1
|
Collins JM, Keane JM, Deady C, Khashan AS, McCarthy FP, O'Keeffe GW, Clarke G, Cryan JF, Caputi V, O'Mahony SM. Prenatal stress impacts foetal neurodevelopment: Temporal windows of gestational vulnerability. Neurosci Biobehav Rev 2024; 164:105793. [PMID: 38971516 DOI: 10.1016/j.neubiorev.2024.105793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Prenatal maternal stressors ranging in severity from everyday occurrences/hassles to the experience of traumatic events negatively impact neurodevelopment, increasing the risk for the onset of psychopathology in the offspring. Notably, the timing of prenatal stress exposure plays a critical role in determining the nature and severity of subsequent neurodevelopmental outcomes. In this review, we evaluate the empirical evidence regarding temporal windows of heightened vulnerability to prenatal stress with respect to motor, cognitive, language, and behavioural development in both human and animal studies. We also explore potential temporal windows whereby several mechanisms may mediate prenatal stress-induced neurodevelopmental effects, namely, excessive hypothalamic-pituitary-adrenal axis activity, altered serotonin signalling and sympathetic-adrenal-medullary system, changes in placental function, immune system dysregulation, and alterations of the gut microbiota. While broadly defined developmental windows are apparent for specific psychopathological outcomes, inconsistencies arise when more complex cognitive and behavioural outcomes are considered. Novel approaches to track molecular markers reflective of the underlying aetiologies throughout gestation to identify tractable biomolecular signatures corresponding to critical vulnerability periods are urgently required.
Collapse
Affiliation(s)
- James M Collins
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - James M Keane
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Clara Deady
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Ali S Khashan
- School of Public Health, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Fergus P McCarthy
- The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Obstetrics and Gynaecology, University College Cork, Cork, Ireland.
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland.
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Valentina Caputi
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | | |
Collapse
|
2
|
Martz J, Shelton MA, Geist L, Seney ML, Kentner AC. Sex differences in offspring risk and resilience following 11β-hydroxylase antagonism in a rodent model of maternal immune activation. Neuropsychopharmacology 2024; 49:1078-1090. [PMID: 38007547 PMCID: PMC11109257 DOI: 10.1038/s41386-023-01771-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/21/2023] [Accepted: 11/08/2023] [Indexed: 11/27/2023]
Abstract
Maternal immune activation (MIA) puts offspring at greater risk for neurodevelopmental disorders associated with impaired social behavior. While it is known that immune signaling through maternal, placental, and fetal compartments contributes to these phenotypical changes, it is unknown to what extent the stress response to illness is involved and how it can be harnessed for potential interventions. To this end, on gestational day 15, pregnant rat dams were administered the bacterial mimetic lipopolysaccharide (LPS; to induce MIA) alongside metyrapone, a clinically available 11β-hydroxylase (11βHSD) inhibitor used to treat hypercortisolism in pregnant, lactating, and neonatal populations. Maternal, placental, and fetal brain levels of corticosterone and placental 11βHSD enzymes type 1 and 2 were measured 3-hrs post treatment. Offspring social behaviors were evaluated across critical phases of development. MIA was associated with increased maternal, placental, and fetal brain corticosterone concentrations that were diminished with metyrapone exposure. Metyrapone protected against reductions in placental 11βHSD2 in males only, suggesting that less corticosterone was inactivated in female placentas. Behaviorally, metyrapone-exposure attenuated MIA-induced social disruptions in juvenile, adolescent, and adult males, while females were unaffected or performed worse. Metyrapone-exposure reversed MIA-induced transcriptional changes in monoamine-, glutamate-, and GABA-related genes in adult male ventral hippocampus, but not in females. Taken together, these findings illustrate that MIA-induced HPA responses act alongside the immune system to produce behavioral deficits. As a clinically available drug, the sex-specific benefits and constraints of metyrapone should be investigated further as a potential means of reducing neurodevelopmental risks due to gestational MIA.
Collapse
Affiliation(s)
- Julia Martz
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, 02115, USA
| | - Micah A Shelton
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Laurel Geist
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, 02115, USA
| | - Marianne L Seney
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, 02115, USA.
| |
Collapse
|
3
|
Sze Y, Brunton PJ. How is prenatal stress transmitted from the mother to the fetus? J Exp Biol 2024; 227:jeb246073. [PMID: 38449331 DOI: 10.1242/jeb.246073] [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] [Indexed: 03/08/2024]
Abstract
Prenatal stress programmes long-lasting neuroendocrine and behavioural changes in the offspring. Often this programming is maladaptive and sex specific. For example, using a rat model of maternal social stress in late pregnancy, we have demonstrated that adult prenatally stressed male, but not prenatally stressed female offspring display heightened anxiety-like behaviour, whereas both sexes show hyperactive hypothalamo-pituitary-adrenal (HPA) axis responses to stress. Here, we review the current knowledge of the mechanisms underpinning dysregulated HPA axis responses, including evidence supporting a role for reduced neurosteroid-mediated GABAergic inhibitory signalling in the brains of prenatally stressed offspring. How maternal psychosocial stress is signalled from the mother to the fetuses is unclear. Direct transfer of maternal glucocorticoids to the fetuses is often considered to mediate the programming effects of maternal stress on the offspring. However, protective mechanisms including attenuated maternal stress responses and placental 11β-hydroxysteroid dehydrogenase-2 (which inactivates glucocorticoids) should limit materno-fetal glucocorticoid transfer during pregnancy. Moreover, a lack of correlation between maternal stress, circulating maternal glucocorticoid levels and circulating fetal glucocorticoid levels is reported in several studies and across different species. Therefore, here we interrogate the evidence for a role for maternal glucocorticoids in mediating the effects of maternal stress on the offspring and consider the evidence for alternative mechanisms, including an indirect role for glucocorticoids and the contribution of changes in the placenta in signalling the stress status of the mother to the fetus.
Collapse
Affiliation(s)
- Ying Sze
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| | - Paula J Brunton
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
- Zhejiang University-University of Edinburgh Joint Institute, Haining, Zhejiang 314400, P.R. China
| |
Collapse
|
4
|
Bakhireva LN, Solomon E, Roberts MH, Ma X, Rai R, Wiesel A, Jacobson SW, Weinberg J, Milligan ED. Independent and Combined Effects of Prenatal Alcohol Exposure and Prenatal Stress on Fetal HPA Axis Development. Int J Mol Sci 2024; 25:2690. [PMID: 38473937 PMCID: PMC10932119 DOI: 10.3390/ijms25052690] [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: 01/19/2024] [Revised: 02/10/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Prenatal alcohol exposure (PAE) and prenatal stress (PS) are highly prevalent conditions known to affect fetal programming of the hypothalamic-pituitary-adrenal (HPA) axis. The objectives of this study were to assess the effect of light PAE, PS, and PAE-PS interaction on fetal HPA axis activity assessed via placental and umbilical cord blood biomarkers. Participants of the ENRICH-2 cohort were recruited during the second trimester and classified into the PAE and unexposed control groups. PS was assessed by the Perceived Stress Scale. Placental tissue was collected promptly after delivery; gene and protein analysis for 11β-HSD1, 11β-HSD2, and pCRH were conducted by qPCR and ELISA, respectively. Umbilical cord blood was analyzed for cortisone and cortisol. Pearson correlation and multivariable linear regression examined the association of PAE and PS with HPA axis biomarkers. Mean alcohol consumption in the PAE group was ~2 drinks/week. Higher PS was observed in the PAE group (p < 0.01). In multivariable modeling, PS was associated with pCRH gene expression (β = 0.006, p < 0.01), while PAE was associated with 11β-HSD2 protein expression (β = 0.56, p < 0.01). A significant alcohol-by-stress interaction was observed with respect to 11β-HSD2 protein expression (p < 0.01). Results indicate that PAE and PS may independently and in combination affect fetal programming of the HPA axis.
Collapse
Affiliation(s)
- Ludmila N. Bakhireva
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.H.R.); (X.M.); (R.R.); (A.W.)
| | - Elizabeth Solomon
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87106, USA; (E.S.); (E.D.M.)
| | - Melissa H. Roberts
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.H.R.); (X.M.); (R.R.); (A.W.)
| | - Xingya Ma
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.H.R.); (X.M.); (R.R.); (A.W.)
| | - Rajani Rai
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.H.R.); (X.M.); (R.R.); (A.W.)
| | - Alexandria Wiesel
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.H.R.); (X.M.); (R.R.); (A.W.)
| | - Sandra W. Jacobson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Joanne Weinberg
- Department of Cellular and Physiological Sciences, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
| | - Erin D. Milligan
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87106, USA; (E.S.); (E.D.M.)
| |
Collapse
|
5
|
Sheng JA, Handa RJ, Tobet SA. Evaluating different models of maternal stress on stress-responsive systems in prepubertal mice. Front Neurosci 2023; 17:1292642. [PMID: 38130695 PMCID: PMC10733493 DOI: 10.3389/fnins.2023.1292642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction Maternal adversity during pregnancy influences neurodevelopment in human and model animal offspring. Adversity can result from stressors coming from many different directions ranging from environmental to nutritional and physiological to immune (e.g., infection). Most stressors result in fetal overexposure to glucocorticoids that have been directly linked to long- and short-term negative impacts on neurological health of offspring. Neuropsychiatric diseases postulated to have fetal origins are diverse and include such things cardiovascular disease, obesity, affective disorders, and metabolic and immune disorders. Methods The experiments in the current study compare 3 stressors: prenatal exposure to dexamethasone (DEX), maternal high fat diet (HFD), and maternal caloric restriction (CR). Offspring of mothers with these treatments were examined prepubertally to evaluate stress responsiveness and stress-related behaviors in in male and female mice. Results Prenatal exposure to synthetic glucocorticoid, DEX, resulted in decreased neonatal body weights, reduced social interaction behavior, and hypoactive stress response offspring exposed to maternal DEX. Maternal CR resulted in decreased body weights and social interaction behavior in males and females and increased anxiety-like behavior and acute stress response only in males. HFD resulted in altered body weight gain in both sex offspring with decreased anxiety-like behavior in a female-biased manner. Discussion The idea that glucocorticoid responses to different stressors might serve as a common stimulus across stress paradigms is insufficient, given that different modes of prenatal stress produced differential effects. Opposite nutritional stressors produced similar outcomes for anxiety-like behavior in both sexes, social-like behavior in females, and a hyperactive adrenal stress response in males. One common theme among the three models of maternal stress (DEX, CR, and HFD) was consistent data showing their role in activating the maternal and fetal immune response. By tuning in on the more immediate immunological aspect on the developing fetus (e.g., hormones, cytokines), additional studies may tease out more direct outcomes of maternal stress in rodents and increase their translational value to human studies.
Collapse
Affiliation(s)
- Julietta A. Sheng
- Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Robert J. Handa
- Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Stuart A. Tobet
- Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Psychiatry, Mass General Hospital, Harvard Medical School, Boston, MA, United States
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
- Innovation Center on Sex Differences in Medicine, Mass General Hospital, Cambridge, MA, United States
| |
Collapse
|
6
|
Herzberg MP, Triplett R, McCarthy R, Kaplan S, Alexopoulos D, Meyer D, Arora J, Miller JP, Smyser TA, Herzog ED, England SK, Zhao P, Barch DM, Rogers CE, Warner BB, Smyser CD, Luby J. The Association Between Maternal Cortisol and Infant Amygdala Volume Is Moderated by Socioeconomic Status. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:837-846. [PMID: 37881545 PMCID: PMC10593881 DOI: 10.1016/j.bpsgos.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 02/25/2023] [Accepted: 03/11/2023] [Indexed: 10/27/2023] Open
Abstract
Background It has been well established that socioeconomic status is associated with mental and physical health as well as brain development, with emerging data suggesting that these relationships begin in utero. However, less is known about how prenatal socioeconomic environments interact with the gestational environment to affect neonatal brain volume. Methods Maternal cortisol output measured at each trimester of pregnancy and neonatal brain structure were assessed in 241 mother-infant dyads. We examined associations between the trajectory of maternal cortisol output across pregnancy and volumes of cortisol receptor-rich regions of the brain, including the amygdala, hippocampus, medial prefrontal cortex, and caudate. Given the known effects of poverty on infant brain structure, socioeconomic disadvantage was included as a moderating variable. Results Neonatal amygdala volume was predicted by an interaction between maternal cortisol output across pregnancy and socioeconomic disadvantage (standardized β = -0.31, p < .001), controlling for postmenstrual age at scan, infant sex, and total gray matter volume. Notably, amygdala volumes were positively associated with maternal cortisol for infants with maternal disadvantage scores 1 standard deviation below the mean (i.e., less disadvantage) (simple slope = 123.36, p < .01), while the association was negative in infants with maternal disadvantage 1 standard deviation above the mean (i.e., more disadvantage) (simple slope = -82.70, p = .02). Individuals with disadvantage scores at the mean showed no association, and there were no significant interactions in the other brain regions examined. Conclusions These data suggest that fetal development of the amygdala is differentially affected by maternal cortisol production at varying levels of socioeconomic advantage.
Collapse
Affiliation(s)
- Max P. Herzberg
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri
| | - Regina Triplett
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri
| | - Ronald McCarthy
- Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, Missouri
| | - Sydney Kaplan
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri
| | | | - Dominique Meyer
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri
| | - Jyoti Arora
- Department of Biostatistics, Washington University in St. Louis, St. Louis, Missouri
| | - J. Philip Miller
- Department of Biostatistics, Washington University in St. Louis, St. Louis, Missouri
| | - Tara A. Smyser
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri
| | - Erik D. Herzog
- Department of Biology, Washington University in St. Louis, St. Louis, Missouri
| | - Sarah K. England
- Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, Missouri
| | - Peinan Zhao
- Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, Missouri
| | - Deanna M. Barch
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri
- Department of Radiology, Washington University in St. Louis, St. Louis, Missouri
| | - Cynthia E. Rogers
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri
- Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri
| | - Barbara B. Warner
- Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri
| | - Christopher D. Smyser
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri
- Department of Radiology, Washington University in St. Louis, St. Louis, Missouri
- Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri
| | - Joan Luby
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri
| |
Collapse
|
7
|
Gundacker A, Cuenca Rico L, Stoehrmann P, Tillmann KE, Weber-Stadlbauer U, Pollak DD. Interaction of the pre- and postnatal environment in the maternal immune activation model. DISCOVER MENTAL HEALTH 2023; 3:15. [PMID: 37622027 PMCID: PMC10444676 DOI: 10.1007/s44192-023-00042-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 08/11/2023] [Indexed: 08/26/2023]
Abstract
Adverse influences during pregnancy are associated with a range of unfavorable outcomes for the developing offspring. Maternal psychosocial stress, exposure to infections and nutritional imbalances are known risk factors for neurodevelopmental derangements and according psychiatric and neurological manifestations later in offspring life. In this context, the maternal immune activation (MIA) model has been extensively used in preclinical research to study how stimulation of the maternal immune system during gestation derails the tightly coordinated sequence of fetal neurodevelopment. The ensuing consequence of MIA for offspring brain structure and function are majorly manifested in behavioral and cognitive abnormalities, phenotypically presenting during the periods of adolescence and adulthood. These observations have been interpreted within the framework of the "double-hit-hypothesis" suggesting that an elevated risk for neurodevelopmental disorders results from an individual being subjected to two adverse environmental influences at distinct periods of life, jointly leading to the emergence of pathology. The early postnatal period, during which the caregiving parent is the major determinant of the newborn´s environment, constitutes a window of vulnerability to external stimuli. Considering that MIA not only affects the developing fetus, but also impinges on the mother´s brain, which is in a state of heightened malleability during pregnancy, the impact of MIA on maternal brain function and behavior postpartum may importantly contribute to the detrimental consequences for her progeny. Here we review current information on the interaction between the prenatal and postnatal maternal environments in the modulation of offspring development and their relevance for the pathophysiology of the MIA model.
Collapse
Affiliation(s)
- Anna Gundacker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090 Vienna, Austria
| | - Laura Cuenca Rico
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090 Vienna, Austria
| | - Peter Stoehrmann
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090 Vienna, Austria
| | - Katharina E. Tillmann
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090 Vienna, Austria
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Daniela D. Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090 Vienna, Austria
| |
Collapse
|
8
|
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
|
9
|
Sze Y, Fernandes J, Kołodziejczyk ZM, Brunton PJ. Maternal glucocorticoids do not directly mediate the effects of maternal social stress on the fetus. J Endocrinol 2022; 255:143-158. [PMID: 36256689 PMCID: PMC9716396 DOI: 10.1530/joe-22-0226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022]
Abstract
Stress during pregnancy negatively affects the fetus and increases the risk for affective disorders in adulthood. Excess maternal glucocorticoids are thought to mediate fetal programming; however, whether they exert their effects directly or indirectly remains unclear. During pregnancy, protective mechanisms including maternal hypothalamic-pituitary-adrenal (HPA) axis hyporesponsiveness and placental 11β-hydroxysteroid dehydrogenase (11βHSD) type 2, which inactivates glucocorticoids, limit mother-to-fetus glucocorticoid transfer. However, whether repeated stress negatively impacts these mechanisms is not known. Pregnant rats were exposed to repeated social stress on gestational days (GD) 16-20 and several aspects of HPA axis and glucocorticoid regulation, including concentrations of glucocorticoids, gene expression for their receptors (Nr3c1, Nr3c2), receptor chaperones (Fkbp51, Fkbp52) and enzymes that control local glucocorticoid availability (Hsd11b1, Hsd11b2), were investigated in the maternal, placental and fetal compartments on GD20. The maternal HPA axis was activated following stress, though the primary driver was vasopressin, rather than corticotropin-releasing hormone. Despite the stress-induced increase in circulating corticosterone in the dams, only a modest increase was detected in the circulation of female fetuses, with no change in the fetal brain of either sex. Moreover, there was no change in the expression of genes that mediate glucocorticoid actions or modulate local concentrations in the fetal brain. In the placenta labyrinth zone, stress increased Hsd11b2 expression only in males and Fkbp51 expression only in females. Our results indicate that any role glucocorticoids play in fetal programming is likely indirect, perhaps through sex-dependent alterations in placental gene expression, rather than exerting effects via direct crossover into the fetal brain.
Collapse
Affiliation(s)
- Ying Sze
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | - Joana Fernandes
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | | | - Paula J Brunton
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, UK
- Zhejiang University-University of Edinburgh Institute, International Campus, Haining, Zhejiang, P.R. China
- Correspondence should be addressed to P J Brunton:
| |
Collapse
|
10
|
Durbagula S, Korlimarla A, Ravikumar G, Valiya Parambath S, Kaku SM, Visweswariah AM. Prenatal epigenetic factors are predisposing for neurodevelopmental disorders—Considering placenta as a model. Birth Defects Res 2022; 114:1324-1342. [DOI: 10.1002/bdr2.2119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022]
Affiliation(s)
- Srividhya Durbagula
- St. John's Medical College Bangalore India
- St. John's Research Institute Bangalore India
| | - Aruna Korlimarla
- St. John's Research Institute Bangalore India
- Department of Research Sri Shankara Cancer Hospital and Research Center Bangalore India
| | | | - Snijesh Valiya Parambath
- St. John's Medical College Bangalore India
- Department of Molecular Medicine St. John's Research Institute Bangalore India
| | - Sowmyashree Mayur Kaku
- St. John's Medical College Bangalore India
- Centre for Advanced Research and Excellence in Autism and Developmental Disorders (CARE ADD) St. John's Research Institute Bangalore India
| | - Ashok Mysore Visweswariah
- St. John's Medical College Bangalore India
- Centre for Advanced Research and Excellence in Autism and Developmental Disorders (CARE ADD) St. John's Research Institute Bangalore India
| |
Collapse
|
11
|
Hong JY. Developmental Programming by Perinatal Glucocorticoids. Mol Cells 2022; 45:685-691. [PMID: 36254710 PMCID: PMC9589377 DOI: 10.14348/molcells.2022.0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/29/2022] [Accepted: 06/15/2022] [Indexed: 11/07/2022] Open
Abstract
Early-life environmental factors can have persistent effects on physiological functions by altering developmental procedures in various organisms. Recent experimental and epidemiological studies now further support the idea that developmental programming is also present in mammals, including humans, influencing long-term health. Although the mechanism of programming is still largely under investigation, the role of endocrine glucocorticoids in developmental programming is gaining interest. Studies found that perinatal glucocorticoids have a persistent effect on multiple functions of the body, including metabolic, behavioral, and immune functions, in adulthood. Several mechanisms have been proposed to play a role in long-term programming. In this review, recent findings on this topic are summarized and the potential biological rationale behind this phenomenon is discussed.
Collapse
Affiliation(s)
- Jun Young Hong
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
12
|
Gene Dysregulation in the Adult Rat Paraventricular Nucleus and Amygdala by Prenatal Exposure to Dexamethasone. LIFE (BASEL, SWITZERLAND) 2022; 12:life12071077. [PMID: 35888164 PMCID: PMC9316520 DOI: 10.3390/life12071077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/24/2022] [Accepted: 07/15/2022] [Indexed: 12/02/2022]
Abstract
Fetal programming is the concept that maternal stressors during critical periods of fetal development can alter offspring phenotypes postnatally. Excess glucocorticoids can interact with the fetus to effect genetic and epigenetic changes implicated in adverse developmental outcomes. The present study investigates how chronic exposure to the synthetic glucocorticoid dexamethasone during late gestation alters the expression of genes related to behavior in brain areas relevant to the regulation and function of the hypothalamic–pituitary–adrenal axis. Pregnant Wistar Kyoto rats received subcutaneous injections of dexamethasone (100 μg/kg) daily from gestational day 15–21 or vehicle only as sham controls. The amygdala and paraventricular nucleus (PVN) were micro-punched to extract mRNA for reverse transcription and quantitative polymerase chain reaction for the analysis of the expression of specific genes. In the PVN, the expression of the glucocorticoid receptor NR3C1 was downregulated in female rats in response to programming. The expression of CACNA1C encoding the Cav1.2 pore subunit of L-type voltage-gated calcium channels was downregulated in male and female rats prenatally exposed to dexamethasone. Collectively, the results suggest that prenatal exposure to elevated levels of glucocorticoids plays a role in the dysregulation of the hypothalamic–pituitary–adrenal axis and potentially learning and memory by altering the expression of specific genes within the amygdala and PVN.
Collapse
|
13
|
Anifantaki F, Pervanidou P, Lambrinoudaki I, Panoulis K, Vlahos N, Eleftheriades M. Maternal Prenatal Stress, Thyroid Function and Neurodevelopment of the Offspring: A Mini Review of the Literature. Front Neurosci 2021; 15:692446. [PMID: 34566560 PMCID: PMC8455916 DOI: 10.3389/fnins.2021.692446] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/06/2021] [Indexed: 11/29/2022] Open
Abstract
Fetal brain is extremely plastic and vulnerable to environmental influences that may have long-term impact on health and development of the offspring. Both the Hypothalamic-Pituitary-Adrenal (HPA) and the Hypothalamic-Pituitary-Thyroid (HPT) axes are involved in stress responses, whereas, their final effectors, the Glucocorticoids (GCs) and the Thyroid Hormones (TH s), mediate several fundamental processes involved in neurodevelopment. The effects of these hormones on brain development are found to be time and dose-dependent. Regarding THs, the developing fetus depends on maternal supply of hormones, especially in the first half of pregnancy. It is acknowledged that inadequate or excess concentrations of both GCs and THs can separately cause abnormalities in the neuronal and glial structures and functions, with subsequent detrimental effects on postnatal neurocognitive function. Studies are focused on the direct impact of maternal stress and GC excess on growth and neurodevelopment of the offspring. Of particular interest, as results from recent literature data, is building understanding on how chronic stress and alterations of the HPA axis interacts and influences HPT axis and TH production. Animal studies have shown that increased GC concentrations related to maternal stress, most likely reduce maternal and thus fetal circulating THs, either directly or through modifications in the expression of placental enzymes responsible for regulating hormone levels in fetal microenvironment. The purpose of this review is to provide an update on data regarding maternal stress and its impact on fetal neurodevelopment, giving particular emphasis in the interaction of two axes and the subsequent thyroid dysfunction resulting from such circumstances.
Collapse
Affiliation(s)
- Foteini Anifantaki
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiota Pervanidou
- First Department of Paediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Irene Lambrinoudaki
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Panoulis
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikos Vlahos
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Makarios Eleftheriades
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
14
|
Creutzberg KC, Sanson A, Viola TW, Marchisella F, Begni V, Grassi-Oliveira R, Riva MA. Long-lasting effects of prenatal stress on HPA axis and inflammation: A systematic review and multilevel meta-analysis in rodent studies. Neurosci Biobehav Rev 2021; 127:270-283. [PMID: 33951412 DOI: 10.1016/j.neubiorev.2021.04.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/25/2021] [Accepted: 04/27/2021] [Indexed: 12/22/2022]
Abstract
Exposure to prenatal stress (PNS) can lead to long-lasting neurobiological and behavioral consequences for the offspring, which may enhance the susceptibility for mental disorders. The hypothalamus-pituitary-adrenal (HPA) axis and the immune system are two major factors involved in the stress response. Here, we performed a systematic review and meta-analysis of rodent studies that investigated the effects of PNS exposure on the HPA axis and inflammatory cytokines in adult offspring. Our analysis shows that animals exposed to PNS display a consistent increase in peripheral corticosterone (CORT) levels and central corticotrophin-releasing hormone (CRH), while decreased levels of its receptor 2 (CRHR2). Meta-regression revealed that sex and duration of PNS protocol are covariates that moderate these results. There was no significant effect of PNS in glucocorticoid receptor (GR), CRH receptor 1 (CRHR1), pro- and anti-inflammatory cytokines. Our findings suggest that PNS exposure elicits long-lasting effects on the HPA axis function, providing an important tool to investigate in preclinical settings key pathological aspects related to early-life stress exposure. Furthermore, researchers should be aware of the mixed outcomes of PNS on inflammatory markers in the adult brain.
Collapse
Affiliation(s)
- Kerstin Camile Creutzberg
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy.
| | - Alice Sanson
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy.
| | - Thiago Wendt Viola
- School of Medicine, Developmental Cognitive Neuroscience Lab, Pontifical Catholic University of Rio Grande do Sul, Avenida Ipiranga 6681, Building 12A, 90619-900, Porto Alegre, RS, Brazil.
| | - Francesca Marchisella
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy.
| | - Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy.
| | - Rodrigo Grassi-Oliveira
- School of Medicine, Developmental Cognitive Neuroscience Lab, Pontifical Catholic University of Rio Grande do Sul, Avenida Ipiranga 6681, Building 12A, 90619-900, Porto Alegre, RS, Brazil.
| | - Marco Andrea Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy; Biological Psychiatry Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| |
Collapse
|
15
|
Camm EJ, Cross CM, Kane AD, Tarry-Adkins JL, Ozanne SE, Giussani DA. Maternal antioxidant treatment protects adult offspring against memory loss and hippocampal atrophy in a rodent model of developmental hypoxia. FASEB J 2021; 35:e21477. [PMID: 33891326 DOI: 10.1096/fj.202002557rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/11/2021] [Accepted: 02/11/2021] [Indexed: 02/02/2023]
Abstract
Chronic fetal hypoxia is one of the most common outcomes in complicated pregnancy in humans. Despite this, its effects on the long-term health of the brain in offspring are largely unknown. Here, we investigated in rats whether hypoxic pregnancy affects brain structure and function in the adult offspring and explored underlying mechanisms with maternal antioxidant intervention. Pregnant rats were randomly chosen for normoxic or hypoxic (13% oxygen) pregnancy with or without maternal supplementation with vitamin C in their drinking water. In one cohort, the placenta and fetal tissues were collected at the end of gestation. In another, dams were allowed to deliver naturally, and offspring were reared under normoxic conditions until 4 months of age (young adult). Between 3.5 and 4 months, the behavior, cognition and brains of the adult offspring were studied. We demonstrated that prenatal hypoxia reduced neuronal number, as well as vascular and synaptic density, in the hippocampus, significantly impairing memory function in the adult offspring. These adverse effects of prenatal hypoxia were independent of the hypoxic pregnancy inducing fetal growth restriction or elevations in maternal or fetal plasma glucocorticoid levels. Maternal vitamin C supplementation during hypoxic pregnancy protected against oxidative stress in the placenta and prevented the adverse effects of prenatal hypoxia on hippocampal atrophy and memory loss in the adult offspring. Therefore, these data provide a link between prenatal hypoxia, placental oxidative stress, and offspring brain health in later life, providing insight into mechanism and identifying a therapeutic strategy.
Collapse
Affiliation(s)
- Emily J Camm
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - Christine M Cross
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - Andrew D Kane
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - Jane L Tarry-Adkins
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.,Cambridge Strategic Initiative in Reproduction, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK.,Cambridge Strategic Initiative in Reproduction, Cambridge, UK
| |
Collapse
|
16
|
Rensel MA, Schlinger BA. 11ß hydroxysteroid dehydrogenases regulate circulating glucocorticoids but not central gene expression. Gen Comp Endocrinol 2021; 305:113734. [PMID: 33548254 PMCID: PMC7954975 DOI: 10.1016/j.ygcen.2021.113734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/17/2021] [Accepted: 01/30/2021] [Indexed: 11/15/2022]
Abstract
Regulation of glucocorticoids (GCs), important mediators of physiology and behavior at rest and during stress, is multi-faceted and dynamic. The 11ß hydroxysteroid dehydrogenases 11ß-HSD1 and 11ß-HSD2 catalyze the regeneration and inactivation of GCs, respectively, and provide peripheral and central control over GC actions in mammals. While these enzymes have only recently been investigated in just two songbird species, central expression patterns suggest that they may function differently in birds and mammals, and little is known about how peripheral expression regulates circulating GCs. In this study, we utilized the 11ß-HSD inhibitor carbenoxolone (CBX) to probe the functional effects of 11ß-HSD activity on circulating GCs and central GC-dependent gene expression in the adult zebra finch (Taeniopygia guttata). Peripheral CBX injection produced a marked increase in baseline GCs 60 min after injection, suggestive of a dominant role for 11ß-HSD2 in regulating circulating GCs. In the adult zebra finch brain, where 11ß-HSD2 but not 11ß-HSD1 is expressed, co-incubation of micro-dissected brain regions with CBX and stress-level GCs had no impact on expression of several GC-dependent genes. These results suggest that peripheral 11ß-HSD2 attenuates circulating GCs, whereas central 11ß-HSD2 has little impact on gene expression. Instead, rapid 11ß-HSD2-based regulation of local GC levels might fine-tune membrane GC actions in brain. These results provide new insights into the dynamics of GC secretion and action in this important model organism.
Collapse
Affiliation(s)
- Michelle A Rensel
- Institute for Society and Genetics, University of California Los Angeles, 621 Charles E Young Drive S, Los Angeles, CA 90095, USA; Laboratory of Neuroendocrinology, Brain Research Institute UCLA, Box 951761, University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - Barney A Schlinger
- Laboratory of Neuroendocrinology, Brain Research Institute UCLA, Box 951761, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E Young Drive E, Los Angeles, CA 90095, USA; Department of Ecology and Evolutionary Biology, University of California Los Angeles, 621 Charles E Young Drive S, Los Angeles, CA 90095, USA
| |
Collapse
|
17
|
Kim HS, Choi HD, Pack JK, Kim N, Ahn YH. Biological Effects of Exposure to a Radiofrequency Electromagnetic Field on the Placental Barrier in Pregnant Rats. Bioelectromagnetics 2021; 42:191-199. [PMID: 33527465 PMCID: PMC8048814 DOI: 10.1002/bem.22322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 10/27/2020] [Accepted: 12/30/2020] [Indexed: 02/04/2023]
Abstract
The placenta protects the fetus against excessive stress‐associated maternal cortisol during pregnancy. We studied whether exposure to radiofrequency electromagnetic field (RF‐EMF) radiation during pregnancy can cause changes in dams and their placentas. Pregnant Sprague–Dawley rats were divided into cage‐control, sham‐exposed, and RF‐exposed groups. They were exposed to RF‐EMF signals at a whole‐body specific absorption rate of 4 W/kg for 8 h/day from gestational Day 1 to 19. Levels of cortisol in the blood, adrenal gland, and placenta were measured by enzyme‐linked immunosorbent assay. Levels of adrenocorticotropic hormone and corticotropin‐releasing hormone were monitored in maternal blood. Expression levels of placental 11β‐hydroxysteroid dehydrogenase type 2 (11β‐HSD2) messenger RNA (mRNA) were measured by reverse transcription polymerase chain reaction. Morphological changes in the placenta were analyzed using hematoxylin and eosin staining. Fetal parts of the placenta were measured using Zen 2.3 blue edition software. Maternal cortisol in circulating blood (RF: 230 ± 24.6 ng/ml and Sham: 156 ± 8.3 ng/ml) and the adrenal gland (RF: 58.3 ± 4.5 ng/ml and Sham: 30 ± 3.8 ng/ml) was significantly increased in the RF‐exposed group (P < 0.05). Placental cortisol was stably maintained, and the level of placental 11β‐HSD2 mRNA expression was not changed in the RF‐exposed group. RF‐EMF exposure during pregnancy caused a significant elevation of cortisol levels in circulating blood; however, no changes in the placental barrier were observed in pregnant rats. Bioelectromagnetics. © 2021 Bioelectromagnetics Society
Collapse
Affiliation(s)
- Hye Sun Kim
- Department of Neurosurgery, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hyung-Do Choi
- Radio Technology Research Department, Electronics and Telecommunications Research Institute, Daejeon, Republic of Korea
| | - Jeong-Ki Pack
- Department of Radio Sciences and Engineering, College of Engineering, Chungnam National University, Daejeon, Republic of Korea
| | - Nam Kim
- School of Electrical and Computer Engineering, Chungbuk National University, Cheongju, Republic of Korea
| | - Young Hwan Ahn
- Department of Neurosurgery, Ajou University School of Medicine, Suwon, Republic of Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
| |
Collapse
|
18
|
Stoye DQ, Blesa M, Sullivan G, Galdi P, Lamb GJ, Black GS, Quigley AJ, Thrippleton MJ, Bastin ME, Reynolds RM, Boardman JP. Maternal cortisol is associated with neonatal amygdala microstructure and connectivity in a sexually dimorphic manner. eLife 2020; 9:60729. [PMID: 33228850 PMCID: PMC7685701 DOI: 10.7554/elife.60729] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 10/25/2020] [Indexed: 12/21/2022] Open
Abstract
The mechanisms linking maternal stress in pregnancy with infant neurodevelopment in a sexually dimorphic manner are poorly understood. We tested the hypothesis that maternal hypothalamic-pituitary-adrenal axis activity, measured by hair cortisol concentration (HCC), is associated with microstructure, structural connectivity, and volume of the infant amygdala. In 78 mother-infant dyads, maternal hair was sampled postnatally, and infants underwent magnetic resonance imaging at term-equivalent age. We found a relationship between maternal HCC and amygdala development that differed according to infant sex. Higher HCC was associated with higher left amygdala fractional anisotropy (β = 0.677, p=0.010), lower left amygdala orientation dispersion index (β = −0.597, p=0.034), and higher fractional anisotropy in connections between the right amygdala and putamen (β = 0.475, p=0.007) in girls compared to boys. Furthermore, altered amygdala microstructure was only observed in boys, with connectivity changes restricted to girls. Maternal cortisol during pregnancy is related to newborn amygdala architecture and connectivity in a sexually dimorphic manner. Given the fundamental role of the amygdala in the emergence of emotion regulation, these findings offer new insights into mechanisms linking maternal health with neuropsychiatric outcomes of children. Stress during pregnancy, for example because of mental or physical disorders, can have long-term effects on child development. Epidemiological studies have shown that individuals exposed to stress in the womb are at higher risk of developmental and mood conditions, such as ADHD and depression. This effect is different between the sexes, and the biological mechanisms that underpin these observations are poorly understood. One possibility is that a baby’s developing amygdala, the part of the brain that processes emotions, is affected by a signal known as cortisol. This hormone is best known for its role in coordinating the stress response, but it also directs the growth of a fetus. Tracking fetal amygdala changes as well as cortisol levels in the pregnant individual could explain how stress during pregnancy affects development. To investigate, Stoye et al. recruited nearly 80 volunteers and their newborn children. MRI scans were used to examine the structure of the amygdala, and how it is connected to other parts of the brain. In parallel, the amount of cortisol was measured in hair samples collected from the volunteers around the time of birth, which reflects stress levels during the final three months of pregnancy. Linking the brain imaging results to the volunteers’ cortisol levels showed that being exposed to higher cortisol levels in the womb affected babies in different ways based on their sex: boys showed alterations in the fine structure of their amygdala, while girls displayed changes in the way that brain region connected to other neural networks. The work by Stoye et al. potentially reveals a biological mechanism by which early exposure to stress could affect brain development differently between the sexes, potentially informing real-world interventions.
Collapse
Affiliation(s)
- David Q Stoye
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Manuel Blesa
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Gemma Sullivan
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Paola Galdi
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Gillian J Lamb
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Gill S Black
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Alan J Quigley
- Department of Radiology, Royal Hospital for Sick Children, Edinburgh, United Kingdom
| | - Michael J Thrippleton
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Mark E Bastin
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Rebecca M Reynolds
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - James P Boardman
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
19
|
Psychological stress and cortisol during pregnancy: An ecological momentary assessment (EMA)-Based within- and between-person analysis. Psychoneuroendocrinology 2020; 121:104848. [PMID: 32927180 PMCID: PMC7895320 DOI: 10.1016/j.psyneuen.2020.104848] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Although the linkage between psychological stress and cortisol is believed to mediate the association of stress with health outcomes, several studies have been unable to demonstrate this association. We suggest this inability may be a consequence of limitations in the measurement approach and/or reliance on analytic strategies that focus on associations across, rather than within individuals. The link between psychological stress and cortisol is of particular interest in the context of pregnancy and fetal development. Using an ecological momentary assessment (EMA) design, we examined the association between psychological stress and cortisol at the between- and the within-person level. METHODS 152 participants completed a 4-day long EMA protocol serially in early, mid and late pregnancy to provide momentary stress appraisals (average of 150 measures/subject) and saliva samples (average of 55 samples/subject) for quantification of cortisol. The association between stress and cortisol was estimated using linear mixed models. RESULTS After accounting for the effects of key determinants of variation in cortisol, momentary stress was significantly and positively associated with cortisol at the within-person level (B = .030, p = .031), but not at the between-person level. No association was evident for traditional retrospective measures of stress with cortisol at either the between- or the within-person level. CONCLUSIONS Our study highlights the value of EMA methods and linear mixed-modeling approaches in linking maternal psychological and physiological states across pregnancy. These findings may have important implications for the development of personalized risk identification and "just-in-time" intervention strategies to optimize maternal and child health.
Collapse
|
20
|
Wang G, Huang Y, Hu T, Zhang B, Tang Z, Yao R, Huang Y, Fan X, Ni X. Contribution of placental 11β-HSD2 to the pathogenesis of preeclampsia. FASEB J 2020; 34:15379-15399. [PMID: 32978833 DOI: 10.1096/fj.202001003rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Preeclampsia, a major human pregnancy-specific disorder, leads to maternal and fetal morbidity and mortality. Here we reported that 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2), an enzyme that degrades active glucocorticoids, is one of the key factors that contributes to preeclampsia development. In the pregnant rat model, we firstly confirmed that administration of 11β-HSD2 inhibitor carbenoxolone (CBX) subcutaneously or by placenta-targeted delivery system could lead to a decrease in placental 11β-HSD2 expression and activity and an increase in corticosterone level in placenta and maternal circulation. Then, we showed that subcutaneous administration and placenta-targeted delivery of CBX resulted in the hallmark of preeclampsia-like features including hypertension, proteinuria, renal damages as well as elevated circulatory soluble fms-like tyrosine kinase 1 (sFlt1) and increased sFlt1/placental growth factor (PlGF) ratio in pregnant rats. These animals displayed decreased trophoblast invasion in uterus, impaired spiral artery remodeling, and reduced placental blood flow. Preeclampsia-like features could also be induced by administration of dexamethasone in pregnant rats. In the cultured human trophoblast models, we found that cortisol only inhibited migration and invasion of the extravillous trophoblasts with 11β-HSD2 knockdown, and promoted sFlt1 release in the cultured syncytiotrophoblasts with 11β-HSD2 knockdown. Furthermore, we elucidated that cortisol stimulated a disintegrin and metalloprotease (ADAM)17 expression in placentas, thereby promoting sFlt1 release in placenta. Collectively, our study provided the evidence that placental 11β-HSD2 dysfunction plays a key role in the development of preeclampsia and immediately identified innovative target to counteract preeclampsia.
Collapse
Affiliation(s)
- Gang Wang
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, China.,Department of Physiology, Second Military Medical University, Shanghai, China
| | - Yan Huang
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, China.,Department of Physiology, Second Military Medical University, Shanghai, China
| | - Tianxiao Hu
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Baozhen Zhang
- Center for Reproduction and Health Development, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhengshan Tang
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, China
| | - Ruojing Yao
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, China
| | - Ying Huang
- Maternity and Child Health Hospital of Pudong New District, Shanghai, China
| | - Xiujun Fan
- Center for Reproduction and Health Development, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xin Ni
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, China.,Department of Physiology, Second Military Medical University, Shanghai, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
| |
Collapse
|
21
|
Dachew BA, Scott JG, Mamun A, Fetene DM, Alati R. Maternal hypertensive disorders during pregnancy and the trajectories of offspring emotional and behavioral problems: the ALSPAC birth cohort study. Ann Epidemiol 2020; 53:63-68.e1. [PMID: 32950656 DOI: 10.1016/j.annepidem.2020.08.015] [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] [Received: 12/04/2019] [Revised: 06/20/2020] [Accepted: 08/18/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE This study examined the effect of hypertensive disorders during pregnancy on trajectories of emotional and behavioral problems in offspring. METHODS We used data from the Avon Longitudinal Study of Parents and Children, a prospective birth cohort study in Avon, United Kingdom. A group-based trajectory modeling was used to identify the distinct trajectories of emotional and behavioral problems in children at four time points: at age 3.5, 6.75, 9, and 11 years. Multinomial logistic regression analyses were used to examine the association between hypertensive disorders during pregnancy and trajectories of emotional and behavioral problems. RESULTS We identified four trajectories of offspring emotional and behavioral problems: normal (42.6%), borderline decreasing (40.6%), borderline stable (10.0%), and persistently elevated (6.8%). We found that children exposed to maternal pre-eclampsia were more likely to be in the persistently elevated symptom trajectory (OR = 2.72; 95% CI: 1.10-6.74) than in the normal trajectory group. We found no associations between maternal gestational hypertension and trajectories of offspring emotional and behavioral problems. CONCLUSIONS Maternal pre-eclampsia, but not gestational hypertension was associated with persistently elevated trajectory of offspring emotional and behavioral problems. Our findings highlight that the antenatal environment is important for children's behavioral and emotional development.
Collapse
Affiliation(s)
- Berihun Assefa Dachew
- Institute for Social Science Research, The University of Queensland, Brisbane, Australia; School of Public Health, Curtin University, Perth, Australia; Department of Epidemiology and Biostatistics, Institute of Public Health, University of Gondar, Gondar, Ethiopia.
| | - James G Scott
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, Australia; Metro North Mental Health, Royal Brisbane and Women's Hospital, Herston, Australia
| | - Abdullah Mamun
- Institute for Social Science Research, The University of Queensland, Brisbane, Australia
| | | | - Rosa Alati
- Institute for Social Science Research, The University of Queensland, Brisbane, Australia; School of Public Health, Curtin University, Perth, Australia
| |
Collapse
|
22
|
Shallie PD, Margolis D, Shallie OF, Naicker T. Placental 11β-HSD2 downregulated in HIV associated preeclampsia. J Reprod Immunol 2020; 142:103185. [PMID: 32853845 DOI: 10.1016/j.jri.2020.103185] [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: 05/20/2020] [Revised: 06/29/2020] [Accepted: 08/08/2020] [Indexed: 11/24/2022]
Abstract
Preeclampsia (PE) and human immunodeficiency virus (HIV) have been linked with marked increases in maternal stress, resulting in a significant change in placental function ranging from alterations in placental structure to the precise and delicate transformations in placental gene expression. Such changes may lead to altered transport of essential signals to the fetus, which can have long-term impacts on offspring health and consequently affect fetal neurodevelopment. Therefore, this work investigated the role of placental 11β-hydroxysteroid dehydrogenase types 2 (11β-HSD2) in HIV associated preeclampsia. The placenta were obtained from 76 pregnant women, which were stratified based on pregnancy type and HIV status into; Normotensive HIV negative, normotensive HIV positive, PE HIV negative and PE HIV positive. The placental tissue was processed for immunocytochemistry and stained with rabbit polyclonal to 11β-HSD2 Our results showed significant downregulation in the placental expression of 11β-HSD2 in both the conducting and exchange villi of PE and HIV-positive patients when compared with Normotensive and HIV-negative individuals, respectively. Our results provide inferential evidence for comorbidity of PE and HIV in the downregulation of placental 11β-HSD2 enzyme function, which mediates the programmed outcomes of an adverse maternal environment during pregnancy and long-term impacts on offspring health and consequently affects fetal neurodevelopment.
Collapse
Affiliation(s)
- Philemon Dauda Shallie
- Optics and Imaging Centre, School of Laboratory Medicine and Medical Sciences, Nelson Mandela Medical School, University of KwaZulu-Natal, Durban, South Africa; Department of Anatomy, Obafemi Awolowo College of Health Sciences, Olabisi Onabanjo University, Ago-Iwoye, Ogun State, Nigeria.
| | - Denise Margolis
- Optics and Imaging Centre, School of Laboratory Medicine and Medical Sciences, Nelson Mandela Medical School, University of KwaZulu-Natal, Durban, South Africa
| | - Oluwadamilola Faith Shallie
- Discipline of Physiology, School of Laboratory Medicine and Medical Sciences, Nelson Mandela Medical School, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, School of Laboratory Medicine and Medical Sciences, Nelson Mandela Medical School, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
23
|
Carrera SC, Sen S, Heistermann M, Lu A, Beehner JC. Low rank and primiparity increase fecal glucocorticoid metabolites across gestation in wild geladas. Gen Comp Endocrinol 2020; 293:113494. [PMID: 32333913 DOI: 10.1016/j.ygcen.2020.113494] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 03/07/2020] [Accepted: 04/21/2020] [Indexed: 11/20/2022]
Abstract
Integrative behavioral ecology requires accurate and non-invasive measures of hormone mediators for the study of wild animal populations. Biologically sensitive assay systems for the measurement of hormones and their metabolites need to be validated for the species and sample medium (e.g. urine, feces, saliva) of interest. Where more than one assay is available for hormone (metabolite) measurement, antibody selection is useful in identifying the assay that tracks changes in an individuaĺs endocrine activity best, i.e., the most biologically sensitive assay. This is particularly important when measuring how glucocorticoids (GCs) respond to the subtle, additive effects of acute stressors during a predictable metabolic challenge, such as gestation. Here, we validate a group-specific enzyme immunoassay, measuring immunoreactive 11β-hydroxyetiocholanolone, for use in a wild primate, geladas (Theropithecus gelada). This group-specific assay produced values correlated with those from a previously validated double-antibody, corticosterone 125I radioimmunoassay. However, the results with the group-specific assay showed a stronger response to an ACTH challenge and identified greater variation in gelada immunoreactive fecal glucocorticoid metabolites (iGCMs) compared with the corticosterone assay, indicating a higher biological sensitivity for assessing adrenocortical activity. We then used the group-specific assay to: (1) determine the normative pattern of iGCM levels across gelada gestation, and (2) identify the ecological, social, and individual factors that influence GC output for pregnant females. Using a general additive mixed model, we found that higher iGCM levels were associated with low rank (compared to high rank) and first time mothers (compared to multiparous mothers). This study highlights the importance of assay selection and the efficacy of group-specific assays for hormonal research in non-invasively collected samples. Additionally, in geladas, our results identify some of the factors that increase GC output over and above the already-elevated GC concentrations associated with gestation. In the burgeoning field of maternal stress, these factors can be examined to identify the effects that GC elevations may have on offspring development.
Collapse
Affiliation(s)
- Sofia C Carrera
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Sharmi Sen
- Department of Anthropology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michael Heistermann
- Endocrinology Laboratory, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Amy Lu
- Department of Anthropology, SUNY Stony Brook, Stony Brook, NY 11794, USA
| | - Jacinta C Beehner
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Anthropology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
24
|
Seo M, Anderson G. Gut-Amygdala Interactions in Autism Spectrum Disorders: Developmental Roles via regulating Mitochondria, Exosomes, Immunity and microRNAs. Curr Pharm Des 2020; 25:4344-4356. [PMID: 31692435 DOI: 10.2174/1381612825666191105102545] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 11/01/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Autism Spectrum Disorders (ASD) have long been conceived as developmental disorder. A growing body of data highlights a role for alterations in the gut in the pathoetiology and/or pathophysiology of ASD. Recent work shows alterations in the gut microbiome to have a significant impact on amygdala development in infancy, suggesting that the alterations in the gut microbiome may act to modulate not only amygdala development but how the amygdala modulates the development of the frontal cortex and other brain regions. METHODS This article reviews wide bodies of data pertaining to the developmental roles of the maternal and foetal gut and immune systems in the regulation of offspring brain development. RESULTS A number of processes seem to be important in mediating how genetic, epigenetic and environmental factors interact in early development to regulate such gut-mediated changes in the amygdala, wider brain functioning and inter-area connectivity, including via regulation of microRNA (miR)-451, 14-3-3 proteins, cytochrome P450 (CYP)1B1 and the melatonergic pathways. As well as a decrease in the activity of monoamine oxidase, heightened levels of in miR-451 and CYP1B1, coupled to decreased 14-3-3 act to inhibit the synthesis of N-acetylserotonin and melatonin, contributing to the hyperserotonemia that is often evident in ASD, with consequences for mitochondria functioning and the content of released exosomes. These same factors are likely to play a role in regulating placental changes that underpin the association of ASD with preeclampsia and other perinatal risk factors, including exposure to heavy metals and air pollutants. Such alterations in placental and gut processes act to change the amygdala-driven biological underpinnings of affect-cognitive and affect-sensory interactions in the brain. CONCLUSION Such a perspective readily incorporates previously disparate bodies of data in ASD, including the role of the mu-opioid receptor, dopamine signaling and dopamine receptors, as well as the changes occurring to oxytocin and taurine levels. This has a number of treatment implications, the most readily applicable being the utilization of sodium butyrate and melatonin.
Collapse
Affiliation(s)
- Moonsang Seo
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - George Anderson
- CRC Scotland & London, Eccleston Square, London, United Kingdom
| |
Collapse
|
25
|
Finik J, Buthmann J, Zhang W, Go K, Nomura Y. Placental Gene Expression and Offspring Temperament Trajectories: Predicting Negative Affect in Early Childhood. JOURNAL OF ABNORMAL CHILD PSYCHOLOGY 2020; 48:783-795. [PMID: 32185610 DOI: 10.1007/s10802-020-00632-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Exposure to prenatal stress increases offspring risk for long-term neurobehavioral impairments and psychopathology, such as Attention Deficit Hyperactivity Disorder (ADHD). Epigenetic regulation of glucocorticoid pathway genes may be a potential underlying mechanism by which maternal conditions 'program' the fetal brain for downstream vulnerabilities. The present study aims to investigate whether mRNA expression of glucocorticoid pathway genes in the placenta predict offspring negative affect during early childhood (between 6 and 24 months). Participants include 318 mother-child dyads participating in a longitudinal birth cohort study. Placental mRNA expression of glucocorticoid pathway genes (HSD11B1, HSD11B2, NR3C1, NCOR2) were profiled and negative affect traits of the offspring were measured at 6, 12, 18, and 24 months. HSD11B1 mRNA expression significantly predicted negative affect (β = -.09, SE = .04; p = .036), and Distress to Limitations trajectories (β = -.13, SE = .06; p = .016). NCOR2 mRNA expression significantly predicted Distress to Limitations (β = .43, SE = .21; p = .047), and marginally predicted Sadness trajectories (β = .39, SE = .21; p = .068). HSD11B2 and NR3C1 did not predict trajectories of Negative Affect or subscale scores. Infant negative affect traits were assessed via maternal self-report, and deviated from linearity across follow-up. mRNA expression of glucocorticoid pathway genes in the placenta may be a potentially novel tool for early identification of infants at greater risk for elevated negative affect. Further study is needed to validate the utility of mRNA expression of glucocorticoid pathway genes in the placenta.
Collapse
Affiliation(s)
- J Finik
- CUNY Graduate School of Public Health and Health Policy, Department of Epidemiology and Biostatistics, 55 W 125th St., New York, NY, 10027, USA.
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY, 11367, USA.
| | - J Buthmann
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY, 11367, USA
- CUNY Graduate Center, Department of Psychology, 365 5th Avenue, New York, NY, 10016, USA
| | - W Zhang
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY, 11367, USA
- New Jersey City University, Department of Psychology, 2039 John Fitzgerald Kennedy Blvd, Jersey City, NJ, 07305, USA
| | - K Go
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY, 11367, USA
- Nova Southeastern University, Dr. Kiran C. Patel College of Osteopathic Medicine, 3301 College Avenue, Fort Lauderdale, FL, 33314, USA
| | - Y Nomura
- CUNY Queens College, Department of Psychology, 65-30 Kissena Blvd, Flushing, NY, 11367, USA
- CUNY Graduate Center, Department of Psychology, 365 5th Avenue, New York, NY, 10016, USA
- CUNY Advanced Science Research Center, Graduate Center, 85 St Nicholas Terrace, New York, NY, 10031, USA
- Icahn School of Medicine at Mount Sinai, Department of Psychiatry, 1 Gustave Levy Pl, New York, NY, 10029, USA
| |
Collapse
|
26
|
Taka-Eilola Nèe Riekki T, Veijola J, Miettunen J, Koskela J, Kantojärvi L, Mäki P. Antisocial and borderline personality disorders in the offspring of antenatally depressed mothers - a follow-up until mid-adulthood in the Northern Finland 1966 birth cohort. Nord J Psychiatry 2020; 74:138-146. [PMID: 31647361 DOI: 10.1080/08039488.2019.1681508] [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] [Indexed: 01/22/2023]
Abstract
Background: Maternal depression is common during pregnancy, affecting 10-15% of mothers. In previous reports, the offspring of antenatally depressed mothers have had an elevated risk for antisocial, criminal and violent behaviour in adolescence, and for borderline personality features in childhood, but long-term outcomes are unknown.Aims: To study whether the adult offspring of antenatally depressed mothers have an elevated risk for antisocial (ASPD) or borderline personality disorder (BPD) when followed until mid-adulthood.Methods: In the general population-based Northern Finland 1966 Birth Cohort, mothers of 12,058 children were asked during mid-gestation if they felt depressed. Of the mothers, 14% reported being depressed. The offspring were followed for 49 years. The diagnoses of in- and outpatient-treated ASPD and BPD in the offspring were detected using the Finnish Care Register for Healthcare. Maternal antenatal smoking, newborn´s low birthweight or short gestational age, father's social class, and family type at birth were considered as confounding variables. Logistic regression analyses on the potential confounders were performed. Maternal postnatal depression and paternal ASPD information was not available.Results: In the male offspring of antenatally depressed mothers, the risk for ASPD was elevated (adjusted odds ratio 5.6; 95% confidence interval 1.8-17.8), but not in female offspring. The risk for BPD was not elevated in the offspring of antenatally depressed mothers in this study.Conclusions: The sons of antenatally depressed mothers had an increased risk for ASPD. Prevention and treatment of antenatal depression might present an opportunity to decrease the risk of antisocial personality in the offspring.
Collapse
Affiliation(s)
- Tiina Taka-Eilola Nèe Riekki
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, Oulu, Finland.,Basic Health Care District of Kallio, Finland
| | - Juha Veijola
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, Oulu, Finland.,Department of Psychiatry, Oulu University Hospital, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Jouko Miettunen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Center for Life Course Health Research, University of Oulu, Oulu, Finland
| | - Jari Koskela
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, Oulu, Finland
| | - Liisa Kantojärvi
- Department of Psychiatry, Oulu University Hospital, Oulu, Finland
| | - Pirjo Mäki
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, Oulu, Finland.,Department of Psychiatry, Oulu University Hospital, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Department of Psychiatry, Länsi-Pohja Healthcare District, Kemi, Finland.,Department of Psychiatry, The Middle Ostrobothnia Central Hospital, Soite Mental Health Services, Joint Municipal Authority of Wellbeing in Raahe District, Mental Health Services and Basic Health Care District of Kallio, Finland.,Department of Psychiatry, Kainuu Central Hospital, Kainuu Social and Healthcare District, Kainuu, Finland
| |
Collapse
|
27
|
Sze Y, Brunton PJ. Sex, stress and steroids. Eur J Neurosci 2019; 52:2487-2515. [DOI: 10.1111/ejn.14615] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Ying Sze
- Centre for Discovery Brain Sciences University of Edinburgh Edinburgh UK
| | - Paula J. Brunton
- Centre for Discovery Brain Sciences University of Edinburgh Edinburgh UK
- Zhejiang University‐University of Edinburgh Joint Institute Haining Zhejiang China
| |
Collapse
|
28
|
Abstract
Early Life Stress Stress and daily hassles are a normal part of day-to day-life. The amount of control that is experienced strongly contributes to resilience and coping. Children very frequently do not experience control over the stressors within their lives. Starting from pregnancy, they are subjected - via the maternal endocrine system - to a variety of stressors ranging from normal stress regarding the transition to parenthood to maternal abuse or torture. This article collects research of the last two decades regarding the influence of stress on the developing brain. Both, animal and human studies will shed light on the effect of pre- and postnatal stress demonstrating an influence of early life stressors reaching far into adulthood. A direct influence of stress on multiple developmental characteristics has been postulated and shown. The results of this review will underline the necessity of early life programs focusing stress reduction and resilience in children and their parents. Also, a need for programs targeting stress reduction in pregnancy will be demonstrated and emphasized.
Collapse
|
29
|
Sawyer KM, Zunszain PA, Dazzan P, Pariante CM. Intergenerational transmission of depression: clinical observations and molecular mechanisms. Mol Psychiatry 2019; 24:1157-1177. [PMID: 30283036 DOI: 10.1038/s41380-018-0265-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 08/16/2018] [Accepted: 08/20/2018] [Indexed: 02/06/2023]
Abstract
Maternal mental illness can have a devastating effect during the perinatal period, and has a profound impact on the care that the baby receives and on the relationships that the baby forms. This review summarises clinical evidence showing the effects of perinatal depression on offspring physical and behavioural development, and on the transmission of psychopathology between generations. We then evaluate a number of factors which influence this relationship, such as genetic factors, the use of psychotropic medications during pregnancy, the timing within the perinatal period, the sex of the foetus, and exposure to maltreatment in childhood. Finally, we examine recent findings regarding the molecular mechanisms underpinning these clinical observations, and identify relevant epigenetic and biomarker changes in the glucocorticoid, oxytocin, oestrogen and immune systems, as key biological mediators of these clinical findings. By understanding these molecular mechanisms in more detail, we will be able to improve outcomes for both mothers and their offspring for generations.
Collapse
Affiliation(s)
- Kristi M Sawyer
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Patricia A Zunszain
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Paola Dazzan
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Carmine M Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
| |
Collapse
|
30
|
Agnew EJ, Garcia-Burgos A, Richardson RV, Manos H, Thomson AJW, Sooy K, Just G, Homer NZM, Moran CM, Brunton PJ, Gray GA, Chapman KE. Antenatal dexamethasone treatment transiently alters diastolic function in the mouse fetal heart. J Endocrinol 2019; 241:279-292. [PMID: 31013474 PMCID: PMC6541236 DOI: 10.1530/joe-18-0666] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 04/23/2019] [Indexed: 12/27/2022]
Abstract
Endogenous glucocorticoid action is important in the structural and functional maturation of the fetal heart. In fetal mice, although glucocorticoid concentrations are extremely low before E14.5, glucocorticoid receptor (GR) is expressed in the heart from E10.5. To investigate whether activation of cardiac GR prior to E14.5 induces precocious fetal heart maturation, we administered dexamethasone in the drinking water of pregnant dams from E12.5 to E15.5. To test the direct effects of glucocorticoids upon the cardiovascular system we used SMGRKO mice, with Sm22-Cre-mediated disruption of GR in cardiomyocytes and vascular smooth muscle. Contrary to expectations, echocardiography showed no advancement of functional maturation of the fetal heart. Moreover, litter size was decreased 2 days following cessation of antenatal glucocorticoid exposure, irrespective of fetal genotype. The myocardial performance index and E/A wave ratio, markers of fetal heart maturation, were not significantly affected by dexamethasone treatment in either genotype. Dexamethasone treatment transiently decreased the myocardial deceleration index (MDI; a marker of diastolic function), in control fetuses at E15.5, with recovery by E17.5, 2 days after cessation of treatment. MDI was lower in SMGRKO than in control fetuses and was unaffected by dexamethasone. The transient decrease in MDI was associated with repression of cardiac GR in control fetuses following dexamethasone treatment. Measurement of glucocorticoid levels in fetal tissue and hypothalamic corticotropin-releasing hormone (Crh) mRNA levels suggest complex and differential effects of dexamethasone treatment upon the hypothalamic-pituitary-adrenal axis between genotypes. These data suggest potentially detrimental and direct effects of antenatal glucocorticoid treatment upon fetal heart function.
Collapse
Affiliation(s)
- E J Agnew
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - A Garcia-Burgos
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - R V Richardson
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - H Manos
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - A J W Thomson
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - K Sooy
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - G Just
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - N Z M Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - C M Moran
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - P J Brunton
- Centre for Discovery Brain Sciences, The University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, UK
| | - G A Gray
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - K E Chapman
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
- Correspondence should be addressed to K E Chapman:
| |
Collapse
|
31
|
Dachew BA, Scott JG, Mamun A, Alati R. Hypertensive disorders of pregnancy and the risk of anxiety disorders in adolescence: Findings from the Avon Longitudinal Study of Parents and Children. J Psychiatr Res 2019; 110:159-165. [PMID: 30641349 DOI: 10.1016/j.jpsychires.2019.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/10/2018] [Accepted: 01/02/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND The effect of hypertensive disorders of pregnancy (HDP) on offspring anxiety disorders in adolescence is not yet known. This study aims to examine the association between HDP and offspring anxiety disorders at age 15 years. METHODS We used data from 5231 mother-offspring pairs from the United Kingdom based Avon Longitudinal Study of Parents and Children (ALSPAC). Anxiety disorder was diagnosed in the offspring at the age of 15 years using the Development and Well-Being Assessment (DAWBA). RESULTS Among those who had anxiety disorders, 16.4% were exposed to HDP. After adjusting for a wide range of known confounders, we found that adolescents of women with HDP had a 2.43 fold (95% CI: 1.41-4.19) increase risk of anxiety disorders compared with adolescents of women without HDP. CONCLUSIONS Our study showed that adolescents exposed to HDP had higher risk of anxiety disorders compared with unexposed adolescents and suggests that prevention and treatment of maternal HDP could possibly prevent offspring anxiety in adolescence. Early screening for anxiety disorders in offspring of women with HDP may also be warranted. Further research is needed to explain the pathways by which HDP may increase the risk of offspring psychopathology.
Collapse
Affiliation(s)
- Berihun Assefa Dachew
- Institute for Social Science Research, The University of Queensland, Brisbane, Australia; Department of Epidemiology and Biostatistics, Institute of Public Health, University of Gondar, Gondar, Ethiopia.
| | - James G Scott
- Faculty of Medicine, School of Public Health, The University of Queensland, Herston, Australia; Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, Australia; Metro North Mental Health, Royal Brisbane and Women's Hospital, Herston, Australia
| | - Abdullah Mamun
- Institute for Social Science Research, The University of Queensland, Brisbane, Australia
| | - Rosa Alati
- Institute for Social Science Research, The University of Queensland, Brisbane, Australia; School of Public Health, Curtin University, Western Australia, Australia
| |
Collapse
|
32
|
Graham AM, Rasmussen JM, Entringer S, Ben Ward E, Rudolph MD, Gilmore JH, Styner M, Wadhwa PD, Fair DA, Buss C. Maternal Cortisol Concentrations During Pregnancy and Sex-Specific Associations With Neonatal Amygdala Connectivity and Emerging Internalizing Behaviors. Biol Psychiatry 2019; 85:172-181. [PMID: 30122286 PMCID: PMC6632079 DOI: 10.1016/j.biopsych.2018.06.023] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/06/2018] [Accepted: 06/22/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND Maternal cortisol during pregnancy has the potential to influence rapidly developing fetal brain systems that are commonly altered in neurodevelopmental and psychiatric disorders. Research examining maternal cortisol concentrations across pregnancy and offspring neurodevelopment proximal to birth is needed to advance understanding in this area and lead to insight into the etiology of these disorders. METHODS Participants were 70 adult women recruited during early pregnancy and their infants born after 34 weeks gestation. Maternal cortisol concentrations were assessed serially over 4 days in early, mid, and late gestation. Resting state functional connectivity magnetic resonance imaging of the neonatal amygdala was examined. Mothers reported on children's internalizing behavior problems at 24 months of age. RESULTS Maternal cortisol concentrations during pregnancy were significantly associated with neonatal amygdala connectivity in a sex-specific manner. Elevated maternal cortisol was associated with stronger amygdala connectivity to brain regions involved in sensory processing and integration, as well as the default mode network in girls, and with weaker connectivity to these brain regions in boys. Elevated maternal cortisol was associated with higher internalizing symptoms in girls only, and this association was mediated by stronger neonatal amygdala connectivity. CONCLUSIONS Normative variation in maternal cortisol during pregnancy is associated with the coordinated functioning of the amygdala soon after birth in a sex-specific manner. The identified pathway from maternal cortisol to higher internalizing symptoms in girls via alterations in neonatal amygdala connectivity may be relevant for the etiology of sex differences in internalizing psychiatric disorders, which are more prevalent in women.
Collapse
Affiliation(s)
- Alice M Graham
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| | - Jerod M Rasmussen
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, California
| | - Sonja Entringer
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, California; Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Medical Psychology, Berlin, Germany
| | - Elizabeth Ben Ward
- Department of Computer Science, University of California, Irvine, Irvine, California
| | - Marc D Rudolph
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, North Carolina
| | - John H Gilmore
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - Martin Styner
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - Pathik D Wadhwa
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, California.
| | - Damien A Fair
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon; Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon
| | - Claudia Buss
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, California; Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Medical Psychology, Berlin, Germany
| |
Collapse
|
33
|
Schmidt M, Rauh M, Schmid MC, Huebner H, Ruebner M, Wachtveitl R, Cordasic N, Rascher W, Menendez-Castro C, Hartner A, Fahlbusch FB. Influence of Low Protein Diet-Induced Fetal Growth Restriction on the Neuroplacental Corticosterone Axis in the Rat. Front Endocrinol (Lausanne) 2019; 10:124. [PMID: 30915031 PMCID: PMC6421269 DOI: 10.3389/fendo.2019.00124] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 02/11/2019] [Indexed: 01/21/2023] Open
Abstract
Objectives: Placental steroid metabolism is linked to the fetal hypothalamus-pituitary-adrenal axis. Intrauterine growth restriction (IUGR) might alter this cross-talk and lead to maternal stress, in turn contributing to the pathogenesis of anxiety-related disorders of the offspring, which might be mediated by fetal overexposure to, or a reduced local enzymatic protection against maternal glucocorticoids. So far, direct evidence of altered levels of circulating/local glucocorticoids is scarce. Liquid chromatography tandem-mass spectrometry (LC-MS/MS) allows quantitative endocrine assessment of blood and tissue. Using a rat model of maternal protein restriction (low protein [LP] vs. normal protein [NP]) to induce IUGR, we analyzed fetal and maternal steroid levels via LC-MS/MS along with the local expression of 11beta-hydroxysteroid-dehydrogenase (Hsd11b). Methods: Pregnant Wistar dams were fed a low protein (8%, LP; IUGR) or an isocaloric normal protein diet (17%, NP; controls). At E18.5, the expression of Hsd11b1 and 2 was determined by RT-PCR in fetal placenta and brain. Steroid profiling of maternal and fetal whole blood, fetal brain, and placenta was performed via LC-MS/MS. Results: In animals with LP-induced reduced body (p < 0.001) and placental weights (p < 0.05) we did not observe any difference in the expressional Hsd11b1/2-ratio in brain or placenta. Moreover, LP diet did not alter corticosterone (Cort) or 11-dehydrocorticosterone (DH-Cort) levels in dams, while fetal whole blood levels of Cort were significantly lower in the LP group (p < 0.001) and concomitantly in LP brain (p = 0.003) and LP placenta (p = 0.002). Maternal and fetal progesterone levels (whole blood and tissue) were not influenced by LP diet. Conclusion: Various rat models of intrauterine stress show profound alterations in placental Hsd11b2 gatekeeper function and fetal overexposure to corticosterone. In contrast, LP diet in our model induced IUGR without altering maternal steroid levels or placental enzymatic glucocorticoid barrier function. In fact, IUGR offspring showed significantly reduced levels of circulating and local corticosterone. Thus, our LP model might not represent a genuine model of intrauterine stress. Hypothetically, the observed changes might reflect a fetal attempt to maintain anabolic conditions in the light of protein restriction to sustain regular brain development. This may contribute to fetal origins of later neurodevelopmental sequelae.
Collapse
Affiliation(s)
- Marius Schmidt
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Matthias C. Schmid
- Institute of Medical Biometry, Informatics and Epidemiology, Faculty of Medicine, Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Hanna Huebner
- Department of Gynaecology and Obstetrics/Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Matthias Ruebner
- Department of Gynaecology and Obstetrics/Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Rainer Wachtveitl
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Nada Cordasic
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Wolfgang Rascher
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Carlos Menendez-Castro
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Andrea Hartner
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Fabian B. Fahlbusch
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
- *Correspondence: Fabian B. Fahlbusch
| |
Collapse
|
34
|
Zhao X, Liu Q, Cao S, Pang J, Zhang H, Feng T, Deng Y, Yao J, Li H. A meta-analysis of selective serotonin reuptake inhibitors (SSRIs) use during prenatal depression and risk of low birth weight and small for gestational age. J Affect Disord 2018; 241:563-570. [PMID: 30153640 DOI: 10.1016/j.jad.2018.08.061] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 06/10/2018] [Accepted: 08/12/2018] [Indexed: 01/23/2023]
Abstract
BACKGROUND Selective Serotonin Reuptake Inhibitors (SSRIs) act as the first-line antidepressants prescribed for the treatment of prenatal depression. Evidence from previous studies has suggested that the use of SSRIs treatment for prenatal depression has adversely affected fetal growth. However, these results are inconsistent and inconclusive. METHODS In this study, we examined whether SSRIs use during pregnancy was related to low birth weight (LBW) and small for gestational age (SGA) using a meta-analysis approach. Relevant studies were retrieved by database searching and selected according to established inclusion criteria. RESULTS Fifteen articles involved 1,977,446 subjects were identified that tested the relationship between the SSRIs use, LBW and SGA outcomes. Statistical analyses revealed a significant association between SSRIs use and suboptimal fetal growth (RR = 1.45, 95% CI = 1.18 - 1.76, Z = 3.62, p = 0.00 for SGA; RR = 1.38, 95% CI = 1.13 - 1.69, Z = 3.14, p = 0.00 for LBW). LIMITATIONS These results must be treated with caution as we did not take the confounding factors into account (e.g., trimester SSRIs taken, specific SSRIs prescribed and maternal lifestyle during pregnancy) to elucidate their specific roles in the relationship between SSRIs use during pregnancy and fetal growth. CONCLUSION Our findings suggested that SSRIs use for prenatal depression is associated with suboptimal fetal growth.
Collapse
Affiliation(s)
- Xiaofeng Zhao
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, PR China
| | - Qian Liu
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, PR China
| | - Suxia Cao
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, PR China
| | - Jianyue Pang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, PR China
| | - Huijie Zhang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, PR China
| | - Tingting Feng
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, PR China
| | - Yajie Deng
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, PR China
| | - Jing Yao
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, PR China
| | - Hengfen Li
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, PR China.
| |
Collapse
|
35
|
Keenan K, Hipwell AE, Class QA, Mbayiwa K. Extending the developmental origins of disease model: Impact of preconception stress exposure on offspring neurodevelopment. Dev Psychobiol 2018; 60:753-764. [PMID: 30144041 PMCID: PMC6342272 DOI: 10.1002/dev.21773] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/18/2018] [Accepted: 07/20/2018] [Indexed: 01/03/2023]
Abstract
The concept of the developmental origins of health and disease via prenatal programming has informed many etiologic models of health and development. Extensive experimental research in non-human animal models has revealed the impact of in utero exposure to stress on fetal development and neurodevelopment later in life. Stress exposure, however, is unlikely to occur de novo following conception, and pregnancy health is not independent of the health of the system prior to conception. For these reasons, the preconception period is emerging as an important new focus for research on adverse birth outcomes and offspring neurodevelopment. In this review, we summarize the existing evidence for the role of preconception stress exposure on pregnancy health and offspring neurodevelopment across species and discuss the implications of this model for addressing health disparities in obstetrics and offspring outcomes.
Collapse
Affiliation(s)
- Kate Keenan
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois
| | - Alison E Hipwell
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Quetzal A Class
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, Illinois
| | - Kimberley Mbayiwa
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois
| |
Collapse
|
36
|
Ni L, Pan Y, Tang C, Xiong W, Wu X, Zou C. Antenatal exposure to betamethasone induces placental 11β-hydroxysteroid dehydrogenase type 2 expression and the adult metabolic disorders in mice. PLoS One 2018; 13:e0203802. [PMID: 30212527 PMCID: PMC6136781 DOI: 10.1371/journal.pone.0203802] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 08/07/2018] [Indexed: 12/23/2022] Open
Abstract
Antenatal overexposure to glucocorticoids causes fetal intrauterine growth restriction (IUGR) and adult metabolic disorders. 11β-hydroxysteroid dehydrogenase (11β-HSD) 1 and 2 are key enzymes for glucocorticoid metabolism, however, the detailed effects of antenatal overexposure to glucocorticoids on placental 11β-HSD1 and 2 expression and adult metabolic disorders remain obscure. Here, we report that, in placenta 11β-HSD1 is diffusely localized, whereas 11β-HSD2 is specifically expressed in labyrinthine layer. Exposure of pregnant dams to betamethasone significantly increases the expression of placental 11β-HSD2 but not 11β-HSD1, and decreases the weights of fetuses but not placentas. Antenatal exposure to betamethasone leads to either significant weight loss in the offspring younger than 10-week-old, or weight gain in those older than 14-week-old. Furthermore, antenatal exposure to betamethasone results in coexistence of various metabolic disorders in adult offspring, including hyperglycemia, glucose intolerance, low insulin secretory capacity and hyperlipidemia. The present study demonstrates that exposure of pregnant dams to betamethasone induces the expression of placental 11β-HSD2 but not 11β-HSD1, leads to fetal IUGR and causes adult metabolic disorders, providing evidence for fetal origins of adult diseases and the potential role of placental 11β-HSD2 in them.
Collapse
Affiliation(s)
- Li Ni
- Department of Endocrinology, the Children Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China
- Jiaxing Maternity and Child Health Care Hospital, Jiaxing, China
| | - Yibin Pan
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chao Tang
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenyi Xiong
- Department of Endocrinology, the Children Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ximei Wu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China
- * E-mail: (XW); (CZ)
| | - Chaochun Zou
- Department of Endocrinology, the Children Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- * E-mail: (XW); (CZ)
| |
Collapse
|
37
|
Walker SE, Papilloud A, Huzard D, Sandi C. The link between aberrant hypothalamic–pituitary–adrenal axis activity during development and the emergence of aggression—Animal studies. Neurosci Biobehav Rev 2018; 91:138-152. [DOI: 10.1016/j.neubiorev.2016.10.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 10/09/2016] [Accepted: 10/12/2016] [Indexed: 11/29/2022]
|
38
|
Social dominance predicts hippocampal glucocorticoid receptor recruitment and resilience to prenatal adversity. Sci Rep 2018; 8:9595. [PMID: 29941995 PMCID: PMC6018627 DOI: 10.1038/s41598-018-27988-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/12/2018] [Indexed: 12/19/2022] Open
Abstract
The developing fetus is highly sensitive to prenatal stress, which may alter Hypothalamic-Pituitary-Adrenal (HPA) axis programming and increase the risk of behavioral disorders. There is high variability among the human population, wherein many offspring of stressed pregnancies display resilience to adversity, while the remainder displays vulnerability. In order to identify biological substrates mediating between resilience or vulnerability to prenatal adversity, we exposed stress-resistant Dominant (Dom) and stress-sensitive Submissive (Sub) mice to mild prenatal restraint stress (PRS, 45 min on gestational days (GD) 15, 16 and 17). We hypothesized that PRS would differentially alter prenatal programming of limbic regions regulating the HPA axis and affect among Dom and Sub offspring. Indeed, PRS increased Sub offspring’s serum corticosterone, and exaggerated their anxiety- and depressive-like behavior, while Dom offspring remained resilient to the hormonal and behavioral consequences of PRS. Moreover, PRS exposure markedly facilitated glucocorticoid receptor (GR) recruitment to the hippocampus among Dom mice in response to restraint stress, which may be responsible for their resilience to stressful challenge. These findings suggest proclivity to adaptive or maladaptive prenatal programming of hippocampal GR recruitment to be inheritable and predictable by social dominance or submissiveness.
Collapse
|
39
|
Gobinath AR, Wong S, Chow C, Lieblich SE, Barr AM, Galea LAM. Maternal exercise increases but concurrent maternal fluoxetine prevents the increase in hippocampal neurogenesis of adult offspring. Psychoneuroendocrinology 2018; 91:186-197. [PMID: 29579632 DOI: 10.1016/j.psyneuen.2018.02.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/14/2018] [Accepted: 02/23/2018] [Indexed: 12/21/2022]
Abstract
Treating postpartum depression (PPD) with pharmacological antidepressants like fluoxetine (FLX) is complicated because these drugs can remain active in breast milk and potentially affect infant development. Alternatively, non-pharmacological treatments such as exercise are associated with beneficial effects on infant development but its potential ability to counter the effects of PPD are largely unknown. To investigate this, we treated dams with corticosterone (CORT) or vehicle (sesame oil) from postpartum days 2-25 to model PPD. Within oil and CORT treatments, dams were also assigned to one of these treatments: 1) exercise (voluntary running wheel) + FLX (10 mg/kg, i.p.), 2) exercise + saline (vehicle for FLX), 3) no exercise + FLX, 4) no exercise + saline. Both male and female offspring were analyzed, and this generated a total of 16 experimental groups for this study. Adult male and female offspring (125 d old) of these dams were tested for anxiety-like behavior in the novelty suppressed feeding test and stress reactivity in the dexamethasone suppression test. Hippocampal tissue was processed for doublecortin, a protein expressed in immature neurons. Regardless of sex, maternal exercise increased neurogenesis in the dorsal hippocampus of adult offspring, but concurrent exposure to maternal fluoxetine prevented this effect. Exposure to either maternal exercise or maternal FLX facilitated HPA negative feedback in adult males but not females. Maternal postpartum CORT also facilitated HPA feedback in adult offspring of both sexes. Collectively, these data indicate that maternal exercise increased dorsal hippocampal neurogenesis in both sexes but differentially affected offspring HPA axis based on sex. Alternatively, maternal postpartum FLX facilitated HPA axis negative feedback only in males. These findings indicate that different types of maternal interventions bear long-term effects on offspring outcome with implications for treating PPD.
Collapse
Affiliation(s)
- Aarthi R Gobinath
- Graduate Program in Neuroscience, University of British Columbia, Canada
| | - Sarah Wong
- Department of Psychology, University of British Columbia, Canada
| | - Carmen Chow
- Department of Psychology, University of British Columbia, Canada
| | | | - Alasdair M Barr
- Graduate Program in Neuroscience, University of British Columbia, Canada; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Canada; Centre for Brain Health, University of British Columbia, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Canada; Department of Psychology, University of British Columbia, Canada; Centre for Brain Health, University of British Columbia, Canada.
| |
Collapse
|
40
|
Effects of early-life malnutrition on neurodevelopment and neuropsychiatric disorders and the potential mechanisms. Prog Neuropsychopharmacol Biol Psychiatry 2018; 83:64-75. [PMID: 29287829 DOI: 10.1016/j.pnpbp.2017.12.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 12/21/2017] [Accepted: 12/24/2017] [Indexed: 02/08/2023]
Abstract
Lines of evidence have demonstrated that early-life malnutrition is highly correlated with neurodevelopment and adulthood neuropsychiatric disorders, while some findings are conflicting with each other. In addition, the biological mechanisms are less investigated. We systematically reviewed the evidence linking early-life nutrition status with neurodevelopment and clinical observations in human and animal models. We summarized the effects of special nutritious on neuropsychiatric disorders and explored the underlying potential mechanisms. The further understanding of the biological regulation of early-life nutritional status on neurodevelopment might shed light on precision nutrition at an integrative systems biology framework.
Collapse
|
41
|
St-Cyr S, McGowan PO. Adaptation or pathology? The role of prenatal stressor type and intensity in the developmental programing of adult phenotype. Neurotoxicol Teratol 2018; 66:113-124. [DOI: 10.1016/j.ntt.2017.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/25/2017] [Accepted: 12/04/2017] [Indexed: 01/06/2023]
|
42
|
Miranda A, Sousa N. Maternal hormonal milieu influence on fetal brain development. Brain Behav 2018; 8:e00920. [PMID: 29484271 PMCID: PMC5822586 DOI: 10.1002/brb3.920] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [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
|
43
|
Xu J, Wang R, Liu Y, Liu D, Jiang H, Pan F. FKBP5 and specific microRNAs via glucocorticoid receptor in the basolateral amygdala involved in the susceptibility to depressive disorder in early adolescent stressed rats. J Psychiatr Res 2017; 95:102-113. [PMID: 28826069 DOI: 10.1016/j.jpsychires.2017.08.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/28/2017] [Accepted: 08/11/2017] [Indexed: 12/11/2022]
Abstract
Exposure to stressful events induces depressive-like symptoms and increases susceptibility to depression. However, the molecular mechanisms are not fully understood. Studies reported that FK506 binding protein51 (FKBP5), the co-chaperone protein of glucocorticoid receptors (GR), plays a crucial role. Further, miR-124a and miR-18a are involved in the regulation of FKBP5/GR function. However, few studies have referred to effects of early life stress on depressive-like behaviours, GR and FKBP5, as well as miR-124a and miR-18a in the basolateral amygdala (BLA) from adolescence to adulthood. This study aimed to examine the dynamic alternations of depressive-like behaviours, GR and FKBP5, as well as miR-124a and miR-18a expressions in the BLA of chronic unpredictable mild stress (CUMS) rats and dexamethasone administration rats during the adolescent period. Meanwhile, the GR antagonist, RU486, was used as a means of intervention. We found that CUMS and dexamethasone administration in the adolescent period induced permanent depressive-like behaviours and memory impairment, decreased GR expression, and increased FKBP5 and miR-124a expression in the BLA of both adolescent and adult rats. However, increased miR-18a expression in the BLA was found only in adolescent rats. Depressive-like behaviours were positively correlated with the level of miR-124a, whereas GR levels were negatively correlated with those in both adolescent and adult rats. Our results suggested FKBP5/GR and miR-124a in the BLA were associated with susceptibility to depressive disorder in the presence of stressful experiences in early life.
Collapse
Affiliation(s)
- Jingjing Xu
- Department of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong, 250012, China.
| | - Rui Wang
- Department of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong, 250012, China.
| | - Yuan Liu
- Department of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong, 250012, China.
| | - Dexiang Liu
- Department of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong, 250012, China.
| | - Hong Jiang
- Department of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong, 250012, China.
| | - Fang Pan
- Department of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong, 250012, China.
| |
Collapse
|
44
|
Child neurodevelopmental outcomes following preterm and term birth: What can the placenta tell us? Placenta 2017; 57:79-86. [DOI: 10.1016/j.placenta.2017.06.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/17/2017] [Accepted: 06/12/2017] [Indexed: 11/21/2022]
|
45
|
van Bodegom M, Homberg JR, Henckens MJAG. Modulation of the Hypothalamic-Pituitary-Adrenal Axis by Early Life Stress Exposure. Front Cell Neurosci 2017; 11:87. [PMID: 28469557 PMCID: PMC5395581 DOI: 10.3389/fncel.2017.00087] [Citation(s) in RCA: 331] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/13/2017] [Indexed: 12/20/2022] Open
Abstract
Exposure to stress during critical periods in development can have severe long-term consequences, increasing overall risk on psychopathology. One of the key stress response systems mediating these long-term effects of stress is the hypothalamic-pituitary-adrenal (HPA) axis; a cascade of central and peripheral events resulting in the release of corticosteroids from the adrenal glands. Activation of the HPA-axis affects brain functioning to ensure a proper behavioral response to the stressor, but stress-induced (mal)adaptation of the HPA-axis' functional maturation may provide a mechanistic basis for the altered stress susceptibility later in life. Development of the HPA-axis and the brain regions involved in its regulation starts prenatally and continues after birth, and is protected by several mechanisms preventing corticosteroid over-exposure to the maturing brain. Nevertheless, early life stress (ELS) exposure has been reported to have numerous consequences on HPA-axis function in adulthood, affecting both its basal and stress-induced activity. According to the match/mismatch theory, encountering ELS prepares an organism for similar ("matching") adversities during adulthood, while a mismatching environment results in an increased susceptibility to psychopathology, indicating that ELS can exert either beneficial or disadvantageous effects depending on the environmental context. Here, we review studies investigating the mechanistic underpinnings of the ELS-induced alterations in the structural and functional development of the HPA-axis and its key external regulators (amygdala, hippocampus, and prefrontal cortex). The effects of ELS appear highly dependent on the developmental time window affected, the sex of the offspring, and the developmental stage at which effects are assessed. Albeit by distinct mechanisms, ELS induced by prenatal stressors, maternal separation, or the limited nesting model inducing fragmented maternal care, typically results in HPA-axis hyper-reactivity in adulthood, as also found in major depression. This hyper-activity is related to increased corticotrophin-releasing hormone signaling and impaired glucocorticoid receptor-mediated negative feedback. In contrast, initial evidence for HPA-axis hypo-reactivity is observed for early social deprivation, potentially reflecting the abnormal HPA-axis function as observed in post-traumatic stress disorder, and future studies should investigate its neural/neuroendocrine foundation in further detail. Interestingly, experiencing additional (chronic) stress in adulthood seems to normalize these alterations in HPA-axis function, supporting the match/mismatch theory.
Collapse
Affiliation(s)
| | | | - Marloes J. A. G. Henckens
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and BehaviourRadboudumc, Nijmegen, Netherlands
| |
Collapse
|
46
|
Vismara L. Perspectives on perinatal stressful and traumatic experiences. EUROPEAN JOURNAL OF TRAUMA & DISSOCIATION 2017. [DOI: 10.1016/j.ejtd.2017.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
47
|
Zouikr I, Karshikoff B. Lifetime Modulation of the Pain System via Neuroimmune and Neuroendocrine Interactions. Front Immunol 2017; 8:276. [PMID: 28348566 PMCID: PMC5347117 DOI: 10.3389/fimmu.2017.00276] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/24/2017] [Indexed: 12/12/2022] Open
Abstract
Chronic pain is a debilitating condition that still is challenging both clinicians and researchers. Despite intense research, it is still not clear why some individuals develop chronic pain while others do not or how to heal this disease. In this review, we argue for a multisystem approach to understand chronic pain. Pain is not only to be viewed simply as a result of aberrant neuronal activity but also as a result of adverse early-life experiences that impact an individual's endocrine, immune, and nervous systems and changes which in turn program the pain system. First, we give an overview of the ontogeny of the central nervous system, endocrine, and immune systems and their windows of vulnerability. Thereafter, we summarize human and animal findings from our laboratories and others that point to an important role of the endocrine and immune systems in modulating pain sensitivity. Taking "early-life history" into account, together with the past and current immunological and endocrine status of chronic pain patients, is a necessary step to understand chronic pain pathophysiology and assist clinicians in tailoring the best therapeutic approach.
Collapse
Affiliation(s)
- Ihssane Zouikr
- Laboratory for Molecular Mechanisms of Thalamus Development, RIKEN BSI , Wako , Japan
| | - Bianka Karshikoff
- Department of Clinical Neuroscience, Division for Psychology, Karolinska Institutet, Solna, Sweden; Stress Research Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
48
|
Räikkönen K, Martikainen S, Pesonen AK, Lahti J, Heinonen K, Pyhälä R, Lahti M, Tuovinen S, Wehkalampi K, Sammallahti S, Kuula L, Andersson S, Eriksson JG, Ortega-Alonso A, Reynolds RM, Strandberg TE, Seckl JR, Kajantie E. Maternal Licorice Consumption During Pregnancy and Pubertal, Cognitive, and Psychiatric Outcomes in Children. Am J Epidemiol 2017; 185:317-328. [PMID: 28158597 DOI: 10.1093/aje/kww172] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 09/19/2016] [Indexed: 12/15/2022] Open
Abstract
Earlier puberty, especially in girls, is associated with physical and mental disorders. Prenatal glucocorticoid exposure influences the timing of puberty in animal models, but the human relevance of those findings is unknown. We studied whether voluntary consumption of licorice, which contains glycyrrhizin (a potent inhibitor of placental 11β-hydroxysteroid dehydrogenase type 2, the "barrier" to maternal glucocorticoids), by pregnant women was associated with pubertal maturation (height, weight, body mass index for age, difference between current and expected adult height, Tanner staging, score on the Pubertal Development Scale), neuroendocrine function (diurnal salivary cortisol, dexamethasone suppression), cognition (neuropsychological tests), and psychiatric problems (as measured by the Child Behavior Checklist) in their offspring. The children were born in 1998 in Helsinki, Finland, and examined during 2009-2011 (mean age = 12.5 (standard deviation (SD), 0.4) years; n = 378). Girls exposed to high maternal glycyrrhizin consumption (≥500 mg/week) were taller (mean difference (MD) = 0.4 SD, 95% confidence interval (CI): 0.1, 0.8), were heavier (MD = 0.6 SD, 95% CI: 0.2, 1.9), and had higher body mass index for age (MD = 0.6 SD, 95% CI: 0.2, 0.9). They were also 0.5 standard deviations (95% CI: 0.2, 0.8) closer to adult height and reported more advanced pubertal development (P < 0.04). Girls and boys exposed to high maternal glycyrrhizin consumption scored 7 (95% CI: 3.1, 11.2) points lower on tests of intelligence quotient, had poorer memory (P < 0.04), and had 3.3-fold (95% CI: 1.4, 7.7) higher odds of attention deficit/hyperactivity disorder problems compared with children whose mothers consumed little to no glycyrrhizin (≤249 mg/week). No differences in cortisol levels were found. Licorice consumption during pregnancy may be associated with harm for the developing offspring.
Collapse
|
49
|
Beck KR, Bächler M, Vuorinen A, Wagner S, Akram M, Griesser U, Temml V, Klusonova P, Yamaguchi H, Schuster D, Odermatt A. Inhibition of 11β-hydroxysteroid dehydrogenase 2 by the fungicides itraconazole and posaconazole. Biochem Pharmacol 2017; 130:93-103. [PMID: 28131847 DOI: 10.1016/j.bcp.2017.01.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/23/2017] [Indexed: 02/01/2023]
Abstract
Impaired 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2)-dependent cortisol inactivation can lead to electrolyte dysbalance, hypertension and cardiometabolic disease. Furthermore, placental 11β-HSD2 essentially protects the fetus from high maternal glucocorticoid levels, and its impaired function has been associated with altered fetal growth and a higher risk for cardio-metabolic diseases in later life. Despite its important role, 11β-HSD2 is not included in current off-target screening approaches. To identify potential 11β-HSD inhibitors among approved drugs, a pharmacophore model was used for virtual screening, followed by biological assessment of selected hits. This led to the identification of several azole fungicides as 11β-HSD inhibitors, showing a significant structure-activity relationship between azole scaffold size, 11β-HSD enzyme selectivity and inhibitory potency. A hydrophobic linker connecting the azole ring to the other, more polar end of the molecule was observed to be favorable for 11β-HSD2 inhibition and selectivity over 11β-HSD1. The most potent 11β-HSD2 inhibition, using cell lysates expressing recombinant human 11β-HSD2, was obtained for itraconazole (IC50 139±14nM), its active metabolite hydroxyitraconazole (IC50 223±31nM) and posaconazole (IC50 460±98nM). Interestingly, experiments with mouse and rat kidney homogenates showed considerably lower inhibitory activity of these compounds towards 11β-HSD2, indicating important species-specific differences. Thus, 11β-HSD2 inhibition by these compounds is likely to be overlooked in preclinical rodent studies. Inhibition of placental 11β-HSD2 by these compounds, in addition to the known inhibition of cytochrome P450 enzymes and P-glycoprotein efflux transport, might contribute to elevated local cortisol levels, thereby affecting fetal programming.
Collapse
Affiliation(s)
- Katharina R Beck
- Swiss Center for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| | - Murielle Bächler
- Swiss Center for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| | - Anna Vuorinen
- Swiss Center for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| | - Sandra Wagner
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), Computer Aided Molecular Design Group, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Muhammad Akram
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), Computer Aided Molecular Design Group, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Ulrich Griesser
- Institute of Pharmacy/Pharmaceutical Technology, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Veronika Temml
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), Computer Aided Molecular Design Group, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Petra Klusonova
- Swiss Center for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| | - Hideaki Yamaguchi
- Department of Applied Biological Chemistry, Meijo University, Nagoya 468-8502, Japan.
| | - Daniela Schuster
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), Computer Aided Molecular Design Group, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Alex Odermatt
- Swiss Center for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| |
Collapse
|
50
|
Prenatal Stress and Neurodevelopmental Plasticity: Relevance to Psychopathology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1015:117-129. [DOI: 10.1007/978-3-319-62817-2_7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|