
Affiliation(s)
- Ping Zhu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Department of Obstetrics and Gynecology, No. 401 Hospital, Qingdao, People's Republic of China
| | - Wangsheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Rujuan Zuo
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Kang Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China.
| |
Collapse
|
43
|
Slotkin TA, Ko A, Seidler FJ. Does growth impairment underlie the adverse effects of dexamethasone on development of noradrenergic systems? Toxicology 2018; 408:11-21. [PMID: 29935188 DOI: 10.1016/j.tox.2018.06.008] [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] [Received: 04/13/2018] [Revised: 05/15/2018] [Accepted: 06/19/2018] [Indexed: 12/24/2022]
Abstract
Glucocorticoids are given in preterm labor to prevent respiratory distress but these agents evoke neurobehavioral deficits in association with reduced brain region volumes. To determine whether the neurodevelopmental effects are distinct from growth impairment, we gave developing rats dexamethasone at doses below or within the therapeutic range (0.05, 0.2 or 0.8 mg/kg) at different stages: gestational days (GD) 17-19, postnatal days (PN) 1-3 or PN7-9. In adolescence and adulthood, we assessed the impact on noradrenergic systems in multiple brain regions, comparing the effects to those on somatic growth or on brain region growth. Somatic growth was reduced with exposure in all three stages, with greater sensitivity for the postnatal regimens; brain region growth was impaired to a lesser extent. Norepinephrine content and concentration were reduced depending on the treatment regimen, with a rank order of deficits of PN7-9 > PN1-3 > GD17-19. However, brain growth impairment did not parallel reduced norepinephrine content in magnitude, dose threshold, sex or regional selectivity, or temporal pattern, and even when corrected for reduced brain region weights (norepinephrine per g tissue), the dexamethasone-exposed animals showed subnormal values. Regression analysis showed that somatic growth impairment accounted for an insubstantial amount of the reduction in norepinephrine content, and brain growth impairment accounted for only 12%, whereas specific effects on norepinephrine accounted for most of the effect. The adverse effects of dexamethasone on noradrenergic system development are not simply related to impaired somatic or brain region growth, but rather include specific targeting of neurodifferentiation.
Collapse
Affiliation(s)
- Theodore A Slotkin
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina, 27710, USA.
| | - Ashley Ko
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Frederic J Seidler
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| |
Collapse
|
44
|
Brachetta V, Schleich CE, Cutrera AP, Merlo JL, Kittlein MJ, Zenuto RR. Prenatal predatory stress in a wild species of subterranean rodent: Do ecological stressors always have a negative effect on the offspring? Dev Psychobiol 2018; 60:567-581. [DOI: 10.1002/dev.21635] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 03/01/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Valentina Brachetta
- Laboratorio de Ecología Fisiológica y del Comportamiento, Instituto de Investigaciones Marinas y Costeras (IIMyC); Universidad Nacional de Mar del Plata - CONICET; de Mar del Plata Argentina
| | - Cristian E. Schleich
- Laboratorio de Ecología Fisiológica y del Comportamiento, Instituto de Investigaciones Marinas y Costeras (IIMyC); Universidad Nacional de Mar del Plata - CONICET; de Mar del Plata Argentina
| | - Ana P. Cutrera
- Laboratorio de Ecología Fisiológica y del Comportamiento, Instituto de Investigaciones Marinas y Costeras (IIMyC); Universidad Nacional de Mar del Plata - CONICET; de Mar del Plata Argentina
| | - Julieta L. Merlo
- Laboratorio de Ecología Fisiológica y del Comportamiento, Instituto de Investigaciones Marinas y Costeras (IIMyC); Universidad Nacional de Mar del Plata - CONICET; de Mar del Plata Argentina
| | - Marcelo J. Kittlein
- Laboratorio de Ecología Fisiológica y del Comportamiento, Instituto de Investigaciones Marinas y Costeras (IIMyC); Universidad Nacional de Mar del Plata - CONICET; de Mar del Plata Argentina
| | - Roxana R. Zenuto
- Laboratorio de Ecología Fisiológica y del Comportamiento, Instituto de Investigaciones Marinas y Costeras (IIMyC); Universidad Nacional de Mar del Plata - CONICET; de Mar del Plata Argentina
| |
Collapse
|
45
|
Kroska EB, O'Hara MW, Elgbeili G, Hart KJ, Laplante DP, Dancause KN, King S. The impact of maternal flood-related stress and social support on offspring weight in early childhood. Arch Womens Ment Health 2018; 21:225-233. [PMID: 29080991 PMCID: PMC7318548 DOI: 10.1007/s00737-017-0786-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 10/04/2017] [Indexed: 11/24/2022]
Abstract
The current study examined the moderating role of social support in the association between prenatal maternal stress (PNMS) and childhood body mass index (BMI) in the context of the Iowa floods of 2008. In addition, the mediating role of offspring birthweight was examined in the association between PNMS and childhood BMI. We recruited women from eastern Iowa who were pregnant in 2008 when disastrous floods occurred. Self-report measures of PNMS and cognitive appraisal of the flood's consequences were obtained shortly after the disaster. Social support was assessed during pregnancy. Offspring anthropometric measures were collected at birth and 30 months. Moderated mediation results indicated that greater PNMS predicted greater BMI at age 30 months through effects on higher birthweight as a mediator, but only for participants with low social support. High social support (satisfaction or number) buffered the effect of PNMS or a negative appraisal of the flood on birthweight. The combination of high PNMS or a negative appraisal of the flood's consequences and low social support resulted in higher offspring birthweight, which predicted greater BMI at 30 months. Providing strong social support to pregnant women following a stressor might buffer the effects of PNMS on offspring birthweight and later obesity.
Collapse
Affiliation(s)
- Emily B Kroska
- Department of Psychological and Brain Sciences, University of Iowa, 311 Seashore Hall W., Iowa City, IA, 52242, USA.
| | - Michael W O'Hara
- Department of Psychological and Brain Sciences, University of Iowa, 311 Seashore Hall W., Iowa City, IA, 52242, USA
| | - Guillaume Elgbeili
- Douglas Institute Research Centre, McGill University, Montreal, QC, Canada
| | - Kimberly J Hart
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - David P Laplante
- Douglas Institute Research Centre, McGill University, Montreal, QC, Canada
| | - Kelsey N Dancause
- Department of Physical Activity Sciences, University of Quebec at Montreal, Montreal, QC, Canada
| | - Suzanne King
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
46
|
Slotkin TA, Skavicus S, Seidler FJ. Developmental neurotoxicity resulting from pharmacotherapy of preterm labor, modeled in vitro: Terbutaline and dexamethasone, separately and together. Toxicology 2018. [PMID: 29524569 DOI: 10.1016/j.tox.2018.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Terbutaline and dexamethasone are used in the management of preterm labor, often for durations of treatment exceeding those recommended, and both have been implicated in increased risk of neurodevelopmental disorders. We used a variety of cell models to establish the critical stages at which neurodifferentiation is vulnerable to these agents and to determine whether combined exposures produce a worsened outcome. Terbutaline selectively promoted the initial emergence of glia from embryonic neural stem cells (NSCs). The target for terbutaline shifted with developmental stage: at later developmental stages modeled with C6 and PC12 cells, terbutaline had little effect on glial differentiation (C6 cells) but impaired the differentiation of neuronotypic PC12 cells into neurotransmitter phenotypes. In contrast to the specificity shown by terbutaline, dexamethasone affected both neuronal and glial differentiation at all stages, impairing the emergence of both cell types in NSCs but with a much greater impairment for glia. At later stages, dexamethasone promoted glial cell differentiation (C6 cells), while shifting neuronal cell differentiation so as to distort the balance of neurotransmitter phenotypes (PC12 cells). Finally, terbutaline and dexamethasone interacted synergistically at the level of late stage glial cell differentiation, with dexamethasone boosting the ability of terbutaline to enhance indices of glial cell growth and neurite formation while producing further decrements in glial cell numbers. Our results support the conclusion that terbutaline and dexamethasone are directly-acting neuroteratogens, and further indicate the potential for their combined use in preterm labor to worsen neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Theodore A Slotkin
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC, 27710, USA.
| | - Samantha Skavicus
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Frederic J Seidler
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC, 27710, USA
| |
Collapse
|
47
|
|
48
|
DNA methylation mediates the effect of maternal cognitive appraisal of a disaster in pregnancy on the child's C-peptide secretion in adolescence: Project Ice Storm. PLoS One 2018; 13:e0192199. [PMID: 29401509 PMCID: PMC5798828 DOI: 10.1371/journal.pone.0192199] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 01/19/2018] [Indexed: 12/31/2022] Open
Abstract
Animal and human studies suggest that prenatal exposure to stress is associated with adverse health outcomes such as type 2 diabetes. Epigenetic modification, such as DNA methylation, is considered one possible underlying mechanism. The 1998 Quebec ice storm provides a unique opportunity to study an independent prenatal stressor on child outcomes. C-peptide is the best measure of endogenous insulin secretion and is widely used in the clinical management of patients with diabetes. The objectives of this study are to determine 1) the extent to which prenatal exposure to disaster-related stress (maternal objective hardship and maternal cognitive appraisal) influences children’s C-peptide secretion, and 2) whether DNA methylation of diabetes-related genes mediates the effects of prenatal stress on C-peptide secretion. Children’s (n = 30) C-peptide secretion in response to an oral glucose tolerance test were assessed in blood at 13½ years. DNA methylation levels of selected type 1 and 2 diabetes-related genes were chosen based upon the genes associated with prenatal maternal objective hardship and/or cognitive appraisal levels. Bootstrapping analyses were performed to determine the mediation effect of DNA methylation. We found that children whose mothers experienced higher objective hardship exhibited higher C-peptide secretion. Cognitive appraisal was not directly associated with C-peptide secretion. DNA methylation of diabetes-related genes had a positive mediation effect of objective hardship on C-peptide secretion: higher objective hardship predicted higher C-peptide secretion through DNA methylation. Negative mediation effects of cognitive appraisal were observed: negative cognitive appraisal predicted higher C-peptide secretion through DNA methylation. However, only one gene, LTA, remained a significant mediator of cognitive appraisal on C-peptide secretion after the conservative Bonferroni multiple corrections. Our findings suggest that DNA methylation could act as an intervening variable between prenatal stress and metabolic outcomes, highlighting the importance of epigenetic mechanisms in response to environmental factors.
Collapse
|
49
|
Miranda A, Sousa N. Maternal hormonal milieu influence on fetal brain development. Brain Behav 2018; 8:e00920. [PMID: 29484271 PMCID: PMC5822586 DOI: 10.1002/brb3.920] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/15/2017] [Accepted: 12/06/2017] [Indexed: 12/23/2022] Open
Abstract
An adverse maternal hormonal environment during pregnancy can be associated with abnormal brain growth. Subtle changes in fetal brain development have been observed even for maternal hormone levels within the currently accepted physiologic ranges. In this review, we provide an update of the research data on maternal hormonal impact on fetal neurodevelopment, giving particular emphasis to thyroid hormones and glucocorticoids. Thyroid hormones are required for normal brain development. Despite serum TSH appearing to be the most accurate indicator of thyroid function in pregnancy, maternal serum free T4 levels in the first trimester of pregnancy are the major determinant of postnatal psychomotor development. Even a transient period of maternal hypothyroxinemia at the beginning of neurogenesis can confer a higher risk of expressive language and nonverbal cognitive delays in offspring. Nevertheless, most recent clinical guidelines advocate for targeted high-risk case finding during first trimester of pregnancy despite universal thyroid function screening. Corticosteroids are determinant in suppressing cell proliferation and stimulating terminal differentiation, a fundamental switch for the maturation of fetal organs. Not surprisingly, intrauterine exposure to stress or high levels of glucocorticoids, endogenous or synthetic, has a molecular and structural impact on brain development and appears to impair cognition and increase anxiety and reactivity to stress. Limbic regions, such as hippocampus and amygdala, are particularly sensitive. Repeated doses of prenatal corticosteroids seem to have short-term benefits of less respiratory distress and fewer serious health problems in offspring. Nevertheless, neurodevelopmental growth in later childhood and adulthood needs further clarification. Future studies should address the relevance of monitoring the level of thyroid hormones and corticosteroids during pregnancy in the risk stratification for impaired postnatal neurodevelopment.
Collapse
Affiliation(s)
- Alexandra Miranda
- Life and Health Sciences Research Institute (ICVS)School of MedicineUniversity of MinhoBragaPortugal
- ICVS/3B's ‐ PT Government Associate LaboratoryBraga/GuimarãesPortugal
- Department of Obstetrics and GynecologyHospital de BragaBragaPortugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS)School of MedicineUniversity of MinhoBragaPortugal
- ICVS/3B's ‐ PT Government Associate LaboratoryBraga/GuimarãesPortugal
- Clinic Academic Center ‐ 2CABragaPortugal
| |
Collapse
|
50
|
Ihde ES, Zamudio S, Loh JM, Zhu Y, Woytanowski J, Rosen L, Liu M, Buckley B. Application of a novel mass spectrometric (MS) method to examine exposure to Bisphenol-A and common substitutes in a maternal fetal cohort. HUMAN AND ECOLOGICAL RISK ASSESSMENT : HERA 2017; 24:331-346. [PMID: 31588171 PMCID: PMC6777866 DOI: 10.1080/10807039.2017.1381831] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 09/16/2017] [Indexed: 05/26/2023]
Abstract
The use of Bisphenol A (BPA) has widely been replaced in consumer products by analogs BPB, BPE, BPF, BPS, and BPAF. Recent studies have linked these substitutes to similar adverse health outcomes as BPA, including disruption of endocrine pathways in animal and human studies. We designed a novel MS method, developed specifically for this study, to capture the most relevant BPA alternatives, BPB, BPE, BPF, BPS, BPAF and 4-NP in human blood and urine to quantify potential in utero exposures. To our knowledge, this is the first study to explore in utero exposure to these BPA analogs and the first U.S. study to test for BPA in maternal/fetal pairs. The method was run on 30 paired maternal urine and fetal cord blood samples from mothers undergoing elective Caesarean sections. 90% of mothers and 77% of babies tested positive for at least one BP analog. 83% of mothers tested positive for BPAF, 60% for BPS, 57% for BPB, 17% for BPF and 7% for BPA. 57% of babies tested positive for BPAF and 50% for BPF. BPA and BPB were detected in one cord blood sample each. BPS was not detected in cord blood. BPE was not detected in any fetal cord blood or maternal urine samples. These findings demonstrate the pervasiveness of some BP analogs in pregnant women and their babies at birth.
Collapse
Affiliation(s)
- Erin Speiser Ihde
- The Deirdre Imus Environmental Health Center®, Hackensack University Medical Center, 30 Prospect Ave, Research Building, Hackensack NJ 07601, USA
| | - Stacy Zamudio
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine and Surgery, Hackensack University Medical Center, 30 Prospect Ave, Hackensack NJ 07601, USA
| | - Ji Meng Loh
- Dept. of Mathematical Sciences, NJ Institute of Technology, University Heights, Newark, NJ 07102, USA
| | - Yalin Zhu
- Dept. of Mathematical Sciences, NJ Institute of Technology, University Heights, Newark, NJ 07102, USA
| | - John Woytanowski
- St. George’s University School of Medicine, Grenada, West Indies
- Drexel University College of Medicine, Dept. of Internal Medicine, 2900 W. Queen Lane, Philadelphia, PA 19129, USA
| | - Lawrence Rosen
- The Deirdre Imus Environmental Health Center®, Hackensack University Medical Center, 30 Prospect Ave, Research Building, Hackensack NJ 07601, USA
| | - Min Liu
- Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Road, Piscataway, NJ08854, USA
| | - Brian Buckley
- Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Road, Piscataway, NJ08854, USA
| |
Collapse
|