1
|
Zhang S, Ma M, Zhang Y, Luo J, Ouyang F, Tian Y, Gao Y. Maternal psychological distress modifies the association between prenatal exposure to per- and polyfluoroalkyl substances and infants' neurodevelopment. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 977:179351. [PMID: 40215636 DOI: 10.1016/j.scitotenv.2025.179351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/03/2025] [Accepted: 04/03/2025] [Indexed: 04/25/2025]
Abstract
Prenatal PFAS exposure and maternal psychological distress may adversely affect child neurodevelopment through shared biological pathways, such as hypothalamic-pituitary-adrenal (HPA) axis disruption and pro-inflammatory responses. However, whether psychological distress modifies PFAS-related neurodevelopmental risks remains unclear. Using data from the Shanghai Birth Cohort including 1779 mother-child pairs, we measured maternal PFAS levels during the first trimester and assessed maternal psychological distress (perceived stress, depression, and anxiety) during the second trimester. Child neurodevelopment was evaluated at 6 months of age using the Ages & Stages Questionnaires-Third Edition (ASQ-3). Multivariable regression models and quantile g-computation were conducted to evaluate the associations of ASQ-3 scores with individual and mixture PFAS. Stratified analyses were conducted between the psychological distress positive group (experienced any one type of stressor) and negative group (experienced no stressors). We found that 41 % of pregnant women experienced at least one type of psychological distress. Among the overall participants, maternal PFAS exposure was associated with reduced gross motor scores in children. Maternal psychological distress status modified the association between PFAS and gross motor scores. Specifically, the adverse associations of PFAS with gross motor development were only observed in the psychological distress positive group, while no association was found in the negative group. Significant interaction effects were observed between maternal psychological distress and most PFAS compounds (all P for interaction <0.1). Furthermore, the adverse associations of prenatal PFAS exposure with gross motor development intensified in women experiencing multiple types of psychological stressors. This study indicates that maternal psychological distress may increase the risk of PFAS-related adverse gross motor development in six-month-old children. Investigating maternal psychological distress could be crucial for identifying vulnerable populations and guiding intervention measures.
Collapse
Affiliation(s)
- Shanyu Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China; Department of Neurology, The Second Affiliated Hospital of Anhui Medical University, 230601 Hefei, China
| | - Mingyue Ma
- Department of Toxicology, School of Public Health, Shenyang Medical College, 110034 Shenyang, China
| | - Yan Zhang
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jiajun Luo
- Institute for Population and Precision Health, the University of Chicago, 60637, Chicago, IL, United States
| | - Fengxiu Ouyang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| | - Ying Tian
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China.
| | - Yu Gao
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China; Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| |
Collapse
|
2
|
Carter SWD, Kemp MW. A review of the potential off-target effects of antenatal steroid exposures on fetal development. J Dev Orig Health Dis 2025; 16:e18. [PMID: 40135629 DOI: 10.1017/s2040174425000078] [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: 03/27/2025]
Abstract
Antenatal steroids (ANS) are one of the most widely prescribed medications in pregnancy, being administered to women at risk of preterm delivery. In the setting of preterm delivery at or below 35 weeks' gestation, systematic review data show ANS reduce perinatal morbidity and mortality, primarily by promoting fetal lung maturation. However, with the expanding use of this intervention has come a growing appreciation for the potential off-target, adverse effects of ANS therapy on wider fetal development. We undertook a narrative literature review of the animal and clinical literature to assess current evidence for adverse effects of ANS exposure and fetal development. This review presents a summary of the evidence relating to the potential for wide-ranging, off-target, adverse effects of ANS therapy on fetal development and programming. We highlight an urgent need for further animal and clinical studies investigating the effects of ANS on the fetal immune, cardiovascular, renal and hepatic systems given a current sparsity of evidence. We also strongly suggest an emphasis on open disclosure, discussion and education of clinicians and patients with regard to the potential benefits and risks of ANS therapy, particularly in late preterm and term gestations where infants derive relatively few benefits from these drugs. We also propose further studies on the optimisation of ANS therapy through improved patient selection and improved dosing regimens based on a pharmacokinetic-pharmacodynamic informed understanding of ANS action on the fetal lung.
Collapse
Affiliation(s)
- Sean W D Carter
- Department of Obstetrics and Gynaecology, National University of Singapore, Singapore, Singapore
- King Edward Memorial Hospital, Perth, Western Australia, Australia
- Women and Infants Research Foundation, Perth, Western Australia, Australia
| | - Matthew W Kemp
- Department of Obstetrics and Gynaecology, National University of Singapore, Singapore, Singapore
- Women and Infants Research Foundation, Perth, Western Australia, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| |
Collapse
|
3
|
Chougula PV, Deshpande S, Datar C, Suryawanshi P. Antenatal diagnosis and early postnatal management of a neonate with type 1 familial glucocorticoid deficiency. BMJ Case Rep 2025; 18:e264598. [PMID: 40000035 DOI: 10.1136/bcr-2024-264598] [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: 02/27/2025] Open
Abstract
Familial glucocorticoid deficiency (FGD) is a rare inherited cause of primary adrenal insufficiency, characterised by cortisol deficiency, without mineralocorticoid involvement. Affected patients commonly present in infancy or early childhood with hypoglycaemia, seizures, generalised hyperpigmentation and failure to thrive. Late diagnosis may lead to adverse neurological outcomes, usually resulting from repeated hypoglycaemic episodes. A family history of sibling deaths or affected relatives is often observed. Mutations in the gene encoding adrenocorticotropin receptor (melanocortin 2 receptor, MC2R), comprise about 25% of FGD cases (type 1 FGD). Here, we describe an infant born to parents with third-degree consanguinity and a history of unexplained neonatal deaths in two previous siblings, who had hyperpigmentation and hypoglycaemia. Genetic testing revealed both parents to be heterozygous for the MC2R gene variant c.701C>C/T (p.Pro234Leu). In the current pregnancy, amniocentesis performed for prenatal diagnosis confirmed the fetus to be homozygous for the same mutation as the parents, indicating the fetus would be affected with type I FGD. After birth, the infant was managed in the neonatal intensive care unit, and despite markedly low cortisol levels, prompt initiation of glucocorticoid replacement therapy resulted in the prevention of hypoglycaemia and adrenal crisis, with a favourable outcome. Our case is unique due to the antenatal diagnosis of FGD, allowing for proactive postnatal management and prevention of complications.
Collapse
Affiliation(s)
| | - Sujata Deshpande
- Neonatology, Bharati Vidyapeeth University, Pune, Maharashtra, India
| | | | | |
Collapse
|
4
|
Wen J, McCann S, Balevic SJ, Muller WJ, Hornik CD, Autmizguine J, Anderson SG, Payne EH, Turdalieva S, Gonzalez D. Pharmacokinetics of Dexamethasone in Children and Adolescents with Obesity. J Clin Pharmacol 2024; 64:1491-1504. [PMID: 39120865 PMCID: PMC11598673 DOI: 10.1002/jcph.6108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024]
Abstract
Dexamethasone is a synthetic glucocorticoid approved for treating disorders of various organ systems in both adult and pediatric populations. Currently, approved pediatric dosing recommendations are weight-based, but it is unknown whether differences in dexamethasone drug disposition and exposure exist for children with obesity. This study aimed to develop a population pharmacokinetic (PopPK) model for dexamethasone with data collected from children with obesity. Dexamethasone was given as either IV or oral/enteral administration, and a salt factor correction was used for dexamethasone sodium phosphate injection. A PopPK analysis using dexamethasone plasma concentration versus time was performed using the software NONMEM. A virtual population of 1000 children with obesity across three age groups was generated for dosing simulations. Data from 59 study participants with 82 PK plasma samples were used in the PopPK analysis. A one-compartment model with first-order absorption and the inclusion of total body weight as a covariate characterized the data. No other covariates were included in the PopPK model. Single and multiple IV dose(s) of 0.5 and 1 mg/kg every 8 h resulted in 68% or more of virtual children with obesity attaining simulated exposures that were within exposure ranges previously reported in adult studies. In conclusion, this was the first study to characterize dexamethasone's PopPK in children with obesity. Simulation results suggest that virtual children with obesity receiving oral doses of 0.5 and 1 mg/kg had generally comparable dexamethasone exposures as adult estimates. Additional studies are needed to characterize the dexamethasone's target exposure in children.
Collapse
Affiliation(s)
- Jiali Wen
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sean McCann
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - William J. Muller
- Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Chi D. Hornik
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Julie Autmizguine
- Department of Pediatrics, Center Hospitalier Universitaire Sainte-Justine, Monetreal, Quebec, Canada
| | | | | | | | - Daniel Gonzalez
- Duke Clinical Research Institute, Durham, NC, USA
- Division of Clinical Pharmacology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
5
|
Nakaki A, Denaro E, Crimella M, Castellani R, Vellvé K, Izquierdo N, Basso A, Paules C, Casas R, Benitez L, Casas I, Larroya M, Genero M, Castro‐Barquero S, Gomez‐Gomez A, Pozo ÓJ, Vieta E, Estruch R, Nadal A, Gratacós E, Crovetto F, Crispi F, Youssef L. Effect of Mediterranean diet or mindfulness-based stress reduction during pregnancy on placental volume and perfusion: A subanalysis of the IMPACT BCN randomized clinical trial. Acta Obstet Gynecol Scand 2024; 103:2042-2052. [PMID: 39037192 PMCID: PMC11426209 DOI: 10.1111/aogs.14874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 07/23/2024]
Abstract
INTRODUCTION The IMPACT BCN trial-a parallel-group randomized clinical trial where 1221 pregnant women at high risk for small-for-gestational age (SGA) newborns were randomly allocated at 19- to 23-week gestation into three groups: Mediterranean diet, Mindfulness-based Stress reduction or non-intervention-has demonstrated a positive effect of Mediterranean diet and Stress reduction in the prevention of SGA. However, the mechanism of action of these interventions remains still unclear. The aim of this study is to investigate the effect of Mediterranean diet and Stress reduction on placental volume and perfusion. MATERIAL AND METHODS Participants in the Mediterranean diet group received monthly individual and group educational sessions, and free provision of extra-virgin olive oil and walnuts. Women in the Stress reduction group underwent an 8-week Stress reduction program adapted for pregnancy, consisting of weekly 2.5-h and one full-day sessions. Non-intervention group was based on usual care. Placental volume and perfusion were assessed in a subgroup of randomly selected women (n = 165) using magnetic resonance (MR) at 36-week gestation. Small placental volume was defined as MR estimated volume <10th centile. Perfusion was assessed by intravoxel incoherent motion. RESULTS While mean MR placental volume was similar among the study groups, both interventions were associated with a lower prevalence of small placental volume (3.9% Mediterranean diet and 5% stress reduction vs. 17% non-intervention; p = 0.03 and p = 0.04, respectively). Logistic regression showed that small placental volume was significantly associated with higher risk of SGA in both study groups (OR 7.48 [1.99-28.09] in Mediterranean diet and 20.44 [5.13-81.4] in Stress reduction). Mediation analysis showed that the effect of Mediterranean diet on SGA can be decomposed by a direct effect and an indirect effect (56.6%) mediated by a small placental volume. Similarly, the effect of Stress reduction on SGA is partially mediated (45.3%) by a small placental volume. Results on placental intravoxel incoherent motion perfusion fraction and diffusion coefficient were similar among the study groups. CONCLUSIONS Structured interventions during pregnancy based on Mediterranean diet or Stress reduction are associated with a lower proportion of small placentas, which is consistent with the previously observed beneficial effects of these interventions on fetal growth.
Collapse
Affiliation(s)
- Ayako Nakaki
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS)BarcelonaSpain
| | - Eugenio Denaro
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
| | - Maddalena Crimella
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
| | - Roberta Castellani
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
| | - Kilian Vellvé
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
| | - Nora Izquierdo
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
| | - Annachiara Basso
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
- Department of Obstetrics and Pediatrics ASST LeccoA. Manzoni HospitalLeccoItaly
| | - Cristina Paules
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
- Instituto de Investigación Sanitaria Aragón (IISAragon), Red de Salud Materno Infantil y del Desarrollo (SAMID), RETICS, Instituto de Salud Carlos III (ISCIII)Subdirección General de Evaluación y Fomento de la Investigación y Fondo Europeo de Desarrollo Regional (FEDER)ZaragozaSpain
| | - Rosa Casas
- Department of Internal Medicine Hospital Clinic, IDIBAPSUniversity of BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN)Institute de Salud Carlos IIIMadridSpain
- Institut de Recerca en Nutrició i Seguretat Alimentaria (INSA‐UB)University of BarcelonaBarcelonaSpain
| | - Leticia Benitez
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS)BarcelonaSpain
| | - Irene Casas
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS)BarcelonaSpain
| | - Marta Larroya
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS)BarcelonaSpain
| | - Mariona Genero
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
- Institut de Recerca Sant Joan de DéuEsplugues de LlobregatSpain
| | - Sara Castro‐Barquero
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
- Department of Internal Medicine Hospital Clinic, IDIBAPSUniversity of BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN)Institute de Salud Carlos IIIMadridSpain
- Institut de Recerca en Nutrició i Seguretat Alimentaria (INSA‐UB)University of BarcelonaBarcelonaSpain
| | - Alex Gomez‐Gomez
- Integrative Pharmacology and Systems Neuroscience GroupIMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
| | - Óscar J. Pozo
- Integrative Pharmacology and Systems Neuroscience GroupIMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
| | - Eduard Vieta
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS)BarcelonaSpain
- Department of Psychiatry and Psychology, Hospital Clinic, Neuroscience InstituteUniversity of Barcelona, CIBERSAMBarcelonaSpain
| | - Ramon Estruch
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS)BarcelonaSpain
- Department of Internal Medicine Hospital Clinic, IDIBAPSUniversity of BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN)Institute de Salud Carlos IIIMadridSpain
- Institut de Recerca en Nutrició i Seguretat Alimentaria (INSA‐UB)University of BarcelonaBarcelonaSpain
| | - Alfons Nadal
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS)BarcelonaSpain
- Department of Pathology, Hospital ClinicUniversity of BarcelonaBarcelonaSpain
| | - Eduard Gratacós
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS)BarcelonaSpain
- Institut de Recerca Sant Joan de DéuEsplugues de LlobregatSpain
- Center for Biomedical Research on Rare Diseases (CIBER‐ER)MadridSpain
| | - Francesca Crovetto
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
- Institut de Recerca Sant Joan de DéuEsplugues de LlobregatSpain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin RD21/0012/0003Instituto de Salud Carlos IIIMadridSpain
| | - Fàtima Crispi
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS)BarcelonaSpain
- Center for Biomedical Research on Rare Diseases (CIBER‐ER)MadridSpain
| | - Lina Youssef
- BCNatal – Barcelona Center for Maternal‐Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu)University of BarcelonaBarcelonaSpain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS)BarcelonaSpain
- Institut de Recerca Sant Joan de DéuEsplugues de LlobregatSpain
- Josep Carreras Leukemia Research InstituteHospital Clinic/University of Barcelona CampusBarcelonaSpain
| |
Collapse
|
6
|
Carter SWD, Fee EL, Usuda H, Oguz G, Ramasamy A, Amin Z, Agnihotri B, Wei Q, Xiawen L, Takahashi T, Takahashi Y, Ikeda H, Kumagai Y, Saito Y, Saito M, Mattar C, Evans MI, Illanes SE, Jobe AH, Choolani M, Kemp MW. Antenatal steroids elicited neurodegenerative-associated transcriptional changes in the hippocampus of preterm fetal sheep independent of lung maturation. BMC Med 2024; 22:338. [PMID: 39183288 PMCID: PMC11346182 DOI: 10.1186/s12916-024-03542-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Antenatal steroid therapy for fetal lung maturation is routinely administered to women at risk of preterm delivery. There is strong evidence to demonstrate benefit from antenatal steroids in terms of survival and respiratory disease, notably in infants delivered at or below 32 weeks' gestation. However, dosing remains unoptimized and lung benefits are highly variable. Current treatment regimens generate high-concentration, pulsatile fetal steroid exposures now associated with increased risk of childhood neurodevelopmental diseases. We hypothesized that damage-associated changes in the fetal hippocampal transcriptome would be independent of preterm lung function. METHODS Date-mated ewes carrying a single fetus at 122 ± 2dGA (term = 150dGA) were randomized into 4 groups: (i) Saline Control Group, 4×2ml maternal saline intramuscular(IM) injections at 12hr intervals (n = 11); or (ii) Dex High Group, 2×12mg maternal IM dexamethasone phosphate injections at 12hr intervals followed by 2×2ml IM saline injections at 12hr intervals (n = 12; representing a clinical regimen used in Singapore); or (iii) Dex Low Group, 4×1.5mg maternal IM dexamethasone phosphate injections 12hr intervals (n = 12); or (iv) Beta-Acetate Group, 1×0.125mg/kg maternal IM betamethasone acetate injection followed by 3×2ml IM sterile normal saline injections 12hr intervals (n = 8). Lambs were surgically delivered 48hr after first maternal injection at 122-125dGA, ventilated for 30min to establish lung function, and euthanised for necropsy and tissue collection. RESULTS Preterm lambs from the Dex Low and Beta-Acetate Groups had statistically and biologically significant lung function improvements (measured by gas exchange, lung compliance). Compared to the Saline Control Group, hippocampal transcriptomic data identified 879 differentially significant expressed genes (at least 1.5-fold change and FDR < 5%) in the steroid-treated groups. Pulsatile dexamethasone-only exposed groups (Dex High and Dex Low) had three common positively enriched differentially expressed pathways related in part to neurodegeneration ("Prion Disease", "Alzheimer's Disease", "Arachidonic Acid metabolism"). Adverse changes were independent of respiratory function during ventilation. CONCLUSIONS Our data suggests that exposure to antenatal steroid therapy is an independent cause of damage- associated transcriptomic changes in the brain of preterm, fetal sheep. These data highlight an urgent need for careful reconsideration and balancing of how antenatal steroids are used, both for patient selection and dosing regimens.
Collapse
Affiliation(s)
- Sean W D Carter
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore.
| | - Erin L Fee
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
| | - Haruo Usuda
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Gokce Oguz
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome #02-01, Singapore, 138632, Republic of Singapore
| | - Adaikalavan Ramasamy
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome #02-01, Singapore, 138632, Republic of Singapore
| | - Zubair Amin
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Neonatology Khoo Teck Puat, National University Children's Medical Institute, National University Hospital, Singapore, Singapore
| | - Biswas Agnihotri
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Neonatology Khoo Teck Puat, National University Children's Medical Institute, National University Hospital, Singapore, Singapore
| | - Qin Wei
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Liu Xiawen
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Tsukasa Takahashi
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yuki Takahashi
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Hideyuki Ikeda
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yusaku Kumagai
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yuya Saito
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Masatoshi Saito
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Citra Mattar
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Mark I Evans
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Fetal Medicine Foundation of America, New York, NY, USA
| | - Sebastián E Illanes
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Reproductive Biology Program, Center for Biomedical Research and Innovation, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Alan H Jobe
- Centre for Pulmonary Biology, Cincinnati Children's Hospital Medical Centre, Cincinnati, OH, USA
| | - Mahesh Choolani
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Matthew W Kemp
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
- Women and Infants Research Foundation, Perth, WA, Australia
| |
Collapse
|
7
|
Briceño-Pérez C, Briceño-Sanabria L, Briceño-Sanabria C, Reyna-Villasmil E. Early life corticosteroid overexposure: Epigenetic and fetal origins of adult diseases. Int J Gynaecol Obstet 2024; 164:40-46. [PMID: 37318113 DOI: 10.1002/ijgo.14914] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/30/2023] [Accepted: 05/25/2023] [Indexed: 06/16/2023]
Abstract
The relationship between events occurring during intrauterine development and later-life predisposition to long-term disease, has been described. The fetus responds to excess intrauterine exposure to high levels of corticosteroids, modifying their physiological development and stopping their growth. Fetal exposure to elevated levels of either endogenous (alterations in fetal hypothalamic-pituitary-adrenal axis) or synthetic corticosteroids, is one model of early-life adversity; to developing adult disease. At the molecular level, there are transcriptional changes in metabolic and growth pathways. Epigenetic mechanisms participate in transgenerational inheritance, not genomic. Exposures that change 11β-hydroxysteroid dehydrogenase type 2 enzyme methylation status in the placenta can result in transcriptional repression of the gene, causing the fetus to be exposed to higher levels of cortisol. More precise diagnosis and management of antenatal corticosteroids for preterm birth, would potentially decrease the risk of long-term adverse outcomes. More studies are needed to understand the potential roles of factors to alter fetal corticosteroid exposure. Long-term infant follow-up is required to determine whether methylation changes in placenta may represent useful biomarkers of later disease risk. This review, summarize recent advances in the programming of fetal effects of corticosteroid exposure, the role of corticosteroids in epigenetic gene regulation of placental 11β-hydroxysteroid dehydrogenase type 2 enzyme expression and transgenerational effects.
Collapse
Affiliation(s)
- Carlos Briceño-Pérez
- Department of Obstetrics and Gynecology, University of Zulia, Maracaibo, Venezuela
| | | | | | | |
Collapse
|
8
|
Mariño-Narvaez C, Puertas-Gonzalez JA, Romero-Gonzalez B, Gonzalez-Perez R, Peralta-Ramírez MI. How prenatal cortisol levels may differentially affect the neurodevelopment of boys and girls. Early Hum Dev 2023; 187:105874. [PMID: 37866289 DOI: 10.1016/j.earlhumdev.2023.105874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/24/2023]
Abstract
BACKGROUND Prenatal stress could have serious consequences on maternal and fetal health. In this sense, some studies have stated that maternal HCC during pregnancy could contribute to sex-specific effects on infant neurodevelopment, following the Developmental Origins of Health and Disease Hypothesis. AIM This study aimed to determine whether maternal hair cortisol concentration (HCC) during each trimester of pregnancy and postpartum could predict the neurodevelopmental outcomes of their 12-month-old offspring, with sex-specific differences considered. STUDY DESIGN longitudinal. SUBJECTS The study involved 93 pregnant women and their babies. OUTCOME MEASURE Hair samples collected during each trimester and postpartum and The Bayley Scales for Infant Development III was used to assess the infants' abilities. RESULTS The results showed that maternal HCC during the first and second trimesters could predict language and motor abilities. However, when discriminated by sex, only females' cognitive, expressive language, and fine and gross motor skills were predicted by cortisol, not males. CONCLUSIONS These findings support the idea that non-toxic levels of cortisol can positively influence infants' neurodevelopment.
Collapse
Affiliation(s)
| | | | - Borja Romero-Gonzalez
- Department of Psychology, Faculty of Education, Campus Duques de Soria, University of Valladolid, Soria, Spain.
| | - Raquel Gonzalez-Perez
- Pharmacology Departament, CIBERehd, Faculty of Pharmacy, University de Granada, Spain
| | - Maria Isabel Peralta-Ramírez
- Mind, Brain and Behavior Research Center (CIMCYC), University of Granada, Granada, Spain; Department of Personality, Assessment and Psychological Treatment, Faculty of Psychology, University of Granada, Granada, Spain
| |
Collapse
|
9
|
Bahsoun A, Fakih Y, Zareef R, Bitar F, Arabi M. Corticosteroids in COVID-19: pros and cons. Front Med (Lausanne) 2023; 10:1202504. [PMID: 37644981 PMCID: PMC10461317 DOI: 10.3389/fmed.2023.1202504] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/31/2023] [Indexed: 08/31/2023] Open
Abstract
The wide and rapid spread of the COVID-19 pandemic has placed an unanticipated burden on the global healthcare sector. This necessitated a swift response from the international community to reach a solution. Efforts were made in parallel to develop preventative and therapeutic modalities. Since then, drug repurposing has blossomed as a potentially rapid resolution and has included various agents with anti-viral and anti-inflammatory properties. Corticosteroids, being potent anti-inflammatory agents, have been placed under extensive investigation. Various trials have recorded the beneficial outcome of corticosteroids in decreasing the mortality and morbidity of COVID-19. With the high pace of escalating events, the quality and study design of clinical trials are varied. Therefore, this study aims to explore the role of corticosteroids in COVID-19 disease. It inspects the molecular, pharmacologic, and clinical proof behind this theory.
Collapse
Affiliation(s)
- Aymen Bahsoun
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Yeva Fakih
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rana Zareef
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Pediatric Department, American University of Beirut Medical Center, Beirut, Lebanon
| | - Fadi Bitar
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Pediatric Department, American University of Beirut Medical Center, Beirut, Lebanon
- Pediatric Department, Division of Pediatric Cardiology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mariam Arabi
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Pediatric Department, American University of Beirut Medical Center, Beirut, Lebanon
- Pediatric Department, Division of Pediatric Cardiology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
10
|
Balázs G, Balajthy A, Seri I, Hegyi T, Ertl T, Szabó T, Röszer T, Papp Á, Balla J, Gáll T, Balla G. Prevention of Chronic Morbidities in Extremely Premature Newborns with LISA-nCPAP Respiratory Therapy and Adjuvant Perinatal Strategies. Antioxidants (Basel) 2023; 12:1149. [PMID: 37371878 DOI: 10.3390/antiox12061149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Less invasive surfactant administration techniques, together with nasal continuous airway pressure (LISA-nCPAP) ventilation, an emerging noninvasive ventilation (NIV) technique in neonatology, are gaining more significance, even in extremely premature newborns (ELBW), under 27 weeks of gestational age. In this review, studies on LISA-nCPAP are compiled with an emphasis on short- and long-term morbidities associated with prematurity. Several perinatal preventative and therapeutic investigations are also discussed in order to start integrated therapies as numerous organ-saving techniques in addition to lung-protective ventilations. Two thirds of immature newborns can start their lives on NIV, and one third of them never need mechanical ventilation. With adjuvant intervention, these ratios are expected to be increased, resulting in better outcomes. Optimized cardiopulmonary transition, especially physiologic cord clamping, could have an additively beneficial effect on patient outcomes gained from NIV. Organ development and angiogenesis are strictly linked not only in the immature lung and retina, but also possibly in the kidney, and optimized interventions using angiogenic growth factors could lead to better morbidity-free survival. Corticosteroids, caffeine, insulin, thyroid hormones, antioxidants, N-acetylcysteine, and, moreover, the immunomodulatory components of mother's milk are also discussed as adjuvant treatments, since immature newborns deserve more complex neonatal interventions.
Collapse
Affiliation(s)
- Gergely Balázs
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - András Balajthy
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - István Seri
- First Department of Pediatrics, School of Medicine, Semmelweis University, 1083 Budapest, Hungary
- Keck School of Medicine of USC, Children's Hospital of Los Angeles, Los Angeles, CA 90033, USA
| | - Thomas Hegyi
- Department of Pediatrics, Division of Neonatology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Tibor Ertl
- Departments of Neonatology and Obstetrics & Gynecology, University of Pécs Medical School, 7624 Pécs, Hungary
- MTA-PTE Human Reproduction Scientific Research Group, University of Pécs, 7624 Pécs, Hungary
| | - Tamás Szabó
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Röszer
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Ágnes Papp
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - József Balla
- Department of Internal Medicine, Division of Nephrology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-UD Vascular Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Gáll
- Department of Internal Medicine, Division of Nephrology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - György Balla
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-UD Vascular Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
11
|
Sohn H. Structural Inequities in the Kin Safety Net: Mapping the Three-Generational Network throughout Early Adulthood 1. AJS; AMERICAN JOURNAL OF SOCIOLOGY 2023; 128:1650-1677. [PMID: 38736557 PMCID: PMC11085851 DOI: 10.1086/724817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Research in the intergenerational transmission of socioeconomic status (SES) consistently shows that the SES of one generation benefits the next. Demographic processes shape the kin structures that serve as conduits for the transmission of SES. Few studies have examined these trends together to describe experiences in evolving kin structures throughout the life course and across generations. This article applies demographic techniques to fertility, marital, and mortality data from three generations in the Panel Survey of Income Dynamics to simulate the amount of time young adults would spend within consequential kin structures. High-SES adults spend more years of their young adulthood in advantageous kin structures with greater potential for kin support and capital accumulation, while low-SES adults spend a larger portion of their young adulthoods as single parents, sandwiched between widowed parents and children, and as adult orphans. The kin network inequities have grown since the 1980s, driven by lagging mortality improvements and increasing single parenthood among low-SES families.
Collapse
|
12
|
Berry KJ, Chandran U, Mu F, Deochand DK, Lei T, Pagin M, Nicolis SK, Monaghan-Nichols AP, Rogatsky I, DeFranco DB. Genomic glucocorticoid action in embryonic mouse neural stem cells. Mol Cell Endocrinol 2023; 563:111864. [PMID: 36690169 PMCID: PMC10057471 DOI: 10.1016/j.mce.2023.111864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023]
Abstract
Prenatal exposure to synthetic glucocorticoids (sGCs) reprograms brain development and predisposes the developing fetus towards potential adverse neurodevelopmental outcomes. Using a mouse model of sGC administration, previous studies show that these changes are accompanied by sexually dimorphic alterations in the transcriptome of neural stem and progenitor cells (NSPCs) derived from the embryonic telencephalon. Because cell type-specific gene expression profiles tightly regulate cell fate decisions and are controlled by a flexible landscape of chromatin domains upon which transcription factors and enhancer elements act, we multiplexed data from four genome-wide assays: RNA-seq, ATAC-seq (assay for transposase accessible chromatin followed by genome wide sequencing), dual cross-linking ChIP-seq (chromatin immunoprecipitation followed by genome wide sequencing), and microarray gene expression to identify novel relationships between gene regulation, chromatin structure, and genomic glucocorticoid receptor (GR) action in NSPCs. These data reveal that GR binds preferentially to predetermined regions of accessible chromatin to influence gene programming and cell fate decisions. In addition, we identify SOX2 as a transcription factor that impacts the genomic response of select GR target genes to sGCs (i.e., dexamethasone) in NSPCs.
Collapse
Affiliation(s)
- Kimberly J Berry
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Uma Chandran
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Research Computing, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fangping Mu
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Research Computing, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dinesh K Deochand
- Hospital for Special Surgery Research Institute, The David Rosensweig Genomics Center, New York, USA
| | - T Lei
- Department of Biomedical Sciences, University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| | - Miriam Pagin
- Department of Biotechnology and Biosciences, University Milano-Bicocca, 20126, Milano, Italy
| | - Silvia K Nicolis
- Department of Biotechnology and Biosciences, University Milano-Bicocca, 20126, Milano, Italy
| | - A Paula Monaghan-Nichols
- Department of Biomedical Sciences, University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| | - Inez Rogatsky
- Hospital for Special Surgery Research Institute, The David Rosensweig Genomics Center, New York, USA; Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, USA
| | - Donald B DeFranco
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
13
|
Weiss SJ, Keeton V, Richoux S, Cooper B, Niemann S. Exposure to antenatal corticosteroids and infant cortisol regulation. Psychoneuroendocrinology 2023; 147:105960. [PMID: 36327758 PMCID: PMC9968454 DOI: 10.1016/j.psyneuen.2022.105960] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/16/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022]
Abstract
Administration of antenatal corticosteroids (AC) is the standard of care during pregnancy for women who are at risk of early delivery. Evidence indicates that AC improve survival and reduce morbidity for preterm infants. However, research suggests that infants whose mothers receive AC have an altered hypothalamic-pituitary-axis (HPA) response to stressors in early life. Results are mixed regarding the nature of these effects, with studies showing both suppressed and augmented HPA activity. In addition, research is very limited beyond the 4th month of life. The purpose of this study was to determine if AC exposure was associated with infant cortisol levels in a resting state or in response to a stressor at 1, 6 and 12 months postnatal. We also evaluated the moderating role of preterm birth in this association. 181 women and their infants participated in the study. Women were recruited during the 3rd trimester of pregnancy; at this time, they completed the Perceived Stress Scale and provided 8 salivary samples over a 2-day period for cortisol assay. They provided these data again at 6 and 12 months postnatal. At 1, 6, and 12 months postnatal, salivary samples were collected from infants to examine their cortisol levels before and after participation in a 'stressor protocol'. Data were extracted from the medical record on AC exposure, gestational age, maternal obstetric risk, and neonatal morbidity. Mixed effects multilevel regression modeling was used to examine the aims. Infants whose mothers received AC had significantly lower resting state (B = -2.47, CI: -3.691, -0.0484) and post-stressor (B = -2.51, CI: -4.283, -0.4276) cortisol levels across the first year of life than infants whose mothers did not receive AC. There was no moderating effect of preterm birth on the relationship between AC exposure and cortisol. Results indicate a state of dampened HPA activation and cortisol hypo-arousal that persists across the first year of life among infants who were exposed to corticosteroids in utero. Further research is needed to examine mechanisms responsible for any alterations that occur during development of the fetal HPA axis, including epigenetic and biochemical factors that control hormonal secretion, negative feedback, and glucocorticoid receptor function throughout the HPA axis. Findings warrant careful consideration by obstetric clinicians of the benefits and risks of prescribing AC.
Collapse
Affiliation(s)
- Sandra J. Weiss
- Department of Community Health Systems, University of California, San Francisco, USA,Correspondence to: Department of Community Health Systems, University of California, Box 0608, 2 Koret Way, San Francisco, CA, 94143, USA. (S.J. Weiss)
| | - Victoria Keeton
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, USA
| | - Sarah Richoux
- Department of Community Health Systems, University of California, San Francisco, USA
| | - Bruce Cooper
- Department of Community Health Systems, University of California, San Francisco, USA
| | - Sandra Niemann
- Department of Community Health Systems, University of California, San Francisco, USA
| |
Collapse
|
14
|
Dubey H, Sharma RK, Krishnan S, Knickmeyer R. SARS-CoV-2 (COVID-19) as a possible risk factor for neurodevelopmental disorders. Front Neurosci 2022; 16:1021721. [PMID: 36590303 PMCID: PMC9800937 DOI: 10.3389/fnins.2022.1021721] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Pregnant women constitute one of the most vulnerable populations to be affected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, the cause of coronavirus disease 2019. SARS-CoV-2 infection during pregnancy could negatively impact fetal brain development via multiple mechanisms. Accumulating evidence indicates that mother to fetus transmission of SARS-CoV-2 does occur, albeit rarely. When it does occur, there is a potential for neuroinvasion via immune cells, retrograde axonal transport, and olfactory bulb and lymphatic pathways. In the absence of maternal to fetal transmission, there is still the potential for negative neurodevelopmental outcomes as a consequence of disrupted placental development and function leading to preeclampsia, preterm birth, and intrauterine growth restriction. In addition, maternal immune activation may lead to hypomyelination, microglial activation, white matter damage, and reduced neurogenesis in the developing fetus. Moreover, maternal immune activation can disrupt the maternal or fetal hypothalamic-pituitary-adrenal (HPA) axis leading to altered neurodevelopment. Finally, pro-inflammatory cytokines can potentially alter epigenetic processes within the developing brain. In this review, we address each of these potential mechanisms. We propose that SARS-CoV-2 could lead to neurodevelopmental disorders in a subset of pregnant women and that long-term studies are warranted.
Collapse
Affiliation(s)
- Harikesh Dubey
- Division of Neuroengineering, Institute for Quantitative Health Sciences and Engineering, Michigan State University, East Lansing, MI, United States
| | - Ravindra K. Sharma
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Suraj Krishnan
- Jacobi Medical Center, Albert Einstein College of Medicine, The Bronx, NY, United States
| | - Rebecca Knickmeyer
- Division of Neuroengineering, Institute for Quantitative Health Sciences and Engineering, Michigan State University, East Lansing, MI, United States,Department of Pediatrics and Human Development, Michigan State University, East Lansing, MI, United States,*Correspondence: Rebecca Knickmeyer,
| |
Collapse
|
15
|
Tang E, Wiencke JK, Warrier G, Hansen H, McCoy L, Rice T, Bracci PM, Wrensch M, Taylor JW, Clarke JL, Koestler DC, Salas LA, Christensen BC, Kelsey KT, Molinaro AM. Evaluation of cross-platform compatibility of a DNA methylation-based glucocorticoid response biomarker. Clin Epigenetics 2022; 14:136. [PMID: 36307860 PMCID: PMC9617416 DOI: 10.1186/s13148-022-01352-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 10/11/2022] [Indexed: 11/10/2022] Open
Abstract
Background Identifying blood-based DNA methylation patterns is a minimally invasive way to detect biomarkers in predicting age, characteristics of certain diseases and conditions, as well as responses to immunotherapies. As microarray platforms continue to evolve and increase the scope of CpGs measured, new discoveries based on the most recent platform version and how they compare to available data from the previous versions of the platform are unknown. The neutrophil dexamethasone methylation index (NDMI 850) is a blood-based DNA methylation biomarker built on the Illumina MethylationEPIC (850K) array that measures epigenetic responses to dexamethasone (DEX), a synthetic glucocorticoid often administered for inflammation. Here, we compare the NDMI 850 to one we built using data from the Illumina Methylation 450K (NDMI 450). Results The NDMI 450 consisted of 22 loci, 15 of which were present on the NDMI 850. In adult whole blood samples, the linear composite scores from NDMI 450 and NDMI 850 were highly correlated and had equivalent predictive accuracy for detecting DEX exposure among adult glioma patients and non-glioma adult controls. However, the NDMI 450 scores of newborn cord blood were significantly lower than NDMI 850 in samples measured with both assays. Conclusions We developed an algorithm that reproduces the DNA methylation glucocorticoid response score using 450K data, increasing the accessibility for researchers to assess this biomarker in archived or publicly available datasets that use the 450K version of the Illumina BeadChip array. However, the NDMI850 and NDMI450 do not give similar results in cord blood, and due to data availability limitations, results from sample types of newborn cord blood should be interpreted with care. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-022-01352-1.
Collapse
|
16
|
Lucas-Herald AK, Rodie ME, Ahmed SF. Update on the management of a newborn with a suspected difference of sex development. Arch Dis Child 2022; 107:866-871. [PMID: 34772663 DOI: 10.1136/archdischild-2020-320872] [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] [Received: 04/15/2021] [Accepted: 10/26/2021] [Indexed: 11/04/2022]
Abstract
Differences or disorders of sex development are a group of heterogeneous conditions, which most commonly present in the newborn period, with the appearance of atypical genitalia on newborn examination. Over recent years, the improvement in our knowledge of these conditions has been accompanied by advances in diagnostic technology and therapeutic options, as well as societal shifts in attitudes and expectations. These factors have placed an even greater emphasis than before on the need for early expert input through a multidisciplinary service that can support the patient and the family; perform and interpret the investigations required to reach a diagnosis; and formulate a management plan that lays down the foundation for optimal long-term outcome. While providing a regional service, the expert team should also be committed to research and quality improvement through participation in national and international networks.
Collapse
Affiliation(s)
| | - Martina E Rodie
- Office for Rare Conditions, University of Glasgow, Glasgow, UK
- Department of Neonatology, Queen Elizabeth University Hospital Campus, Glasgow, UK
| | - S Faisal Ahmed
- Developmental Endocrinology Research Group, University of Glasgow, Glasgow, UK
| |
Collapse
|
17
|
Manojlovic-Stojanoski M, Lavrnja I, Stevanovic I, Trifunovic S, Ristic N, Nestorovic N, Sévigny J, Nedeljkovic N, Laketa D. Antenatal Dexamethasone Treatment Induces Sex-dependent Upregulation of NTPDase1/CD39 and Ecto-5'-nucleotidase/CD73 in the Rat Fetal Brain. Cell Mol Neurobiol 2022; 42:1965-1981. [PMID: 33761054 PMCID: PMC11421702 DOI: 10.1007/s10571-021-01081-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/13/2021] [Indexed: 10/21/2022]
Abstract
Dexamethasone (DEX) is frequently used to treat women at risk of preterm delivery, but although indispensable for the completion of organ maturation in the fetus, antenatal DEX treatment may exert adverse sex-dimorphic neurodevelopmental effects. Literature findings implicated oxidative stress in adverse effects of DEX treatment. Purinergic signaling is involved in neurodevelopment and controlled by ectonucleotidases, among which in the brain the most abundant are ectonucleoside triphosphate diphosphohydrolase 1 (NTPDase1/CD39) and ecto-5'-nucleotidase (e5'NT/CD73), which jointly dephosphorylate ATP to adenosine. They are also involved in cell adhesion and migration, processes integral to brain development. Upregulation of CD39 and CD73 after DEX treatment was reported in adult rat hippocampus. We investigated the effects of maternal DEX treatment on CD39 and CD73 expression and enzymatic activity in the rat fetal brain of both sexes, in the context of oxidative status of the brain tissue. Fetuses were obtained at embryonic day (ED) 21, from Wistar rat dams treated with 0.5 mg DEX/kg/day, at ED 16, 17, and 18, and brains were processed and used for further analysis. Sex-specific increase in CD39 and CD73 expression and in the corresponding enzyme activities was induced in the brain of antenatally DEX-treated fetuses, more prominently in males. The oxidative stress induction after antenatal DEX treatment was confirmed in both sexes, although showing a slight bias in males. Due to the involvement of purinergic system in crucial neurodevelopmental processes, future investigations are needed to determine the role of these observed changes in the adverse effects of antenatal DEX treatment.
Collapse
Affiliation(s)
- Milica Manojlovic-Stojanoski
- Institute for Biological Research, "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research, "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ivana Stevanovic
- Medical Faculty of Military Medical Academy, Institute of Medical Research Belgrade, Belgrade, Serbia
| | - Svetlana Trifunovic
- Institute for Biological Research, "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Natasa Ristic
- Institute for Biological Research, "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Natasa Nestorovic
- Institute for Biological Research, "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jean Sévigny
- Département de Microbiologie-Infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec - Université Laval, Québec, Canada
| | - Nadezda Nedeljkovic
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Danijela Laketa
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
18
|
Prenatal Activation of Glucocorticoid Receptors Induces Memory Impairment in a Sex-Dependent Manner: Role of Cyclooxygenase-2. Mol Neurobiol 2022; 59:3767-3777. [PMID: 35396693 DOI: 10.1007/s12035-022-02820-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/30/2022] [Indexed: 10/18/2022]
Abstract
Prenatal exposure to dexamethasone (DEX) results in long-lasting effects on cognitive functions such as learning and memory impairment. However, the mechanisms underlying these DEX-induced deleterious effects are not well known. Here, we assessed whether cyclooxygenase-2 (COX-2) is involved in the impact of prenatal exposure to DEX on learning and memory during adulthood. Pregnant Sprague-Dawley rats received daily injections of either DEX (0.2 mg/kg; i.p.) or saline from gestation day (GD) 14 until GD21. Gene and protein expression of COX-2, as well as presynaptic (synaptophysin) and postsynaptic (postsynaptic density protein-95) proteins, were monitored in the dorsal and ventral hippocampi of adult male and female offspring. A different cohort of adult male and female rat offspring was given daily injections of either vehicle or a specific COX-2 inhibitor (celecoxib 10 mg/kg, i.p.) for 5 consecutive days and was subsequently subjected to Morris water maze memory test. Prenatal DEX enhanced the expression of COX-2 protein and cox-2 mRNA in the dorsal hippocampus of adult female but not male rats. This enhanced COX-2 expression was associated with reduced expression in pre- and postsynaptic proteins and altered memory acquisition and retention. Administration of COX-2-specific inhibitor alleviated prenatal DEX-induced memory impairment in adult female rats. This study suggests that prenatal activation of glucocorticoid receptors stimulates COX-2 gene and protein expression and impairs hippocampal-dependent spatial memory in female but not male rat offspring. Furthermore, COX-2 selective inhibitors can be used to alleviate the long-lasting deleterious effects of corticosteroid medication during pregnancy.
Collapse
|
19
|
Asztalos EV, Murphy KE, Matthews SG. A Growing Dilemma: Antenatal Corticosteroids and Long-Term Consequences. Am J Perinatol 2022; 39:592-600. [PMID: 33053595 DOI: 10.1055/s-0040-1718573] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE A single course of synthetic antenatal corticosteroids is standard care for women considered to be at risk for preterm birth before 34 weeks of gestation. While the intended target is the fetal lung, the fetal brain contains remarkably high levels of glucocorticoid receptors in structures critical in the regulation of behavior and endocrine function. Negative programming signals may occur which can lead to permanent maladaptive changes and predispose the infant/child to an increased risk in physical, mental, and developmental disorders. METHODS Framed around these areas of concerns for physical, mental, and developmental disorders, this narrative review drew on studies (animal and clinical), evaluating the long-term effects of antenatal corticosteroids to present the case that a more targeted approach to the use of antenatal corticosteroids for the betterment of the fetus urgently needed. RESULTS Studies raised concerns about the potential negative long-term consequences, especially for the exposed fetus who was born beyond the period of the greatest benefit from antenatal corticosteroids. The long-term consequences are more subtle in nature and usually manifest later in life, often beyond the scope of most clinical trials. CONCLUSION Continued research is needed to identify sufficient safety data, both short term and long term. Caution in the use of antenatal corticosteroids should be exercised while additional work is undertaken to optimize dosing strategies and better identify women at risk of preterm birth prior to administration of antenatal corticosteroids. KEY POINTS · A single-course ACS is a remarkable therapy with substantial benefits.. · There is a potential of long-term neurodevelopmental consequences in the ACS-exposed fetus.. · There is a need to improve dosing strategies and identification of appropriate at risk women..
Collapse
Affiliation(s)
- Elizabeth V Asztalos
- Department of Newborn and Developmental Paediatrics, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Kellie E Murphy
- Department of Obstetrics and Gynecology, Sinai Health Systems, University of Toronto, Toronto, Ontario, Canada
| | - Stephen G Matthews
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
van Dokkum NH, Bachini S, Verkaik-Schakel RN, Baptist DH, Salavati S, Kraft KE, Scherjon SA, Bos AF, Plösch T. Differential Placental DNA Methylation of NR3C1 in Extremely Preterm Infants With Poorer Neurological Functioning. Front Pediatr 2022; 10:876803. [PMID: 35722484 PMCID: PMC9198301 DOI: 10.3389/fped.2022.876803] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Understanding underlying mechanisms of neurodevelopmental impairment following preterm birth may enhance opportunities for targeted interventions. We aimed to assess whether placental DNA methylation of selected genes affected early neurological functioning in preterm infants. METHODS We included 43 infants, with gestational age <30 weeks and/or birth weight <1,000 g and placental samples at birth. We selected genes based on their associations with several prenatal conditions that may be related to poor neurodevelopmental outcomes. We determined DNA methylation using pyrosequencing, and neurological functioning at 3 months post-term using Prechtl's General Movement Assessment, including the Motor Optimality Score-Revised (MOS-R). RESULTS Twenty-four infants had atypical MOS-R, 19 infants had near-optimal MOS-R. We identified differences in average methylation of NR3C1 (encoding for the glucocorticoid receptor) [3.3% (95%-CI: 2.4%-3.9%) for near-optimal vs. 2.3% (95%-CI: 1.7%-3.0%), p = 0.008 for atypical], and at three of the five individual CpG-sites. For EPO, SLC6A3, TLR4, VEGFA, LEP and HSD11B2 we found no differences between the groups. CONCLUSION Hypomethylation of NR3C1 in placental tissue is associated with poorer neurological functioning at 3 months post-term in extremely preterm infants. Alleviating stress during pregnancy and its impact on preterm infants and their neurodevelopmental outcomes should be further investigated.
Collapse
Affiliation(s)
- Nienke H van Dokkum
- Department of Pediatrics, Division of Neonatology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Sofia Bachini
- Department of Pediatrics, Division of Neonatology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Rikst Nynke Verkaik-Schakel
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Dyvonne H Baptist
- Department of Pediatrics, Division of Neonatology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Sahar Salavati
- Department of Pediatrics, Division of Neonatology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Karianne E Kraft
- Department of Pediatrics, Division of Neonatology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Sicco A Scherjon
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Arend F Bos
- Department of Pediatrics, Division of Neonatology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Torsten Plösch
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
21
|
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: 1.5] [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
|
22
|
Prelipcean I, Wynn JL, Thompson L, Burchfield DJ, James-Woodley L, Chase PB, Barnes CP, Bernier A. Absence of relationship between serum cortisol and critical illness in premature infants. Arch Dis Child Fetal Neonatal Ed 2021; 106:408-412. [PMID: 33541918 PMCID: PMC8852370 DOI: 10.1136/archdischild-2020-319970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 12/05/2020] [Accepted: 12/10/2020] [Indexed: 11/04/2022]
Abstract
BACKGROUND Inadequate cortisol production in response to critical illness in extremely preterm infants may exacerbate poor outcomes. Despite commonly measuring cortisol concentration and administering hydrocortisone for presumed adrenal insufficiency, the relationship between serum cortisol concentration and illness severity remains unclear in this unique population. OBJECTIVE To determine the relationship between cortisol concentrations and illness severity as measured by the Score for Neonatal Acute Physiology II, neonatal Sequential Organ Failure Assessment and Vasoactive-Inotropic Score in premature infants. DESIGN/METHODS This retrospective, single-center cohort study included preterm infants born <30 weeks gestational age admitted to a level IV neonatal intensive care unit (NICU) between June 2011 and July 2018, who had a serum cortisol obtained for clinical indications before 36 weeks PMA. Demographic data were collected on infants and mothers. Nine clinical variables were identified a priori that could potentially modify cortisol concentration including critical illness. Univariate and multivariable analyses determined the relationship between cortisol concentration and each of these variables. RESULTS A total of 224 preterm infants with pretreatment serum cortisol concentration met criteria for inclusion. The median (IQR) gestational age at birth was 25 weeks (24, 26) and at cortisol measurement was 26 weeks (25, 28). The median cortisol was 13.3 ug/dL. Non-survivors had the highest values. Cortisol concentration did not correlate with any of the selected illness severity scores. CONCLUSIONS Cortisol concentrations in extremely preterm infants did not correlate with illness severity regardless of gestational age. Further studies are needed to identify clinically useful mediators of adrenal dysfunction and to guide clinical management.
Collapse
Affiliation(s)
- Irina Prelipcean
- Pediatrics, University of Rochester, Rochester, New York, USA .,Pediatrics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - James L. Wynn
- Department of Pediatrics, University of Florida, Gainesville, Florida,Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Lindsay Thompson
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | | | | | - Philip B. Chase
- Clinical and Translational Science - Informatics and Technology, University of Florida
| | - Christopher P. Barnes
- Clinical and Translational Science - Informatics and Technology, University of Florida
| | - Angelina Bernier
- Department of Pediatrics, University of Florida, Gainesville, Florida
| |
Collapse
|
23
|
Stoye DQ, Sullivan G, Galdi P, Kirschbaum C, Lamb GJ, Black GS, Evans MJ, Boardman JP, Reynolds RM. Perinatal determinants of neonatal hair glucocorticoid concentrations. Psychoneuroendocrinology 2021; 128:105223. [PMID: 33878601 PMCID: PMC8155393 DOI: 10.1016/j.psyneuen.2021.105223] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/21/2021] [Accepted: 04/05/2021] [Indexed: 12/15/2022]
Abstract
Adult hair glucocorticoid concentrations reflect months of hypothalamic-pituitary-adrenal axis activity. However, little is known about the determinants of neonatal hair glucocorticoids. We tested associations between perinatal exposures and neonatal hair glucocorticoids. Cortisol and cortisone were measured by LC-MS/MS in paired maternal and infant hair samples collected within 10 days of birth (n = 49 term, n = 47 preterm), with neonatal samples collected at 6-weeks in n = 54 preterm infants. We demonstrate cortisol accumulation in hair increases with fetal maturity, with hair cortisol being higher in term than preterm born infants after delivery (median 401 vs 106 pg/mg; p < 0.001). In term born infants, neonatal hair cortisol is positively associated with maternal hair cortisol concentration (β = 0.240, p = 0.045) and negatively associated with birthweight z-score (β = -0.340, p = 0.006). Additionally, being born without maternal labour is associated with lower hair cortisol concentrations (β = -0.489, p < 0.001) and a lower ratio of cortisol to cortisone (β = -0.484, p = 0.001). In preterm infants, histological chorioamnionitis is associated with a higher cortisol to cortisone ratio in hair (β = 0.459, p = 0.001). In samples collected 6 weeks after preterm birth, hair cortisol concentration is associated with cortisol hair concentrations measured after birth (β = 0.523, p < 0.001), chorioamnionitis (β = 0.250, p = 0.049) and postnatal exposures including intravenous hydrocortisone therapy (β = 0.343, p < 0.007) and neonatal sepsis (β = 0.290, p = 0.017). In summary, neonatal hair cortisol is associated with birth gestation, maternal hair cortisol concentration and fetal growth. Additionally, exposures at delivery are important determinants of hair cortisol, and should be considered in the design of future research investigating how neonatal hair cortisol relates to prenatal exposures or fetal development.
Collapse
Affiliation(s)
- David Q Stoye
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Gemma Sullivan
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Paola Galdi
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | | | - Gillian J Lamb
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Gill S Black
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Margaret J Evans
- Department of Pathology, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - James P Boardman
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Rebecca M Reynolds
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK; Centre for Cardiovascular Science, University of Edinburgh, UK.
| |
Collapse
|
24
|
Galbally M, Watson SJ, Lappas M, de Kloet ER, van Rossum E, Wyrwoll C, Mark P, Lewis AJ. Fetal programming pathway from maternal mental health to infant cortisol functioning: The role of placental 11β-HSD2 mRNA expression. Psychoneuroendocrinology 2021; 127:105197. [PMID: 33743501 DOI: 10.1016/j.psyneuen.2021.105197] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023]
Abstract
Placental 11β-HSD2 has been a focus of research for understanding potential fetal programming associated with maternal emotional disorders. This study examined the pathway from antenatal mental health via placental 11β-HSD2 mRNA to cortisol regulation in the infant offspring. This study reports on data obtained from 236 participants in the Mercy Pregnancy and Emotional Wellbeing Study (MPEWS). At term, placental tissue was collected within 30 min of birth from 52 participants meeting current criteria for a depressive disorder, and 184 control participants. Depressive disorders were diagnosed using the SCID-IV. In addition, antidepressant use, depressive and anxiety symptoms were measured in early and late pregnancy. Placental 11β-HSD2 mRNA expression was measured using qRT-PCR. Infant salivary cortisol samples were taken at 12 months of age. Women on antidepressant medication and with higher trait anxiety had higher placental 11β-HSD2 expression compared to women not taking medication. Furthermore, the offspring of women taking an antidepressant and who also had a current depressive disorder and high trait anxiety had high cortisol reactivity at 12 months of age and this was mediated through 11β-HSD2 mRNA expression. In contrast, offspring of women not taking antidepressant medication with depressive disorder and high anxiety there was low cortisol reactivity observed. Our findings suggest that the relationship between maternal antenatal depression and anxiety and infant cortisol reactivity is mediated through placental 11β-HSD2 mRNA expression. Furthermore, the direction differed for women taking antidepressants, where infant cortisol reactivity was high whereas when compared to those with unmedicated depression and anxiety, where infant cortisol reactivity was low.
Collapse
Affiliation(s)
- Megan Galbally
- Psychology, Murdoch University, Australia; School of Medicine, University of Notre Dame, Australia; King Edward Memorial Hospital, Subiaco, Australia.
| | - Stuart J Watson
- Psychology, Murdoch University, Australia; School of Medicine, University of Notre Dame, Australia
| | - Martha Lappas
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia; Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - E Ron de Kloet
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Elisabeth van Rossum
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Caitlin Wyrwoll
- School of Human Sciences, The University of Western Australia, Australia
| | - Peter Mark
- School of Human Sciences, The University of Western Australia, Australia
| | | |
Collapse
|
25
|
Marinelli KC, Lyden ER, Peeples ES. Clinical risk factors for the development of late-onset circulatory collapse in premature infants. Pediatr Res 2021; 89:968-973. [PMID: 32492694 DOI: 10.1038/s41390-020-0990-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Very low birth weight (VLBW) infants may be at risk for late-onset circulatory collapse (LCC) where otherwise stable infants develop hypotension resistant to vasoactive agents. The risk factors for LCC development are poorly defined, and it has been theorized that it may be in part due to withdrawal from exogenous prenatal steroids. The goal of this study was to define the clinical characteristics of LCC and investigate its association with antenatal steroid administration. METHODS This is a retrospective cohort study of infants born ≤1500 g. LCC was retrospectively diagnosed in infants requiring glucocorticoids for circulatory instability at >1 week of life. Demographic and clinical characteristics were compared between groups using Mann-Whitney test. RESULTS Three hundred and ten infants were included; 19 (6.1%) developed LCC. Infants with LCC were born at a median 4.6 weeks' lower gestation, 509 g lower birth weight than those without LCC. There was no difference in antenatal steroid delivery between the groups. CONCLUSIONS LCC occurs in a distinct subset of VLBW infants, suggesting the need for monitoring in this high-risk population. Antenatal steroids did not significantly increase the risk of LCC development in this study. IMPACT Late-onset circulatory collapse (LCC) is a life-threatening clinical entity occurring in around 6% in VLBW infants and is likely underdiagnosed in the United States. Targeting specific demographic characteristics such as birth weight (<1000 g) and gestational age at birth (<26 weeks) may allow for early identification of high-risk infants, allowing close monitoring and prompt treatment of LCC. No significant association was found between antenatal steroid administration and LCC development, suggesting that the theoretical risks of antenatal steroids on the fetal HPA axis does not outweigh the benefits of antenatal steroids in fetal lung maturity. To date, no studies characterizing LCC have originated outside of Asia. Therefore, providing a description of LCC in a U.S.-based cohort will provide insight into both its prevalence and presentation to inform clinicians about this potentially devastating disorder and foster early diagnosis and treatment. This study validates LCC characteristics and prevalence previously outlined by Asian studies in a single-center U.S.-based cohort while also identifying potential risk factors for LCC development. This manuscript will provide education for U.S. physicians about the risk factors and clinical presentation of LCC to facilitate early diagnosis and treatment, potentially decreasing neonatal mortality. With prompt recognition and treatment of LCC, infants may have decreased exposure to vasoactive medications that have significant systemic effects.
Collapse
Affiliation(s)
| | - Elizabeth R Lyden
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eric S Peeples
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
26
|
Davis EP, Narayan AJ. Pregnancy as a period of risk, adaptation, and resilience for mothers and infants. Dev Psychopathol 2020; 32:1625-1639. [PMID: 33427164 PMCID: PMC7863987 DOI: 10.1017/s0954579420001121] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The pregnancy period represents a unique window of opportunity to identify risks to both the fetus and mother and to deter the intergenerational transmission of adversity and mental health problems. Although the maternal-fetal dyad is especially vulnerable to the effects of stress during pregnancy, less is known about how the dyad is also receptive to salutary, resilience-promoting influences. The present review adopts life span and intergenerational perspectives to review four key areas of research. The first part describes how pregnancy is a sensitive period for both the mother and fetus. In the second part, the focus is on antecedents of maternal prenatal risks pertaining to prenatal stress response systems and mental health. The third part then turns to elucidating how these alterations in prenatal stress physiology and mental health problems may affect infant and child outcomes. The fourth part underscores how pregnancy is also a time of heightened fetal receptivity to maternal and environmental signals, with profound implications for adaptation. This section also reviews empirical evidence of promotive and protective factors that buffer the mother and fetus from developmental and adaptational problems and covers a sample of rigorous evidence-based prenatal interventions that prevent maladaptation in the maternal-fetal dyad before babies are born. Finally, recommendations elaborate on how to further strengthen understanding of pregnancy as a period of multilevel risk and resilience, enhance comprehensive prenatal screening, and expand on prenatal interventions to promote maternal-fetal adaptation before birth.
Collapse
Affiliation(s)
- Elysia Poggi Davis
- Department of Psychology, University of Denver, Denver, CO, USA
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | | |
Collapse
|
27
|
Nadholta P, Bali P, Singh A, Anand A. Potential benefits of Yoga in pregnancy-related complications during the COVID-19 pandemic and implications for working women. Work 2020; 67:269-279. [PMID: 33044208 DOI: 10.3233/wor-203277] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pregnancy is a vulnerable period of growth and enrichment along with many physiological and psychological challenges. These changes can lead to complications if compounded by external stress and anxiety. COVID-19 has emerged as a chief stressor among the general population and is a serious threat among vulnerable populations. Therefore, there is a need for stress management tools, such as Yoga and physical exercises, both at home and at work. These can be adopted during the pandemic with proper maintenance of social distancing. OBJECTIVE To evaluate and compile literature that has reported the health outcomes of Yoga intervention on pregnancy at the workplace and analyzes both the restrictions as well as advantages of its beneficial effects in comparison to physical exercises. METHODOLOGY A comprehensive literature review was conducted utilizing PubMed and Google Scholar. The keywords used for the search include "Yoga", "work", "complications", "physical exercise", "drugs" and "COVID" indifferent permutations and combinations with "pregnancy". We compiled the literature with respect to pregnancy complications and the effects of drugs, physical activity and Yoga for preventing these complications. RESULTS We noted that pregnancy-related complications are becoming more prevalent because of a sedentary lifestyle, restricted physical activity and growing stress. In such situations, a home or workplace Yoga protocol can combine both exercise and mindfulness-based alleviation of anxiety for both working and non-working women. CONCLUSION Yoga can be effective for combating stress and anxiety besides boosting immunity in pregnant working women confronted with the COVID-19 pandemic.
Collapse
Affiliation(s)
- Pooja Nadholta
- Neuroscience Research Lab, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Parul Bali
- Department of Biophysics, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Singh
- Swami Vivekananda Yoga Research Foundation (SVYASA), Bangalore, India
| | - Akshay Anand
- Neuroscience Research Lab, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
28
|
Zipori Y, Zidan R, Lauterbach R, Hagag A, Ginsberg Y, Solt I, Weiner Z, Kugelman A, Beloosesky R. Antenatal betamethasone and the risk of neonatal hypoglycemia: it's all about timing. Arch Gynecol Obstet 2020; 303:695-701. [PMID: 32960361 DOI: 10.1007/s00404-020-05785-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/31/2020] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Our objective was to evaluate whether there is a relationship between the "time during the day" of maternal betamethasone administration between 24 and 34 weeks' gestation and the risk for neonatal hypoglycemia. MATERIAL AND METHODS A retrospective study included cases between 2008 and 2018. Eligible cases were pregnant women with singleton pregnancies who received a single course of betamethasone between 24 and 34 weeks' gestation. Each woman was allocated into one of four pre-defined groups based on the time when intramuscular betamethasone was administered. Group 1 (23:00-04:59) represents the lowest daily natural corticosteroids' activity, group 2 (05:00-10:59) represents the peak daily natural corticosteroids' activity, whereas group 3 (11:00-16:59) and group 4 (17:00-22:59) present an intermediate natural state of steady corticosteroids' secretion and activity. The primary outcome of the study was the incidence of neonatal hypoglycemia (glucose level of less than 40 mg/dL). RESULTS We have identified 868 women who received a single complete course of betamethasone, of which 353 women (40.7%) had a steroid treatment latency to delivery up to 14 days. The incidence of neonatal hypoglycemia was significantly higher in group 2 (39.5%, 30/76, p = 0.0063), compared to group 1, who had the lowest incidence of neonatal hypoglycemia (16.9%, 12/71), and to group 3 and group 4. CONCLUSIONS The "time during the day" when betamethasone administered is important when considering the risk for neonatal hypoglycemia. The risk was significantly higher when betamethasone was administered during the peak time and significantly lower when administered at the nadir time of maternal endogenous corticosteroid activity.
Collapse
Affiliation(s)
- Yaniv Zipori
- Department of Obstetrics and Gynecology, Rambam Health Care Campus, 3109601, Haifa, Israel.
| | - Ragda Zidan
- Department of Obstetrics and Gynecology, Rambam Health Care Campus, 3109601, Haifa, Israel
| | - Roy Lauterbach
- Department of Obstetrics and Gynecology, Rambam Health Care Campus, 3109601, Haifa, Israel
| | - Arin Hagag
- Department of Obstetrics and Gynecology, Rambam Health Care Campus, 3109601, Haifa, Israel
| | - Yuval Ginsberg
- Department of Obstetrics and Gynecology, Rambam Health Care Campus, 3109601, Haifa, Israel
| | - Ido Solt
- Department of Obstetrics and Gynecology, Rambam Health Care Campus, 3109601, Haifa, Israel.,Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa, Israel
| | - Zeev Weiner
- Department of Obstetrics and Gynecology, Rambam Health Care Campus, 3109601, Haifa, Israel.,Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa, Israel
| | - Amir Kugelman
- Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa, Israel.,Department of Neonatology, Rambam Health Care Campus, Haifa, Israel
| | - Ron Beloosesky
- Department of Obstetrics and Gynecology, Rambam Health Care Campus, 3109601, Haifa, Israel.,Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa, Israel
| |
Collapse
|
29
|
di Pasquo E, Saccone G, Angeli L, Dall'Asta A, Borghi E, Fieni S, Berghella V, Magnani C, Frusca T, Ghi T. Determinants of neonatal hypoglycemia after antenatal administration of corticosteroids (ACS) for lung maturation: Data from two referral centers and review of the literature. Early Hum Dev 2020; 143:104984. [PMID: 32092675 DOI: 10.1016/j.earlhumdev.2020.104984] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/06/2020] [Accepted: 02/11/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND A correlation between ACS and neonatal hypoglycemia has been recently demonstrated. AIMS The aim of the study was to evaluate the determinants of neonatal hypoglycemia in women exposed to ACS for respiratory distress syndrome prevention. MATERIAL AND METHODS Retrospective, multicenter, cohort study conducted in two Tertiary University Units. All fetuses delivered from 2016 to 2017 after ACS (two doses i.m. of Betamethasone 12 mg 24 h apart) were considered eligible for the study purpose. The primary outcome was the incidence of hypoglycemia, defined as a glycemic value ≤45 mg/dl within the first 48 h of neonatal life. The effect on neonatal glycaemia due to timing (interval from exposure to delivery) and type (single completed, single partial or repeated course) of ACS administration was also assessed. RESULTS Overall, 99 neonates met the inclusion criteria. Hypoglycemia occurred in 38/99 (38.4%) of the included newborns. Compared to normoglycemic neonates, those with hypoglycemia had lower gestational age at delivery (33.06 ± 3.37 vs. 35.94 ± 3.17 g; p < 0.0001). Lower birthweight (1747.28 ± 815.29 vs. 2499.24 ± 780.51 g; p < 0.0001), a shorter interval time from administration to delivery (1.85 ± 2.59 vs. 3.34 ± 3.39 weeks; p = 0.02) and a higher incidence of single partial course (23.7 vs. 8.72%; p = 0.03). Multivariate logistic regression found that only birthweight was significantly associated with neonatal hypoglycemia (OR 0.4 95% CI -1.16/-0.04; p < 0.038). CONCLUSION Hypoglycemia occurs in a large proportion of fetuses exposed to ACS independently from the type of exposure (single partial/single completed) and from the time interval between ACS administration and delivery. Birthweight seems to be the strongest determinant for the occurrence neonatal hypoglycemia after antenatal administration of steroids for lung maturation.
Collapse
Affiliation(s)
- Elvira di Pasquo
- Obstetrics and Gynecology Unit, University of Parma, Parma, Italy
| | - Gabriele Saccone
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Laura Angeli
- Obstetrics and Gynecology Unit, University of Parma, Parma, Italy
| | - Andrea Dall'Asta
- Obstetrics and Gynecology Unit, University of Parma, Parma, Italy
| | - Elena Borghi
- Obstetrics and Gynecology Unit, University of Parma, Parma, Italy
| | - Stefania Fieni
- Obstetrics and Gynecology Unit, University of Parma, Parma, Italy
| | - Vincenzo Berghella
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA, USA
| | - Cinzia Magnani
- Pediatric Hospital 'P. Barilla', Neonatology Unit, University of Parma, Parma, Italy
| | - Tiziana Frusca
- Obstetrics and Gynecology Unit, University of Parma, Parma, Italy
| | - Tullio Ghi
- Obstetrics and Gynecology Unit, University of Parma, Parma, Italy.
| |
Collapse
|
30
|
Fassaie S, McAloon J. Maternal distress, HPA activity, and antenatal interventions: A systematic review. Psychoneuroendocrinology 2020; 112:104477. [PMID: 31753328 DOI: 10.1016/j.psyneuen.2019.104477] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 08/05/2019] [Accepted: 10/11/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Elevated antenatal distress has been associated with negative outcomes for both mothers and, as a result, their infants. One mechanism hypothesised to underlie these associations is the maternal hypothalamic pituitary adrenal (HPA) axis. Though research has examined whether biopsychosocial antenatal interventions can reduce maternal HPA activity, only one review has summarized the nature of findings to date. The present study examined randomised control trials (RCTs) specifically; our primary aim was to assess the effectiveness of antenatal interventions in reducing HPA activity in pregnant women, our secondary aim was to examine whether antenatal interventions reduced maternal self-report of depression and/or anxiety. METHODS This study systematically reviewed RCTs that evaluated biopsychosocial interventions that reported subjective and objective markers of maternal distress in pregnant women within the clinical population. RESULTS Eight studies met inclusion criteria and women were in their second or third trimester. HPA-activity was primarily assessed through salivary cortisol (n = 7) and self-reported maternal distress was assessed using a variety of validated screening measures. Included trials demonstrated significant methodological heterogeneity and small sample sizes, poor treatment adherence, and poor reliability in cortisol measurement indicated low methodological quality. CONCLUSIONS Due to the high heterogeneity across studies, small sample sizes, and unreliable sampling methods, firm conclusions about the efficacy and effectiveness of antenatal interventions cannot be drawn. Despite this, interventions which targeted pregnancy-specific influencers of maternal mood were more likely to result in reduced depression and anxiety symptomatology as reported by mothers.
Collapse
Affiliation(s)
- Soha Fassaie
- Graduate School of Health, University of Technology Sydney, PO Box 123, Broadway NSW 2007, Australia
| | - John McAloon
- Graduate School of Health, University of Technology Sydney, PO Box 123, Broadway NSW 2007, Australia.
| |
Collapse
|
31
|
Franks AL, Berry KJ, DeFranco DB. Prenatal drug exposure and neurodevelopmental programming of glucocorticoid signalling. J Neuroendocrinol 2020; 32:e12786. [PMID: 31469457 PMCID: PMC6982551 DOI: 10.1111/jne.12786] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/25/2019] [Accepted: 08/27/2019] [Indexed: 12/21/2022]
Abstract
Prenatal neurodevelopment is dependent on precise functioning of multiple signalling pathways in the brain, including those mobilised by glucocorticoids (GC) and endocannabinoids (eCBs). Prenatal exposure to drugs of abuse, including opioids, alcohol, cocaine and cannabis, has been shown to not only impact GC signalling, but also alter functioning of the hypothalamic-pituitary-adrenal (HPA) axis. Such exposures can have long-lasting neurobehavioural consequences, including alterations in the stress response in the offspring. Furthermore, cannabis contains cannabinoids that signal via the eCB pathway, which is linked to some components of GC signalling in the adult brain. Given that GCs are frequently used in pregnancy to prevent complications of prematurity, and also that rates of cannabis use in pregnancy are increasing, the likelihood of foetal co-exposure to these compounds is high and may have additional implications for long-term neurodevelopment. Here, we present a discussion of GC signalling and the HPA axis, as well as the effects of prenatal drug exposure on these pathways and the stress response, and we explore the interactions between GC and EC signalling in the developing brain and potential for neurodevelopmental consequences.
Collapse
Affiliation(s)
- Alexis L Franks
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kimberly J Berry
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Donald B DeFranco
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology and Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
32
|
Prenatal Glucocorticoid Treatment Completeness and Steroid Hormonal Levels as Related to Infant and Maternal Health. J Perinat Neonatal Nurs 2020; 34:E32-E43. [PMID: 33079812 DOI: 10.1097/jpn.0000000000000519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study was conducted to confirm the negative associations between testosterone and cortisol levels and health and developmental outcomes in very low-birth weight (VLBW) infants after controlling for prenatal glucocorticoid (GC) treatment. Seventy-four VLBW infant-mother pairs were recruited from a neonatal intensive care unit in the Southeastern United States. We divided the pairs into the complete (n = 58) and incomplete (n = 16) GC treatment groups. Data on infants and mothers were obtained at birth, 40 weeks' postmenstrual age, and 3 and 6 months' corrected age. Salivary testosterone and cortisol levels of the pairs were determined at 40 weeks' postmenstrual age using enzyme immunoassay. Log-linear and general linear mixed models showed that gestational age and birth weight were lower when testosterone was 1 pg/mL higher. When cortisol was 1 μg/dL higher, technology dependence at discharge was higher and motor development at 6 months was lower. Mothers with complete GC treatment had greater parity and gravida, more prenatal visits, and more medical complications. The study outcomes supported our hypothesis that steroid hormonal levels are more predictive of infant health and development than GC treatment completeness. Single dose of GC treatment might be just as effective as 2 doses, although further study with more subjects would be needed to confirm. As the associations with steroid hormonal levels lasted longer than the GC treatment associations, we recommend confirming the predictive effects of testosterone and cortisol levels after 6 months.
Collapse
|
33
|
Niwa F, Kawai M, Kanazawa H, Okanoya K, Myowa M. The development of the hypothalamus-pituitary-adrenal axis during infancy may be affected by antenatal glucocorticoid therapy. J Neonatal Perinatal Med 2020; 13:55-61. [PMID: 31609703 DOI: 10.3233/npm-180040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
BACKGROUND Developmental changes in the hypothalamus-pituitary-adrenal (HPA) axis during infancy have been reported in term infants, but those in preterm infants have yet to be elucidated. If developmental changes in the HPA axis of preterm infants are modulated by any factors, it may affect their future health. Few studies have examined the lasting consequences of antenatal glucocorticoids on the development of the HPA axis. METHODS We measured pre- and post-palivizumab vaccination salivary cortisol values in two conforming periods of three-months intervals during infancy, and compared cortisol values and the response of cortisol secretion between groups with and without antenatal glucocorticoid (AG) therapy. RESULTS Although the strength of the response of cortisol secretion to palivizumab fell age-dependently (until late infancy) in the Non-AG group, the opposite pattern was exhibited in the AG group. The changes of the delta cortisol values between the 2 groups were significant. CONCLUSIONS This study suggests that the HPA axis of preterm infants whose mothers receive AG therapy may be upregulated during infancy, possibly leading to long lasting health problems.
Collapse
Affiliation(s)
- F Niwa
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Japan
- Japan Science and Technology Agency, ERATO Okanoya Emotional Information Project, Japan
| | - M Kawai
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Japan
| | - H Kanazawa
- Graduate School of Information Science and Technology, The University of Tokyo, Japan
- Research Fellow of the Japan Society for the Promotion of Science, Japan
| | - K Okanoya
- Joint Research Laboratory for Emotional Information, Riken Brain Science Institute, and Department of Life Sciences, Graduate School of Arts and Sciences, the University of Tokyo, Japan
- Japan Science and Technology Agency, ERATO Okanoya Emotional Information Project, Japan
| | - M Myowa
- Japan Science and Technology Agency, ERATO Okanoya Emotional Information Project, Japan
- Graduate School of Education, Kyoto University, Japan
| |
Collapse
|
34
|
Bulygina VV, Kalinina TS, Lanshakov DA, Dygalo NN. Expression of Neurotrophic Factor 3 in the Hippocampus of Neonatal Rats after Administration of Dexamethasone. NEUROCHEM J+ 2019. [DOI: 10.1134/s181971241903005x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
35
|
Iijima S. Late-onset glucocorticoid-responsive circulatory collapse in premature infants. Pediatr Neonatol 2019; 60:603-610. [PMID: 31564521 DOI: 10.1016/j.pedneo.2019.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 07/16/2019] [Accepted: 09/06/2019] [Indexed: 12/20/2022] Open
Abstract
Late-onset glucocorticoid-responsive circulatory collapse (LGCC) in infants is characterized by sudden onset of hypotension and/or oliguria, which is resistant to volume expanders and inotropes but responds rapidly to intravenous glucocorticoids. LGCC occurs after the first week of life mainly in relatively stable very low birth weight (VLBW) infants. In Japan, the incidence of LGCC is reported to be 8%. Relative adrenal insufficiency (AI) is considered the most likely cause of LGCC, but its detailed pathophysiology remains unclear. Intrinsic and extrinsic factors may affect the pathophysiological mechanism. LGCC should be recognized as one of the high-risk complications in VLBW infants and managed promptly and properly, because if it is not, it may cause life-long neurological problems. To diagnose relative AI, an accurate evaluation of adrenal function is necessary; however, the interpretation of basal serum cortisol levels is difficult in preterm infants after 7 days of life. To recognize LGCC, it is recommended that blood pressure and urine volume be carefully monitored, even outside of the transitional period. If no underlying causes are documented or volume expansion and inotropic support fail, intravenous hydrocortisone should be initiated, and an additional dose of hydrocortisone is required when the response is inadequate. There are few reports to verify or characterize LGCC and this phenomenon has not been recognized worldwide to date. This review summarizes the current knowledge about LGCC in premature infants and evaluates the most significant new findings regarding its pathophysiology, treatment, and prognosis.
Collapse
Affiliation(s)
- Shigeo Iijima
- Department of Pediatrics, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka 431-3192, Japan.
| |
Collapse
|
36
|
Optimizing antenatal corticosteroid therapy for improving outcome of premature infants. Pediatr Res 2019; 86:556-557. [PMID: 31412354 DOI: 10.1038/s41390-019-0538-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 08/05/2019] [Indexed: 11/08/2022]
|
37
|
Plasma cortisol and ACTH levels in 416 VLBW preterm infants during the first month of life: distribution in the AGA/SGA population. J Perinatol 2019; 39:934-940. [PMID: 31068663 DOI: 10.1038/s41372-019-0381-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 03/26/2019] [Accepted: 04/01/2019] [Indexed: 11/08/2022]
Abstract
OBJECTIVE The aim of this study is to establish the serum level distribution of cortisol and ACTH in VLBW preterm newborns and determine which neonates are ideal candidates for the stimulation test for adrenal insufficiency. METHODS Plasma cortisol and ACTH levels were evaluated in 416 VLBW newborns on days 1, 7, and 30 of life. Gender, gestational age, weight, type of delivery, RDS prophylaxis, and perinatal morbidities were considered as potential variability factors. RESULTS Cortisol and ACTH levels significantly decreased between 1, 7, and 30 days. Significantly higher cortisol levels were found at lower gestational ages and in infants born by vaginal delivery, whereas lower levels were observed in those born after maternal corticosteroid treatment. The distribution of cortisol and ACTH levels in healthy infants born by cesarian section is presented. CONCLUSION Even if high or low levels were not frequently linked to illness, the presented distribution data may indicate that the newborns are ideal candidates for the stimulation test.
Collapse
|
38
|
Lv J, Ma Q, Dasgupta C, Xu Z, Zhang L. Antenatal Hypoxia and Programming of Glucocorticoid Receptor Expression in the Adult Rat Heart. Front Physiol 2019; 10:323. [PMID: 31001129 PMCID: PMC6454194 DOI: 10.3389/fphys.2019.00323] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/11/2019] [Indexed: 12/26/2022] Open
Abstract
Glucocorticoid receptor (GR) signaling is critical for development and function of the heart. Our previous study demonstrated that gestational hypoxia induced epigenetic repression of the GR gene in the developing heart. The present study aims to determine that the alterations of promoter methylation level and epigenetic repression of the GR gene in the developing heart in response to maternal hypoxia is sustained in adult offspring and potential gender differences in the programming of GR gene. Pregnant rats were treated with 10.5% O2 from gestational day 15 (E15) to 21 (E21). Hearts were isolated from 5-month-old male and female offspring with the developing stage being equivalent to 18-year-old human. GR mRNA and protein abundance was determined with real time qRT-PCR and Western blot. GR gene promoter methylation and binding of transcription factors were measured with methylated DNA immunoprecipitation (MeDIP) and Chromatin immunoprecipitation (ChIP). The results showed that antenatal hypoxia significantly decreased the expression of GR mRNA and protein in the hearts of adult male offspring, but not in females, which is ascribed to the differential changes of alternative exon1 mRNA variants of GR gene in male and female hearts in response to prenatal hypoxia. In addition, the downregulation of GR expression in the male heart was correlated with increased methylation levels of CpG dinucleotides in promoters of exon 14, 15, 16, 17, and 110, which resulted in a decrease in the binding of their transcription factors. Thus, the study reveals that antenatal hypoxia results in a reprogramming and long-term change in GR gene expression in the heart by hypermethylation of GR promoter in a sex-differential pattern, which provides a novel mechanism regarding the increased vulnerability of heart later in life with exposure of prenatal hypoxia.
Collapse
Affiliation(s)
- Juanxiu Lv
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qingyi Ma
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Chiranjib Dasgupta
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Zhice Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Lubo Zhang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| |
Collapse
|
39
|
Developmental programming of shyness: A longitudinal, prospective study across four decades. Dev Psychopathol 2019; 32:455-464. [DOI: 10.1017/s0954579419000208] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
AbstractAlthough shyness is a ubiquitous phenomenon with early developmental origins, little research has examined the influence of prenatal exposures on the developmental trajectory of shyness. Here, we examined trajectories of shyness from childhood to adulthood in three groups (N = 254), with varying degrees of prenatal adversity as indicated by the number of stressful exposures: extremely low birth weight (ELBW; <1000 g) survivors prenatally exposed to exogenous corticosteroids (ELBW+S, n = 56); ELBW survivors not prenatally exposed to exogenous corticosteroids (ELBW+NS, n = 56); and normal birth weight (NBW, n = 142) controls. Multilevel modeling revealed that the ELBW+S individuals exhibited the highest levels of childhood shyness, which remained stable into adulthood. The ELBW+NS and NBW controls had comparably low levels of childhood shyness; however, the ELBW+NS individuals experienced patterns of increasing shyness, while NBW controls displayed decreases in shyness into adulthood. We speculate that individuals exposed to multiple prenatal stressors (i.e., ELBW+S) may be developmentally programmed to be more sensitive to detecting social threat, with one manifestation being early developing, stable shyness, while increasing shyness among ELBW+NS individuals may reflect a later developing shyness influenced by postnatal context. We discuss the implications of these findings for understanding the developmental origins and developmental course of human shyness from childhood through adulthood.
Collapse
|
40
|
Savoy C, Mathewson KJ, Schmidt LA, Morrison KM, Saigal S, Boyle MH, Van Lieshout RJ. Exposure to antenatal corticosteroids and reduced respiratory sinus arrhythmia in adult survivors of extremely low birth weight. Int J Neurosci 2019; 129:776-783. [PMID: 30633628 DOI: 10.1080/00207454.2019.1567511] [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: 12/21/2022]
Abstract
Purpose/aim: Antenatal corticosteroid (ACS) therapy has dramatically increased survival rates among extremely low birth weight (ELBW) infants. However, the long-term effects of ACS on autonomic nervous system function have not been explored. Using the world's oldest longitudinally followed cohort of ELBW infants we compared respiratory sinus arrhythmia (RSA) among ELBW survivors whose mothers received ACS (ELBW-S), those who did not (ELBW-NS) and normal birth weight (NBW) controls in their 20 and 30 s. Methods: Resting electrocardiogram (ECG) was recorded from ELBW-S (n = 28), ELBW-NS (n = 36), and matched NBW controls (n = 79) at 22-26 and 29-36 years. Resting RSA was compared across groups via analyses of covariance (ANCOVA), adjusting for sex, medication use, postnatal steroid exposure and the presence of chronic health conditions. RSA was also compared across assessments for each group. Results: At 29-36 years, resting RSA in ELBW-S was significantly lower than in NBW controls. RSA in the ELBW-NS group was intermediate between ELBW-S and NBW groups. Although the ELBW-S group also showed nominally reduced RSA compared to NBW controls at the 22-26-year visit, this difference was not statistically significant. Conclusions: ELBW survivors exposed to ACS had lower RSA than NBW controls during their 30 s, suggestive of a decline in parasympathetic input to heart. ELBW survivors who received ACS may be particularly vulnerable to cardiovascular problems in later life.
Collapse
Affiliation(s)
- Calan Savoy
- a Department of Psychiatry and Behavioural Neurosciences , McMaster University , Hamilton , Canada
| | - Karen J Mathewson
- a Department of Psychiatry and Behavioural Neurosciences , McMaster University , Hamilton , Canada
| | - Louis A Schmidt
- b Department of Psychology, Neuroscience and Behaviour , McMaster University , Hamilton , Canada
| | | | - Saroj Saigal
- c Department of Pediatrics , McMaster University , Hamilton , Canada
| | - Michael H Boyle
- a Department of Psychiatry and Behavioural Neurosciences , McMaster University , Hamilton , Canada
| | - Ryan J Van Lieshout
- a Department of Psychiatry and Behavioural Neurosciences , McMaster University , Hamilton , Canada
| |
Collapse
|
41
|
Vora NL, Parker JS, Mieckowski PA, Smeester L, Fry RC, Boggess KA. RNA-Sequencing of Umbilical Cord Blood to Investigate Spontaneous Preterm Birth: A Pilot Study. AJP Rep 2019; 9:e60-e66. [PMID: 30854245 PMCID: PMC6406026 DOI: 10.1055/s-0039-1678717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 11/13/2018] [Indexed: 12/21/2022] Open
Abstract
Objective To analyze the transcriptomic gene expression of umbilical cord blood leukocytes using RNA-sequencing from preterm birth (PTB) and term birth (TB). Study Design Eight women with spontaneous PTB (sPTB) and eight women with unlabored TB were enrolled prospectively. The sPTB and TB cohorts were matched for maternal age, race, mode of delivery, and fetal sex. Cord blood RNA was extracted and a globin depletion protocol was applied, then sequenced on the Illumina HiSeq 4000. Raw read counts were analyzed with DESeq2 to test for gene expression differences between sPTB and TB. Results 148 genes had significant differential expression ( q < 0.01). Cell cycle/metabolism gene expression was significantly higher and immune/inflammatory signaling gene expression significantly lower in the sPTB cohort compared with term. In African American (AA) infants, 18 genes specific to cell signaling, neutrophil activity, and major histocompatibility complex type 1 had lower expression in preterm compared with term cohort; the opposite pattern was seen in non-Hispanic Whites (NHWs). Conclusion Compared with term, preterm fetuses have higher cell cycle/metabolism gene expression, suggesting metabolic focus on growth and development. Immune function gene expression in this pilot study is lower in the sPTB group compared with term and differs in AA compared with NHW infants.
Collapse
Affiliation(s)
- Neeta L Vora
- Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Joel S Parker
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Piotr A Mieckowski
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Lisa Smeester
- Department of Environmental Sciences and Engineering, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina
| | - Rebecca C Fry
- Department of Environmental Sciences and Engineering, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina
| | - Kim A Boggess
- Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
42
|
Lemche E. Early Life Stress and Epigenetics in Late-onset Alzheimer's Dementia: A Systematic Review. Curr Genomics 2018; 19:522-602. [PMID: 30386171 PMCID: PMC6194433 DOI: 10.2174/1389202919666171229145156] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 07/27/2017] [Accepted: 12/12/2017] [Indexed: 11/22/2022] Open
Abstract
Involvement of life stress in Late-Onset Alzheimer's Disease (LOAD) has been evinced in longitudinal cohort epidemiological studies, and endocrinologic evidence suggests involvements of catecholamine and corticosteroid systems in LOAD. Early Life Stress (ELS) rodent models have successfully demonstrated sequelae of maternal separation resulting in LOAD-analogous pathology, thereby supporting a role of insulin receptor signalling pertaining to GSK-3beta facilitated tau hyper-phosphorylation and amyloidogenic processing. Discussed are relevant ELS studies, and findings from three mitogen-activated protein kinase pathways (JNK/SAPK pathway, ERK pathway, p38/MAPK pathway) relevant for mediating environmental stresses. Further considered were the roles of autophagy impairment, neuroinflammation, and brain insulin resistance. For the meta-analytic evaluation, 224 candidate gene loci were extracted from reviews of animal studies of LOAD pathophysiological mechanisms, of which 60 had no positive results in human LOAD association studies. These loci were combined with 89 gene loci confirmed as LOAD risk genes in previous GWAS and WES. Of the 313 risk gene loci evaluated, there were 35 human reports on epigenomic modifications in terms of methylation or histone acetylation. 64 microRNA gene regulation mechanisms were published for the compiled loci. Genomic association studies support close relations of both noradrenergic and glucocorticoid systems with LOAD. For HPA involvement, a CRHR1 haplotype with MAPT was described, but further association of only HSD11B1 with LOAD found; however, association of FKBP1 and NC3R1 polymorphisms was documented in support of stress influence to LOAD. In the brain insulin system, IGF2R, INSR, INSRR, and plasticity regulator ARC, were associated with LOAD. Pertaining to compromised myelin stability in LOAD, relevant associations were found for BIN1, RELN, SORL1, SORCS1, CNP, MAG, and MOG. Regarding epigenetic modifications, both methylation variability and de-acetylation were reported for LOAD. The majority of up-to-date epigenomic findings include reported modifications in the well-known LOAD core pathology loci MAPT, BACE1, APP (with FOS, EGR1), PSEN1, PSEN2, and highlight a central role of BDNF. Pertaining to ELS, relevant loci are FKBP5, EGR1, GSK3B; critical roles of inflammation are indicated by CRP, TNFA, NFKB1 modifications; for cholesterol biosynthesis, DHCR24; for myelin stability BIN1, SORL1, CNP; pertaining to (epi)genetic mechanisms, hTERT, MBD2, DNMT1, MTHFR2. Findings on gene regulation were accumulated for BACE1, MAPK signalling, TLR4, BDNF, insulin signalling, with most reports for miR-132 and miR-27. Unclear in epigenomic studies remains the role of noradrenergic signalling, previously demonstrated by neuropathological findings of childhood nucleus caeruleus degeneration for LOAD tauopathy.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| |
Collapse
|
43
|
Horn SR, Roos LE, Berkman ET, Fisher PA. Neuroendocrine and immune pathways from pre- and perinatal stress to substance abuse. Neurobiol Stress 2018; 9:140-150. [PMID: 30450380 PMCID: PMC6236513 DOI: 10.1016/j.ynstr.2018.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/06/2018] [Accepted: 09/12/2018] [Indexed: 12/26/2022] Open
Abstract
Early life adversity is a documented risk factor for substance abuse and addiction. The pre- and perinatal period (i.e., from implantation, through pregnancy, to 6 months of age) is a critical period marked by high biological plasticity and vulnerability, making perinatal stress a particularly robust form of adversity. The neuroendocrine and immune systems are key mechanisms implicated in the transmission of addiction risk. We review animal and human studies that provide preliminary evidence for links between perinatal stress, neuroendocrine and immune dysregulation, and risk for substance abuse and addiction. A translational neuroscience perspective is employed to elucidate pre- and perinatally-induced biological mechanisms linked to addiction and discuss implications for prevention and intervention efforts. Significant evidence supports associations between pre- and perinatal stress and dysregulation of the hypothalamic-pituitary-adrenal axis and immune systems as well as links between neuroendocrine/immune functioning and addiction risk. More work is needed to explicitly examine the interplay between pre- and perinatal stress and neuroendocrine/immune disruptions that together heighten substance abuse risk. Future work is needed to fully understand how pre- and perinatal stress induces biological alterations to predispose individuals to higher risk for addiction. Such knowledge will strengthen theoretically-driven and empirically-supported prevention efforts for substance abuse and addiction.
Collapse
Affiliation(s)
- Sarah R Horn
- University of Oregon, Department of Psychology, 1227 University of Oregon, Eugene, OR, 97402, USA
| | - Leslie E Roos
- University of Oregon, Department of Psychology, 1227 University of Oregon, Eugene, OR, 97402, USA
| | - Elliot T Berkman
- University of Oregon, Department of Psychology, 1227 University of Oregon, Eugene, OR, 97402, USA
| | - Philip A Fisher
- University of Oregon, Department of Psychology, 1227 University of Oregon, Eugene, OR, 97402, USA
| |
Collapse
|
44
|
Agnew EJ, Ivy JR, Stock SJ, Chapman KE. Glucocorticoids, antenatal corticosteroid therapy and fetal heart maturation. J Mol Endocrinol 2018; 61:R61-R73. [PMID: 29720513 PMCID: PMC5976079 DOI: 10.1530/jme-18-0077] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 05/02/2018] [Indexed: 01/08/2023]
Abstract
Glucocorticoids are essential in mammals to mature fetal organs and tissues in order to survive after birth. Hence, antenatal glucocorticoid treatment (termed antenatal corticosteroid therapy) can be life-saving in preterm babies and is commonly used in women at risk of preterm birth. While the effects of glucocorticoids on lung maturation have been well described, the effects on the fetal heart remain less clear. Experiments in mice have shown that endogenous glucocorticoid action is required to mature the fetal heart. However, whether the potent synthetic glucocorticoids used in antenatal corticosteroid therapy have similar maturational effects on the fetal heart is less clear. Moreover, antenatal corticosteroid therapy may increase the risk of cardiovascular disease in adulthood. Here, we present a narrative review of the evidence relating to the effects of antenatal glucocorticoid action on the fetal heart and discuss the implications for antenatal corticosteroid therapy.
Collapse
Affiliation(s)
- Emma J Agnew
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - Jessica R Ivy
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - Sarah J Stock
- MRC Centre for Reproductive HealthUniversity of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - Karen E Chapman
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
- Correspondence should be addressed to K E Chapman:
| |
Collapse
|
45
|
Frontal EEG asymmetry in extremely low birth weight adult survivors: Links to antenatal corticosteroid exposure and psychopathology. Clin Neurophysiol 2018; 129:1891-1898. [PMID: 30005216 DOI: 10.1016/j.clinph.2018.05.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 04/11/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Extremely low birth weight (ELBW; <1000 g) survivors are exposed to significant perinatal adversity. In many cases, mothers of these infants receive antenatal corticosteroids (ACS), which reduce offspring neonatal mortality but may have lasting neuropsychiatric effects. However, the long-term neurophysiological effects of being born at ELBW and exposed to ACS are unknown. METHODS We compared resting frontal electroencephalogram (EEG) alpha asymmetry in ELBW survivors (n = 51), some of whom were exposed to antenatal corticosteroids (ELBW-S; n = 23) versus non-exposed (ELBW-NS; n = 28), and normal birth weight controls (NBW; n = 66) in adulthood (mage = 32.3 years). RESULTS ELBW survivors exhibited greater relative right frontal EEG alpha (11.5 to 13.5 Hz) asymmetry at rest relative to NBW controls. A linear relation was observed between increased exposure to perinatal adversity (NBW vs. ELBW-NS vs. ELBW-S) and greater relative right frontal asymmetry. Relative right frontal asymmetry was also linked to contemporaneous psychopathology in all individuals. CONCLUSION In the fourth decade of life, exposure to perinatal adversity was associated with patterns of frontal brain activity that reflect risk for psychopathology. Exposure to additional physiological stresses such as antenatal corticosteroids may amplify these effects. SIGNIFICANCE The mental health of ELBW survivors, particularly those exposed to antenatal corticosteroids, should be monitored into adulthood.
Collapse
|
46
|
Epidemiology and Effects of Substance Use in Pregnancy. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2018; 39:906-915. [PMID: 28935056 DOI: 10.1016/j.jogc.2017.07.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/16/2017] [Accepted: 07/16/2017] [Indexed: 12/13/2022]
Abstract
Substance use during pregnancy has important implications for health care providers and policymakers and can negatively affect a woman's health and the health of her children. Understanding trends, patterns of use, and outcomes are critical to developing prevention campaigns, building awareness, and providing effective care. This review critically examines the current literature on substance use in pregnancy and during the postpartum period in terms of epidemiology, risk factors, and implications. The risk factors for substance use in pregnancy, the challenges associated with reporting these cases, and the adverse effects of common substances on maternal and fetal health are discussed.
Collapse
|
47
|
Abstract
The soon-to-be-delivered fetus and preterm infant have been treated with glucocorticoids to prepare for postnatal life, historically for more than 40 years. The use of glucocorticoids is as much for replacement of cortisol in the setting of a poorly functioning hypothalamic-pituitary-adrenal axis in the preterm infant, as it is for prevention of long-term lung dysfunction. Potential negative effects of glucocorticoid treatment on brain development and function have been observed more often with dexamethasone therapy than with use of other glucocorticoids. Overall, glucocorticoid treatment has improved the outcome of the preterm infant.
Collapse
Affiliation(s)
- Susan M Scott
- University of New Mexico School of Medicine, 2211 Lomas Boulevard, Albuquerque, NM 87131, USA.
| | - Susan R Rose
- Cincinnati Children's Hospital Medical Center and University of Cincinnati, MLC 7012, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| |
Collapse
|
48
|
Travers S, Martinerie L, Boileau P, Lombès M, Pussard E. Alterations of adrenal steroidomic profiles in preterm infants at birth. Arch Dis Child Fetal Neonatal Ed 2018; 103:F143-F151. [PMID: 28747362 DOI: 10.1136/archdischild-2016-312457] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 05/22/2017] [Accepted: 05/22/2017] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Preterm infants have relative adrenal and kidney immaturity. Recently, we linked their urine sodium loss to a hypoaldosteronism at variance with an appropriate stimulation of the renin-angiotensin system. To investigate this defective aldosterone secretion, we analyse the biosynthesis pathways of adrenal steroids in neonates according to gestational age (GA). DESIGN Multicentre study (Premaldo) including 152 neonates classified into three groups: group 1 (very preterm (VPT)): <33 gestational weeks (GW); group 2 (preterm (PT)): 33-36 GW and group 3 (term (T)): ≥GW. METHOD Steroidomic profiles of mineralocorticoids, glucocorticoids and adrenal androgens were established from umbilical cord at birth (n=152) and peripheral blood at day 3 (n=70) using a recently developed liquid chromatography mass spectrometry method (LC-MS/MS). The enzymatic activity of each biosynthesis step was estimated by the product-to-substrate ratio. RESULTS At birth, VPT infants exhibit a global defect in adrenal steroid synthesis pathways leading to lower levels of aldosterone, cortisol and androstenedione than in term infants. This defect was strongly related to GA. On day 3, steroid precursors (progesterone, 11-deoxycorticosterone (DOC), 17-hydroxyprogesterone(17-OH-P) and 11-deoxycortisol (S)) were higher in VPT and negatively correlated with GA. Despite of precursors' accumulation, aldosterone and cortisol were similar in the three groups. At birth and day 3, a low cortisol/11-deoxycortisol ratio was found in preterm infants, suggesting an 11-beta-hydroxylase activity (CYP11B1) deficiency. CONCLUSIONS At birth, VPT infants exhibit a global deficit in mineralocorticoids, glucocorticoids and adrenal androgens that attenuates on day 3 of life. Steroid profiling using LC-MS/MS provides evidence for a partial defect in 11-hydroxylase along with prematurity.
Collapse
Affiliation(s)
- Simon Travers
- Inserm, U1185, Le Kremlin-Bicêtre, France.,Fac Med Paris-Sud, Univ, Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France.,Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpital de Bicêtre, Hôpitaux Universitaires Paris Sud, Assistance Publique'Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Laetitia Martinerie
- Inserm, U1185, Le Kremlin-Bicêtre, France.,Service d'Endocrinologie Pédiatrique, Hôpital Robert Debré, Assistance Publique Hôpitaux de Paris, Paris, France.,PremUp Foundation, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Pascal Boileau
- PremUp Foundation, Paris, France.,Service de Réanimation Néonatale, CH Poissy St'Germain en'Laye, Poissy, France.,EA 7285, UFR des Sciences de la Santé, Simone Veil, Université Versailles St-Quentin en Yvelines, Montigny le Bretonneux, France
| | - Marc Lombès
- Inserm, U1185, Le Kremlin-Bicêtre, France.,Fac Med Paris-Sud, Univ, Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France.,PremUp Foundation, Paris, France.,Service d'Endocrinologie et Maladies de la Reproduction, Hôpital de Bicêtre, Hôpitaux Universitaires Paris Sud, Assistance Publique'Hêpitaux de Paris, Le Kremlin Bicêtre, France.,Institut Biomédical de Bicêtre, Le Kremlin-Bicêtre, France
| | - Eric Pussard
- Inserm, U1185, Le Kremlin-Bicêtre, France.,Fac Med Paris-Sud, Univ, Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France.,Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpital de Bicêtre, Hôpitaux Universitaires Paris Sud, Assistance Publique'Hôpitaux de Paris, Le Kremlin Bicêtre, France.,Institut Biomédical de Bicêtre, Le Kremlin-Bicêtre, France
| |
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: 121] [Impact Index Per Article: 17.3] [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
|
Chen Q, Huang R, Hua L, Guo Y, Huang L, Zhao Y, Wang X, Zhang J. Prenatal exposure to perfluoroalkyl and polyfluoroalkyl substances and childhood atopic dermatitis: a prospective birth cohort study. Environ Health 2018; 17:8. [PMID: 29343261 PMCID: PMC5773146 DOI: 10.1186/s12940-018-0352-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 01/05/2018] [Indexed: 05/21/2023]
Abstract
BACKGROUND Perfluoroalkyl and polyfluoroalkyl substances (PFASs) have been reported to suppress immune function. However, previous studies on prenatal exposure to PFASs and allergic disorders in offspring provided inconsistent results. We aimed to examine the association between prenatal exposure to PFASs and childhood atopic dermatitis (AD) in offspring up to 24 months of age. METHODS A prospective birth cohort study involving 1056 pregnant women was conducted in two hospitals in Shanghai from 2012 to 2015. Prenatal information was collected by an interview with the women and from medical records. Fetal umbilical cord blood was collected at birth. Cord blood plasma PFASs were measured. Children were followed at 6, 12 and 24 months and information on the development of AD was recorded. AD was diagnosed by 2 dermatologists independently based on the questionnaires. Multiple logistic regression was used to compute odds ratio (OR) and corresponding 95% confidence interval (CI) for the association between AD and each PFASs, adjusting for potential confounders. RESULTS A total of 687 children completed a 2-year follow-up visit and had PFASs measurement. AD was diagnosed in 173 (25.2%) children during the first 24 months. In female children, a log-unit increase in perfluorooctanoic acid (PFOA) was associated with a 2.1-fold increase in AD risk (AOR 2.07, 95% CI 1.13-3.80) after adjusting for potential confounders. The corresponding risk was 2.22 (1.07-4.58) for perfluorononanoic acid (PFNA). The highest PFOA quartile was significantly associated with AD (2.52, 1.12-5.68) compared with the lowest quartile. The highest quartile of PFNA, perfluorodecanoic acid (PFDA) and perfluorohexane sulfonic acid (PFHxS) were associated with AD with AOR (95% CI) being 2.14 (0.97-4.74), 2.14 (1.00-4.57), and 2.30 (1.03-5.15), respectively. Additionally, the second quartile of perfluorododecanoic acid (PFDoA) was associated with a 3.2-fold increase in AD risk (3.24, 1.44-7.27). However, no significant associations were found in male children. CONCLUSIONS Prenatal exposure to PFOA, PFDA, PFDoA and PFHxS significantly increased the risk of childhood AD in female children during the first 24 months of life. In addition, the associations between AD with prenatal exposure to PFNA were close to statistical significance.
Collapse
Affiliation(s)
- Qian Chen
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092 China
| | - Rong Huang
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092 China
| | - Li Hua
- Department of Pediatric Pulmonology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
| | - Yifeng Guo
- Department of Dermatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
| | - Lisu Huang
- Department of Pediatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
- Clinical research unit, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
| | - Yanjun Zhao
- Department of Child Health Care, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200040 China
| | - Xia Wang
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092 China
| | - Jun Zhang
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092 China
| |
Collapse
|