1
|
Sheng JA, Tobet SA. Maternal immune activation with toll-like receptor 7 agonist during mid-gestation alters juvenile and adult developmental milestones and behavior. J Neuroendocrinol 2024; 36:e13417. [PMID: 38822791 PMCID: PMC11296912 DOI: 10.1111/jne.13417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 05/07/2024] [Accepted: 05/15/2024] [Indexed: 06/03/2024]
Abstract
Infections during pregnancy are associated with increased risk for adult neuropsychiatric disease, such as major depressive disorder, schizophrenia, and autism spectrum disorder. In mouse models of maternal immune activation (MIA), different toll-like receptors (TLRs) are stimulated to initiate inflammatory responses in mother and fetus. The goal of this study was to determine sex-dependent aspects of MIA using a TLR7/8 agonist, Resiquimod (RQ), on neurodevelopment. RQ was administered to timed-pregnant mice on embryonic day (E) 12.5. At E15, maternal/fetal plasma cytokines were measured by enzyme-linked immunosorbent assay (ELISA). Maternal cytokines interleukin (IL)-6 and IL-10 were higher while tumor necrosis factor (TNF)-α and IL-17 were lower in pregnant dams exposed to RQ. Fetal cytokines (E15) were altered at the same timepoint with fetal plasma IL-6 and IL-17 greater after RQ compared to vehicle, while IL-10 and TNF-α were higher in male fetuses but not female. Other timed-pregnant dams were allowed to give birth. MIA with RQ did not alter the female to male ratio of offspring born per litter. Body weights were reduced significantly in both sexes at birth, and over the next 5 weeks. Offspring from RQ-injected mothers opened their eyes 5 days later than controls. Similarly, female offspring from RQ-injected mothers exhibited pubertal delay based on vaginal opening 2-3 days later than control females. On the behavioral side, juvenile and adult male and female MIA offspring exhibited less social-like behavior in a social interaction test. Anhedonia-like behavior was greater in MIA adult female mice. This study provides support for sex-dependent influences of fetal antecedents for altered brain development and behavioral outputs that could be indicative of increased susceptibility for adult disorders through immune mechanisms. Future studies are needed to determine neural cellular and molecular mechanisms for such programming effects.
Collapse
Affiliation(s)
| | - Stuart A. Tobet
- Biomedical Sciences, Colorado State University, Fort Collins, CO
- Department of Psychiatry, Mass General Hospital, Harvard Medical School, Boston, MA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO
- Innovation Center on Sex Differences in Medicine, Mass General Hospital
| |
Collapse
|
2
|
Castillo-Ruiz A, Gars A, Sturgeon H, Ronczkowski NM, Pyaram DN, Dauriat CJG, Chassaing B, Forger NG. Brain effects of gestating germ-free persist in mouse neonates despite acquisition of a microbiota at birth. Front Neurosci 2023; 17:1130347. [PMID: 37207179 PMCID: PMC10188942 DOI: 10.3389/fnins.2023.1130347] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/10/2023] [Indexed: 05/21/2023] Open
Abstract
At birth, mammals experience a massive colonization by microorganisms. We previously reported that newborn mice gestated and born germ-free (GF) have increased microglial labeling and alterations in developmental neuronal cell death in the hippocampus and hypothalamus, as well as greater forebrain volume and body weight when compared to conventionally colonized (CC) mice. To test whether these effects are solely due to differences in postnatal microbial exposure, or instead may be programmed in utero, we cross-fostered GF newborns immediately after birth to CC dams (GF→CC) and compared them to offspring fostered within the same microbiota status (CC→CC, GF→GF). Because key developmental events (including microglial colonization and neuronal cell death) shape the brain during the first postnatal week, we collected brains on postnatal day (P) 7. To track gut bacterial colonization, colonic content was also collected and subjected to 16S rRNA qPCR and Illumina sequencing. In the brains of GF→GF mice, we replicated most of the effects seen previously in GF mice. Interestingly, the GF brain phenotype persisted in GF→CC offspring for almost all measures. In contrast, total bacterial load did not differ between the CC→CC and GF→CC groups on P7, and bacterial community composition was also very similar, with a few exceptions. Thus, GF→CC offspring had altered brain development during at least the first 7 days after birth despite a largely normal microbiota. This suggests that prenatal influences of gestating in an altered microbial environment programs neonatal brain development.
Collapse
Affiliation(s)
- Alexandra Castillo-Ruiz
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
- *Correspondence: Alexandra Castillo-Ruiz,
| | - Aviva Gars
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Hannah Sturgeon
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | | | - Dhanya N. Pyaram
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Charlène J. G. Dauriat
- INSERM U1016, Team “Mucosal Microbiota in Chronic Inflammatory Diseases,” Université Paris Cité, Paris, France
| | - Benoit Chassaing
- INSERM U1016, Team “Mucosal Microbiota in Chronic Inflammatory Diseases,” Université Paris Cité, Paris, France
| | - Nancy G. Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
3
|
Mohr MA, Michael NS, DonCarlos LL, Sisk CL. Sex differences in proliferation and attrition of pubertally born cells in the rat posterior dorsal medial amygdala. Dev Cogn Neurosci 2022; 57:101141. [PMID: 35933923 PMCID: PMC9357828 DOI: 10.1016/j.dcn.2022.101141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/08/2022] [Accepted: 07/21/2022] [Indexed: 11/26/2022] Open
Abstract
The rodent posterodorsal medial amygdala (MePD) evaluates and assigns valence to social sensory stimuli. The perception of social stimuli evolves during puberty, when the focus of social interactions shifts from kin to peers. Using the cell birthdate marker bromo-deoxyuridine (BrdU), we previously discovered that more pubertally born cells are added to the rat MePD in males than females. Here we addressed several questions that remained unanswered by our previous work. First, to determine whether there are sex differences in cell proliferation within the MePD, we examined BrdU-immunoreactive (-ir) cells at 2 and 4 h following BrdU administration on postnatal day 30 (P30). The density of BrdU-ir cells was greater in males than in females, indicating greater proliferation in males. Proliferation was substantiated by double-label immunohistochemistry showing that MePD BrdU-ir cells colocalize proliferating cell nuclear antigen, but not the cell death marker Caspase3. We next studied longer time points (2-21 days) following BrdU administration on P30 and found that the rate of cell attrition is higher in males. Finally, triple-label immunohistochemistry of P30-born MePD cells revealed that some of these cells differentiate into neurons or astrocytes within three weeks of cell birth, with no discernable sex differences. The demonstration of pubertal neuro- and glio-genesis in the MePD of male and female rats adds a new dimension to developmental plasticity of the MePD that may contribute to pubertal changes in the perception of social stimuli in both sexes.
Collapse
Affiliation(s)
- Margaret A Mohr
- Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
| | | | - Lydia L DonCarlos
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| | - Cheryl L Sisk
- Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
4
|
Zuloaga DG, Heck AL, De Guzman RM, Handa RJ. Roles for androgens in mediating the sex differences of neuroendocrine and behavioral stress responses. Biol Sex Differ 2020; 11:44. [PMID: 32727567 PMCID: PMC7388454 DOI: 10.1186/s13293-020-00319-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 07/09/2020] [Indexed: 12/17/2022] Open
Abstract
Estradiol and testosterone are powerful steroid hormones that impact brain function in numerous ways. During development, these hormones can act to program the adult brain in a male or female direction. During adulthood, gonadal steroid hormones can activate or inhibit brain regions to modulate adult functions. Sex differences in behavioral and neuroendocrine (i.e., hypothalamic pituitary adrenal (HPA) axis) responses to stress arise as a result of these organizational and activational actions. The sex differences that are present in the HPA and behavioral responses to stress are particularly important considering their role in maintaining homeostasis. Furthermore, dysregulation of these systems can underlie the sex biases in risk for complex, stress-related diseases that are found in humans. Although many studies have explored the role of estrogen and estrogen receptors in mediating sex differences in stress-related behaviors and HPA function, much less consideration has been given to the role of androgens. While circulating androgens can act by binding and activating androgen receptors, they can also act by metabolism to estrogenic molecules to impact estrogen signaling in the brain and periphery. This review focuses on androgens as an important hormone for modulating the HPA axis and behaviors throughout life and for setting up sex differences in key stress regulatory systems that could impact risk for disease in adulthood. In particular, impacts of androgens on neuropeptide systems known to play key roles in HPA and behavioral responses to stress (corticotropin-releasing factor, vasopressin, and oxytocin) are discussed. A greater knowledge of androgen action in the brain is key to understanding the neurobiology of stress in both sexes.
Collapse
Affiliation(s)
| | - Ashley L Heck
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | | | - Robert J Handa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
5
|
Lee C, Sakurai R, Shin E, Wang Y, Liu J, Rehan VK. Antenatal PPAR-γ agonist pioglitazone stimulates fetal lung maturation equally in males and females. Am J Physiol Lung Cell Mol Physiol 2020; 319:L435-L443. [PMID: 32579381 DOI: 10.1152/ajplung.00376.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Antenatal steroids (ANS) accelerate fetal lung maturation and reduce the incidence of respiratory distress syndrome. However, sex specificity, i.e., being less effective in males, and potential long-term neurodevelopmental sequelae, particularly with repeated courses, remain significant limitations. The differential sex response to ANS is likely mediated via the inhibitory effect of fetal androgens on steroid's stimulatory effect on alveolar epithelial-mesenchymal interactions. Since peroxisome proliferator-activated receptor-γ (PPAR-γ) agonists accelerate lung maturation by stimulating alveolar epithelial-mesenchymal interactions, independent of fetal sex, we hypothesized that the effect of PPAR-γ agonist pioglitazone (PGZ) would be sex-independent. Pregnant Sprague-Dawley rat dams were intraperitoneally administered dexamethasone (DEX) or PGZ on embryonic day (e) 18 and e19. At e20, pups were delivered by cesarean section, and fetal lungs and brains were examined for markers of lung maturation and apoptosis, respectively. Mixed epithelial-fibroblast cell cultures were examined to gain mechanistic insights. Antenatal PGZ increased alveolar epithelial and mesenchymal maturation markers equally in males and females; in contrast, antenatal DEX had sex-specific effects. Additionally, unlike DEX, antenatal PGZ did not increase hippocampal apoptosis. We conclude that PPAR-γ agonist administration is an effective, and probably even a superior, alternative to ANS for accelerating fetal lung maturity equally in both males and females.
Collapse
Affiliation(s)
- Cindy Lee
- Department of Pediatrics, Harbor-University of California, Los Angleles (UCLA) Medical Center, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, Torrance, California
| | - Reiko Sakurai
- Department of Pediatrics, Harbor-University of California, Los Angleles (UCLA) Medical Center, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, Torrance, California
| | - Eugene Shin
- Department of Pediatrics, Harbor-University of California, Los Angleles (UCLA) Medical Center, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, Torrance, California
| | - Ying Wang
- Department of Pediatrics, Harbor-University of California, Los Angleles (UCLA) Medical Center, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, Torrance, California
| | - Jie Liu
- Department of Pediatrics, Harbor-University of California, Los Angleles (UCLA) Medical Center, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, Torrance, California
| | - Virender K Rehan
- Department of Pediatrics, Harbor-University of California, Los Angleles (UCLA) Medical Center, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, Torrance, California
| |
Collapse
|
6
|
Does Birth Trigger Cell Death in the Developing Brain? eNeuro 2020; 7:ENEURO.0517-19.2020. [PMID: 32015098 PMCID: PMC7031855 DOI: 10.1523/eneuro.0517-19.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/27/2022] Open
Abstract
Developmental cell death eliminates half of the neurons initially generated in the mammalian brain, and occurs perinatally in many species. It is possible that the timing of neuronal cell death is developmentally programmed, and only coincidentally associated with birth. Alternatively, birth may play a role in shaping cell death. To test these competing hypotheses, we experimentally advanced or delayed birth by 1 d in mice (within the normal range of gestation for the species) and examined effects on the temporal pattern and magnitude (amount) of neuronal cell death, using immunohistochemical detection of activated caspase-3 as a cell death marker. In order to detect effects of subtle changes in birth timing, we focused on brain areas that exhibit sharp postnatal peaks in cell death. We find that advancing birth advances peak cell death, supporting the hypothesis that birth triggers cell death. However, a delay of birth does not delay cell death. Thus, birth can advance cell death, but if postponed, a developmental program governs. Advancing or delaying birth also caused region-specific changes in the overall magnitude of cell death. Our findings shed light on the long-standing question of what controls the timing and magnitude of developmental neuronal cell death, and position birth as an orchestrator of brain development. Because humans across the world now routinely alter birth timing, these findings may have implications for current obstetric practices.
Collapse
|
7
|
Petrenko A, Kanya MJ, Rosinski L, McKay ER, Bridgett DJ. Effects of infant negative affect and contextual factors on infant regulatory capacity: The moderating role of infant sex. INFANT AND CHILD DEVELOPMENT 2019. [DOI: 10.1002/icd.2157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Anton Petrenko
- Department of PsychologyNorthern Illinois University DeKalb Illinois
| | - Meghan J. Kanya
- Department of PsychologyNorthern Illinois University DeKalb Illinois
| | - Leanna Rosinski
- Department of PsychologyNorthern Illinois University DeKalb Illinois
| | - Erin R. McKay
- Department of PsychologyNorthern Illinois University DeKalb Illinois
| | - David J. Bridgett
- Department of PsychologyNorthern Illinois University DeKalb Illinois
| |
Collapse
|
8
|
Jacobs AJ, Castillo‐Ruiz A, Cisternas CD, Forger NG. Microglial Depletion Causes Region‐Specific Changes to Developmental Neuronal Cell Death in the Mouse Brain. Dev Neurobiol 2019; 79:769-779. [DOI: 10.1002/dneu.22706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/17/2019] [Accepted: 06/12/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Andrew J. Jacobs
- Neuroscience Institute Georgia State University P.O. Box 5030 Atlanta Georgia30302‐5030
| | | | - Carla D. Cisternas
- Neuroscience Institute Georgia State University P.O. Box 5030 Atlanta Georgia30302‐5030
| | - Nancy G. Forger
- Neuroscience Institute Georgia State University P.O. Box 5030 Atlanta Georgia30302‐5030
| |
Collapse
|
9
|
Kolaka R, Chotwiwatthanakun C, Chutabhakdikul N. Fetal exposure to high levels of maternal glucocorticoids alters reelin signaling in the prefrontal cortex of rat pups. Int J Dev Neurosci 2019; 78:185-190. [PMID: 31014819 DOI: 10.1016/j.ijdevneu.2019.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 04/06/2019] [Accepted: 04/10/2019] [Indexed: 10/27/2022] Open
Abstract
Maternal stress (MS) is associated with various neuropsychiatric disorders and cognitive impairment in the offspring. However, it is unclear how early life stress alters the pup's brain development and how it contributes to the pathology of neuropsychiatric disorders later in life. Reelin is a large extracellular matrix glycoprotein that plays essential roles in early brain development such as neural migration, synaptic development, and maturation. Dysregulation of reelin and its signaling proteins is associated with the emergence of neuropsychiatric disorders in adulthood. This study examined the effect of repeated maternal Carbenoxolone (CBX) injection during late gestation on reelin signaling in the prefrontal cortex (PFC) of rat pups. CBX is a selective 11β-HSD2 enzyme inhibitor that promotes the direct transfer of maternal corticosteroids (CORT) to the fetus. Therefore, treatment with CBX can mimic the animal model of early life exposure to high levels of maternal stress hormone. In this study, pregnant rats were injected daily with either saline or CBX during gestation day (GD) 14-21, and the levels of reelin and its signaling proteins were examined in the PFC of rat pups at different postnatal age from P0-P21. The main result of this study is the repeated maternal CBX injections during GD14-21 acutely increase reln mRNA and protein expression in the PFC of rat pups at birth (P0) and follow by a significant decrease during P7-P14. The treatment also causes long term decreases in the amount of VLDLR and Dab1 which are the downstream signaling proteins for the reelin pathway, at least until P21. Our results indicated that fetal exposure to high levels of maternal CORT interferes with reelin signaling which might have profound effects on cortical development associated with neuropsychiatric disorders later in life.
Collapse
Affiliation(s)
- Ratirat Kolaka
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakornpathom, Thailand
| | | | - Nuanchan Chutabhakdikul
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakornpathom, Thailand
| |
Collapse
|
10
|
Goldstein JM, Hale T, Foster SL, Tobet SA, Handa RJ. Sex differences in major depression and comorbidity of cardiometabolic disorders: impact of prenatal stress and immune exposures. Neuropsychopharmacology 2019; 44:59-70. [PMID: 30030541 PMCID: PMC6235859 DOI: 10.1038/s41386-018-0146-1] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/15/2018] [Accepted: 06/21/2018] [Indexed: 12/11/2022]
Abstract
Major depressive disorder topped ischemic heart disease as the number one cause of disability worldwide in 2012, and women have twice the risk of men. Further, the comorbidity of depression and cardiometabolic disorders will be one of the primary causes of disability worldwide by 2020, with women at twice the risk. Thus, understanding the sex-dependent comorbidities has public health consequences worldwide. We propose here that sex differences in MDD-cardiometabolic comorbidity originate, in part, from pathogenic processes initiated in fetal development that involve sex differences in shared pathophysiology between the brain, the vascular system, the CNS control of the heart and associated hormonal, immune, and metabolic physiology. Pathways implicate neurotrophic and angiogenic growth factors, gonadal hormone receptors, and neurotransmitters such as gamma amino butyric acid (GABA) on neuronal and vascular development of HPA axis regions, such as the paraventricular nucleus (PVN), in addition to blood pressure, in part through the renin-angiotensin system, and insulin and glucose metabolism. We show that the same prenatal exposures have consequences for sex differences across multiple organ systems that, in part, share common pathophysiology. Thus, we believe that applying a sex differences lens to understanding shared biologic substrates underlying these comorbidities will provide novel insights into the development of sex-dependent therapeutics. Further, taking a lifespan perspective beginning in fetal development provides the opportunity to target abnormalities early in the natural history of these disorders in a sex-dependent way.
Collapse
Affiliation(s)
- Jill M Goldstein
- Departments of Psychiatry and Obstetrics and Gynecology, Massachusetts General Hospital (MGH), Boston, MA, 02120, USA.
- Departments of Psychiatry and Medicine, Harvard Medical School, Boston, MA, USA.
| | - Taben Hale
- Department of Basic Medical Science, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, 85004, USA
| | - Simmie L Foster
- Department of Psychiatry, Harvard Medical School, at Massachusetts General Hospital, Boston, MA, USA
| | - Stuart A Tobet
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
| | - Robert J Handa
- Department of Basic Medical Science, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, 85004, USA
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| |
Collapse
|
11
|
Abul M, Al-Bader MD, Mouihate A. Exposure to synthetic glucocorticoids during pregnancy alters the expression of p73 gene variants in fetal brains in a sex-specific manner. Brain Res 2018; 1707:117-123. [PMID: 30476470 DOI: 10.1016/j.brainres.2018.11.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 11/22/2018] [Accepted: 11/23/2018] [Indexed: 12/31/2022]
Abstract
Fetal exposure to dexamethasone (DEX) alters brain plasticity and cognitive functions during adulthood in a sex-dependent manner. The mechanisms underlying such long-lasting sex-dependent change of prenatal DEX is not well understood. The p73 gene plays an important role in brain development. It encodes for two protein variants; the neural cell death protein (TAp73) and the anti-neural cell death protein (ΔNp73). Therefore, we sought to determine how prenatal exposure to DEX alters the expression of these p73 gene variants in the brain of male and female fetuses. Pregnant dams received daily injections of either DEX (0.4 mg/kg, i.p.) or saline from gestation day (GD) 14 until GD21. On GD21, body and brain weights were monitored and mRNA and protein levels of TAp73 and ΔNp73 were measured in male and female fetal brains using RT-PCR, Western blot, and immunohistochemistry. Prenatal exposure to DEX significantly reduced the body and brain weights of both male and female fetuses, although reduction in brain weight was less severe than that of the body weight. Administration of DEX to pregnant dams led to enhanced expression of both TAp73 and ΔNp73 gene/protein variants in the brain of male but not in that of female fetuses. Dexamethasone induced a sex-dependent effect on the expression of p73 gene variants. DEX-induced growth restriction in the brain of female fetuses is independent of p73 gene. This study strongly suggests that survival/death programs operate differently during the development of male and female brains.
Collapse
Affiliation(s)
- Mai Abul
- Department of Physiology, Health Sciences Centre, Faculty of Medicine, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait
| | - Maie D Al-Bader
- Department of Physiology, Health Sciences Centre, Faculty of Medicine, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait
| | - Abdeslam Mouihate
- Department of Physiology, Health Sciences Centre, Faculty of Medicine, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait.
| |
Collapse
|
12
|
Birth delivery mode alters perinatal cell death in the mouse brain. Proc Natl Acad Sci U S A 2018; 115:11826-11831. [PMID: 30322936 DOI: 10.1073/pnas.1811962115] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Labor and a vaginal delivery trigger changes in peripheral organs that prepare the mammalian fetus to survive ex utero. Surprisingly little attention has been given to whether birth also influences the brain, and to how alterations in birth mode affect neonatal brain development. These are important questions, given the high rates of cesarean section (C-section) delivery worldwide, many of which are elective. We examined the effect of birth mode on neuronal cell death, a widespread developmental process that occurs primarily during the first postnatal week in mice. Timed-pregnant dams were randomly assigned to C-section deliveries that were yoked to vaginal births to carefully match gestation length and circadian time of parturition. Compared with rates of cell death just before birth, vaginally-born offspring had an abrupt, transient decrease in cell death in many brain regions, suggesting that a vaginal delivery is neuroprotective. In contrast, cell death was either unchanged or increased in C-section-born mice. Effects of delivery mode on cell death were greatest for the paraventricular nucleus of the hypothalamus (PVN), which is central to the stress response and brain-immune interactions. The greater cell death in the PVN of C-section-delivered newborns was associated with a reduction in the number of PVN neurons expressing vasopressin at weaning. C-section-delivered mice also showed altered vocalizations in a maternal separation test and greater body mass at weaning. Our results suggest that vaginal birth acutely impacts brain development, and that alterations in birth mode may have lasting consequences.
Collapse
|
13
|
Jie F, Yin G, Yang W, Yang M, Gao S, Lv J, Li B. Stress in Regulation of GABA Amygdala System and Relevance to Neuropsychiatric Diseases. Front Neurosci 2018; 12:562. [PMID: 30154693 PMCID: PMC6103381 DOI: 10.3389/fnins.2018.00562] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 07/25/2018] [Indexed: 01/19/2023] Open
Abstract
The amygdala is an almond-shaped nucleus located deep and medially within the temporal lobe and is thought to play a crucial role in the regulation of emotional processes. GABAergic neurotransmission inhibits the amygdala and prevents us from generating inappropriate emotional and behavioral responses. Stress may cause the reduction of the GABAergic interneuronal network and the development of neuropsychological diseases. In this review, we summarize the recent evidence investigating the possible mechanisms underlying GABAergic control of the amygdala and its interaction with acute and chronic stress. Taken together, this study may contribute to future progress in finding new approaches to reverse the attenuation of GABAergic neurotransmission induced by stress in the amygdala.
Collapse
Affiliation(s)
- Fan Jie
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Guanghao Yin
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Modi Yang
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shuohui Gao
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiayin Lv
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Jacobskind JS, Rosinger ZJ, Gonzalez T, Zuloaga KL, Zuloaga DG. Chronic Methamphetamine Exposure Attenuates Neural Activation in Hypothalamic-Pituitary-Adrenal Axis-Associated Brain Regions in a Sex-specific Manner. Neuroscience 2018; 380:132-145. [PMID: 29679646 DOI: 10.1016/j.neuroscience.2018.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/16/2018] [Accepted: 04/09/2018] [Indexed: 12/15/2022]
Abstract
Sex differences in methamphetamine (MA) abuse and consequences of MA have been reported with females showing an increased addiction phenotype and withdrawal symptoms. One mechanism through which these effects might occur is via sex-specific alterations in the hypothalamic-pituitary-adrenal (HPA) axis and its associated brain regions. In this study, mice were administered MA (5 mg/kg) or saline for 10 consecutive days. During early withdrawal, anxiety-like behaviors were assessed in the open field, light/dark box, and elevated plus maze. At ten days of withdrawal, mice were injected with a final dose of MA (5 mg/kg) or saline. Chronic MA did not alter anxiety-like behaviors or corticosterone responses to a final dose of MA, although females showed elevated corticosterone responses compared to males. Chronic MA attenuated final MA-induced c-Fos in both sexes in the paraventricular hypothalamus (PVH), bed nucleus of the stria terminalis (BNST), cingulate cortex, central and basolateral amygdala. In CA1 and CA3 hippocampal areas, c-Fos attenuation by chronic MA occurred only in females. Within the PVH, final MA injection increased c-Fos to a greater extent in females compared to males regardless of prior MA exposure. Dual-labeling of c-Fos with glucocorticoid receptor revealed a specific attenuation of neural activation within this cell type in the PVH, central and basolateral amygdala, and BNST. Together these findings demonstrate that chronic MA can suppress subsequent activation of HPA axis-associated brain regions and cell phenotypes. Further, in select regions this reduction is sex-specific. These changes may contribute to reported sex differences in MA abuse patterns.
Collapse
Affiliation(s)
- Jason S Jacobskind
- University at Albany, Department of Psychology, Albany, NY 12222, United States
| | - Zachary J Rosinger
- University at Albany, Department of Psychology, Albany, NY 12222, United States
| | - Tiffany Gonzalez
- University at Albany, Department of Psychology, Albany, NY 12222, United States
| | - Kristen L Zuloaga
- Albany Medical College, Department of Neuroscience & Experimental Therapeutics, Albany, NY 12208, United States
| | - Damian G Zuloaga
- University at Albany, Department of Psychology, Albany, NY 12222, United States.
| |
Collapse
|
15
|
The microbiota influences cell death and microglial colonization in the perinatal mouse brain. Brain Behav Immun 2018; 67:218-229. [PMID: 28890156 PMCID: PMC5696094 DOI: 10.1016/j.bbi.2017.08.027] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/18/2017] [Accepted: 08/23/2017] [Indexed: 12/21/2022] Open
Abstract
The mammalian fetus develops in a largely sterile environment, and direct exposure to a complex microbiota does not occur until birth. We took advantage of this to examine the effect of the microbiota on brain development during the first few days of life. The expression of anti- and pro-inflammatory cytokines, developmental cell death, and microglial colonization in the brain were compared between newborn conventionally colonized mice and mice born in sterile, germ-free (GF) conditions. Expression of the pro-inflammatory cytokines interleukin 1β and tumor necrosis factor α was markedly suppressed in GF newborns. GF mice also had altered cell death, with some regions exhibiting higher rates (paraventricular nucleus of the hypothalamus and the CA1 oriens layer of the hippocampus) and other regions exhibiting no change or lower rates (arcuate nucleus of the hypothalamus) of cell death. Microglial labeling was elevated in GF mice, due to an increase in both microglial cell size and number. The changes in cytokine expression, cell death and microglial labeling were evident on the day of birth, but were absent on embryonic day 18.5, approximately one-half day prior to expected delivery. Taken together, our results suggest that direct exposure to the microbiota at birth influences key neurodevelopmental events and does so within hours. These findings may help to explain some of the behavioral and neurochemical alterations previously seen in adult GF mice.
Collapse
|
16
|
Abdulbasit A, Stephen Michael F, Shukurat Onaopemipo A, Abdulmusawwir AO, Aminu I, Nnaemeka Tobechukwu A, Wahab Imam A, Oluwaseun Aremu A, Folajimi O, Bilikis Aderonke A, Ridwan Babatunde I, Victor Bamidele O. Glucocorticoid receptor activation selectively influence performance of Wistar rats in Y-maze. ACTA ACUST UNITED AC 2017; 25:41-50. [PMID: 29274871 DOI: 10.1016/j.pathophys.2017.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 01/09/2023]
Abstract
Glucocorticoid receptors (GR) are ubiquitously expressed in metazoans. Different and contrasting phenotypes have been reported upon their activation. This study investigated the behavioral phenotypes characteristic of GR stimulation in male Wistar rats. Rats in each of the four groups of rats received one of the following treatments: distilled water (control) or one of three doses of dexamethasone (treatment) injected intraperitoneally for 7 days. The Rats were afterwards subjected to the Y maze, the elevated plus maze (EPM), the Morris water maze (MWM), and the novel object recognition (NOR) test. At the end of the study, the animals were anesthetized and neural activity from the prefrontal cortex recorded. Blood was collected via cardiac puncture to evaluate the levels of plasma insulin and glucose, and the prefrontal cortexes excised to determine the levels of insulin, markers of oxidative stress, and calcium in the homogenate. This study showed that treatment with dexamethasone significantly reduced the total and percentage alternation in the Y maze, but had no significant effect on object recognition in the NOR test, long-term and short-term spatial memory in the MWM, or anxiety-like behavior in the EPM. Plasma and brain insulin and calcium levels were elevated moderately following treatment with the lowest dose of dexamethasone. All doses of dexamethasone decreased brain superoxide dismutase and increased lactate dehydrogenase levels. No significant change in neural activity was observed. This study shows that activation of glucocorticoid receptors differentially affects different behavioral paradigms and provides evidence for a role for glucocorticoids in mediating insulin function in the brain.
Collapse
Affiliation(s)
- Amin Abdulbasit
- Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Nigeria.
| | - Fii Stephen Michael
- Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Nigeria
| | | | | | - Imam Aminu
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Ilorin, Nigeria
| | | | - Abdulmajeed Wahab Imam
- Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Nigeria
| | - Adeyanju Oluwaseun Aremu
- Department of Physiology, College of Medicine, Afe-Babalola University Ado-Ekiti, Ekiti, Nigeria
| | - Olaseinde Folajimi
- Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Nigeria
| | | | | | | |
Collapse
|
17
|
The role of IL-6 in neurodevelopment after prenatal stress. Brain Behav Immun 2017; 65:274-283. [PMID: 28546058 PMCID: PMC5537020 DOI: 10.1016/j.bbi.2017.05.015] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 05/03/2017] [Accepted: 05/20/2017] [Indexed: 12/13/2022] Open
Abstract
Prenatal stress exposure is associated with adverse psychiatric outcomes, including autism and ADHD, as well as locomotor and social inhibition and anxiety-like behaviors in animal offspring. Similarly, maternal immune activation also contributes to psychiatric risk and aberrant offspring behavior. The mechanisms underlying these outcomes are not clear. Offspring microglia and the pro-inflammatory cytokine interleukin-6 (IL-6), known to influence microglia, may serve as common mechanisms between prenatal stress and prenatal immune activation. To evaluate the role of prenatal IL-6 in prenatal stress, microglia morphological analyses were conducted at embryonic days 14 (E14), E15, and in adult mice. Offspring microglia and behavior were evaluated after repetitive maternal restraint stress, repetitive maternal IL-6, or maternal IL-6 blockade during stress from E12 onwards. At E14, novel changes in cortical plate embryonic microglia were documented-a greater density of the mutivacuolated morphology. This resulted from either prenatal stress or IL-6 exposure and was prevented by IL-6 blockade during prenatal stress. Prenatal stress also resulted in increased microglia ramification in adult brain, as has been previously shown. As with embryonic microglia, prenatal IL-6 recapitulated prenatal stress-induced changes in adult microglia. Furthermore, prenatal IL-6 was able to recapitulate the delay in GABAergic progenitor migration caused by prenatal stress. However, IL-6 mechanisms were not necessary for this delay, which persisted after prenatal stress despite IL-6 blockade. As we have previously demonstrated, behavioral effects of prenatal stress in offspring, including increased anxiety-like behavior, decreased sociability, and locomotor inhibition, may be related to these GABAergic delays. While adult microglia changes were ameliorated by IL-6 blockade, these behavioral changes were independent of IL-6 mechanisms, similar to GABAergic delays. This and previous work from our laboratory suggests that multiple mechanisms, including GABAergic delays, may underlie prenatal stress-linked deficits.
Collapse
|
18
|
Jacobskind JS, Rosinger ZJ, Zuloaga DG. Hypothalamic-pituitary-adrenal axis responsiveness to methamphetamine is modulated by gonadectomy in males. Brain Res 2017; 1677:74-85. [PMID: 28941573 DOI: 10.1016/j.brainres.2017.09.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/18/2017] [Accepted: 09/17/2017] [Indexed: 12/14/2022]
Abstract
Sex differences in patterns of methamphetamine (MA) abuse have been reported with females (humans and rodents) showing an elevated addiction phenotype. Previous findings indicate MA-induced hypothalamic-pituitary-adrenal (HPA) axis activation is also sexually dimorphic with females exhibiting an elevated glucocorticoid release and differential neural activation patterns within HPA axis-associated brain regions. These effects may contribute to sex differences in abuse. To determine the role of gonadal hormones in mediating sex differences in MA-induced glucocorticoids, male and female C57BL/6J mice were gonadectomized or sham-operated, and following recovery, injected with MA (5mg/kg) and sacrificed 60min or 120min later. Blood was collected for corticosterone radioimmunoassay, and brains were used to assess c-Fos, and c-Fos co-localization with glucocorticoid receptor (GR). At 120min after MA injection, corticosterone levels were elevated in females compared to males and gonadectomy in males increased corticosterone to female levels. C-Fos was greater in females than males in the medial preoptic area, bed nucleus of the stria terminalis, basolateral amygdala, and central amygdala. Female gonadectomy had little effect on either corticosterone or c-Fos, while male gonadectomy elevated c-Fos in the central amygdala. Relative to sham males, gonadectomized males also showed decreased c-Fos/GR cell number in the CA3 hippocampal area compared to sham males, indicating a central site for attenuated negative feedback. Together, these findings indicate that androgens regulate MA-induced activation of the HPA axis, potentially by enhancing negative feedback. These sex and gonadal hormone effects on the HPA axis may contribute to sex differences in MA abuse patterns.
Collapse
Affiliation(s)
- Jason S Jacobskind
- University at Albany, Department of Psychology, Albany, NY 12222, United States
| | - Zachary J Rosinger
- University at Albany, Department of Psychology, Albany, NY 12222, United States
| | - Damian G Zuloaga
- University at Albany, Department of Psychology, Albany, NY 12222, United States.
| |
Collapse
|
19
|
Nolvi S, Pesonen H, Bridgett DJ, Korja R, Kataja EL, Karlsson H, Karlsson L. Infant Sex Moderates the Effects of Maternal Pre- and Postnatal Stress on Executive Functioning at 8 Months of Age. INFANCY 2017. [DOI: 10.1111/infa.12206] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Saara Nolvi
- The FinnBrain Birth Cohort Study; Turku Brain and Mind Center; Institute of Clinical Medicine; University of Turku
| | - Henri Pesonen
- The FinnBrain Birth Cohort Study; Turku Brain and Mind Center; Institute of Clinical Medicine; University of Turku
- Department of Mathematics and Statistics; University of Turku
| | | | - Riikka Korja
- The FinnBrain Birth Cohort Study; Turku Brain and Mind Center; Institute of Clinical Medicine; University of Turku
- Department of Psychology; University of Turku
| | - Eeva-Leena Kataja
- The FinnBrain Birth Cohort Study; Turku Brain and Mind Center; Institute of Clinical Medicine; University of Turku
- Department of Psychology; University of Turku
| | - Hasse Karlsson
- The FinnBrain Birth Cohort Study; Turku Brain and Mind Center; Institute of Clinical Medicine; University of Turku
- Department of Psychiatry; Turku University Hospital and University of Turku
| | - Linnea Karlsson
- The FinnBrain Birth Cohort Study; Turku Brain and Mind Center; Institute of Clinical Medicine; University of Turku
- Department of Child Psychiatry; Turku University Hospital and University of Turku
| |
Collapse
|
20
|
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: 345] [Impact Index Per Article: 43.1] [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
|
21
|
Menshanov PN, Bannova AV, Dygalo NN. Anoxia ameliorates the dexamethasone-induced neurobehavioral alterations in the neonatal male rat pups. Horm Behav 2017; 87:122-128. [PMID: 27865789 DOI: 10.1016/j.yhbeh.2016.11.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/11/2016] [Accepted: 11/14/2016] [Indexed: 01/03/2023]
Abstract
Glucocorticoids and hypoxia are two essential factors affecting the brain development during labor and delivery. In addition to the neurobehavioral alterations induced separately by these factors, glucocorticoids can attenuate the deleterious consequences of severe hypoxia-ischemia on the brain development, acting as a neuroprotective agent in combination with hypoxia. The role of hypoxia in the combined action with corticosteroids is less clear. Severe hypoxia-ischemia results in the massive activation of caspase-3, masking any other effects of hypoxia on the neonatal brain exposed to glucocorticoids. As a result, the effects of mild hypoxia on the developing brain pretreated with glucocorticoids remain unclear. To analyze this problem, 2-day-old male rats were treated with dexamethasone (DEX) before the subsequent exposure to mild 10-min anoxia or normoxia. The treatment with only DEX resulted in the delay in the development of the negative geotaxis reaction and in the decrease in locomotor activity of the neonatal male pups. The mild anoxic event attenuated these DEX-induced neurobehavioral alterations. The treatment with DEX, but not the mild anoxic exposure alone, resulted in the delayed upregulation of active caspase-3 in the prefrontal cortex and in the brainstem of the male pups. This glucocorticoid-induced upregulation of active caspase-3 was prevented by the anoxic event. The present findings evidence that mild anoxia is capable of ameliorating the glucocorticoid-induced neurodevelopmental alterations in the neonatal rats if the artificial or the naturally occurring increase in the levels of glucocorticoids occurred just before the episode of hypoxia.
Collapse
Affiliation(s)
- Petr N Menshanov
- Functional Neurogenomics Laboratory, Institute of Cytology and Genetics, Russian Academy of Science, Novosibirsk, Russian Federation; Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russian Federation.
| | - Anita V Bannova
- Functional Neurogenomics Laboratory, Institute of Cytology and Genetics, Russian Academy of Science, Novosibirsk, Russian Federation
| | - Nikolay N Dygalo
- Functional Neurogenomics Laboratory, Institute of Cytology and Genetics, Russian Academy of Science, Novosibirsk, Russian Federation; Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russian Federation
| |
Collapse
|
22
|
Mosley M, Shah C, Morse KA, Miloro SA, Holmes MM, Ahern TH, Forger NG. Patterns of cell death in the perinatal mouse forebrain. J Comp Neurol 2017; 525:47-64. [PMID: 27199256 PMCID: PMC5116296 DOI: 10.1002/cne.24041] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 12/15/2022]
Abstract
The importance of cell death in brain development has long been appreciated, but many basic questions remain, such as what initiates or terminates the cell death period. One obstacle has been the lack of quantitative data defining exactly when cell death occurs. We recently created a "cell death atlas," using the detection of activated caspase-3 (AC3) to quantify apoptosis in the postnatal mouse ventral forebrain and hypothalamus, and found that the highest rates of cell death were seen at the earliest postnatal ages in most regions. Here we have extended these analyses to prenatal ages and additional brain regions. We quantified cell death in 16 forebrain regions across nine perinatal ages from embryonic day (E) 17 to postnatal day (P) 11 and found that cell death peaks just after birth in most regions. We found greater cell death in several regions in offspring delivered vaginally on the day of parturition compared with those of the same postconception age but still in utero at the time of collection. We also found massive cell death in the oriens layer of the hippocampus on P1 and in regions surrounding the anterior crossing of the corpus callosum on E18 as well as the persistence of large numbers of cells in those regions in adult mice lacking the pro-death Bax gene. Together these findings suggest that birth may be an important trigger of neuronal cell death and identify transient cell groups that may undergo wholesale elimination perinatally. J. Comp. Neurol. 525:47-64, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Morgan Mosley
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, 30302
| | - Charisma Shah
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, 30302
| | - Kiriana A Morse
- Department of Psychology, Center for Behavioral Neuroscience, Quinnipiac University, Hamden, Connecticut, 06518
| | - Stephen A Miloro
- Department of Psychology, Center for Behavioral Neuroscience, Quinnipiac University, Hamden, Connecticut, 06518
| | - Melissa M Holmes
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, L5L 1C6, Canada
| | - Todd H Ahern
- Department of Psychology, Center for Behavioral Neuroscience, Quinnipiac University, Hamden, Connecticut, 06518
| | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, 30302
| |
Collapse
|
23
|
Strahan JA, Walker WH, Montgomery TR, Forger NG. Minocycline causes widespread cell death and increases microglial labeling in the neonatal mouse brain. Dev Neurobiol 2016; 77:753-766. [PMID: 27706925 DOI: 10.1002/dneu.22457] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 09/09/2016] [Accepted: 09/28/2016] [Indexed: 11/09/2022]
Abstract
Minocycline, an antibiotic of the tetracycline family, inhibits microglia in many paradigms and is among the most commonly used tools for examining the role of microglia in physiological processes. Microglia may play an active role in triggering developmental neuronal cell death, although findings have been contradictory. To determine whether microglia influence developmental cell death, we treated perinatal mice with minocycline (45 mg/kg) and quantified effects on dying cells and microglial labeling using immunohistochemistry for activated caspase-3 (AC3) and ionized calcium-binding adapter molecule 1 (Iba1), respectively. Contrary to our expectations, minocycline treatment from embryonic day 18 to postnatal day (P)1 caused a > tenfold increase in cell death 8 h after the last injection in all brain regions examined, including the primary sensory cortex, septum, hippocampus and hypothalamus. Iba1 labeling was also increased in most regions. Similar effects, although of smaller magnitude, were seen when treatment was delayed to P3-P5. Minocycline treatment from P3 to P5 also decreased overall cell number in the septum at weaning, suggesting lasting effects of the neonatal exposure. When administered at lower doses (4.5 or 22.5 mg/kg), or at the same dose 1 week later (P10-P12), minocycline no longer increased microglial markers or cell death. Taken together, the most commonly used microglial "inhibitor" increases cell death and Iba1 labeling in the neonatal mouse brain. Minocycline is used clinically in infant and pediatric populations; caution is warrented when using minocycline in developing animals, or extrapolating the effects of this drug across ages. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 753-766, 2017.
Collapse
Affiliation(s)
- J Alex Strahan
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia, 30302
| | - William H Walker
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia, 30302
| | - Taylor R Montgomery
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia, 30302
| | - Nancy G Forger
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia, 30302
| |
Collapse
|
24
|
Hiroi R, Carbone DL, Zuloaga DG, Bimonte-Nelson HA, Handa RJ. Sex-dependent programming effects of prenatal glucocorticoid treatment on the developing serotonin system and stress-related behaviors in adulthood. Neuroscience 2016; 320:43-56. [PMID: 26844389 PMCID: PMC4840233 DOI: 10.1016/j.neuroscience.2016.01.055] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 01/12/2016] [Accepted: 01/26/2016] [Indexed: 12/25/2022]
Abstract
Prenatal stress and overexposure to glucocorticoids (GC) during development may be associated with an increased susceptibility to a number of diseases in adulthood including neuropsychiatric disorders, such as depression and anxiety. In animal models, prenatal overexposure to GC results in hyper-responsiveness to stress in adulthood, and females appear to be more susceptible than males. Here, we tested the hypothesis that overexposure to GC during fetal development has sex-specific programming effects on the brain, resulting in altered behaviors in adulthood. We examined the effects of dexamethasone (DEX; a synthetic GC) during prenatal life on stress-related behaviors in adulthood and on the tryptophan hydroxylase-2 (TpH2) gene expression in the adult dorsal raphe nucleus (DRN). TpH2 is the rate-limiting enzyme for serotonin (5-HT) synthesis and has been implicated in the etiology of human affective disorders. Timed-pregnant rats were treated with DEX from gestational days 18-22. Male and female offspring were sacrificed on the day of birth (postnatal day 0; P0), P7, and in adulthood (P80-84) and brains were examined for changes in TpH2 mRNA expression. Adult animals were also tested for anxiety- and depressive- like behaviors. In adulthood, prenatal DEX increased anxiety- and depressive- like behaviors selectively in females, as measured by decreased time spent in the center of the open field and increased time spent immobile in the forced swim test, respectively. Prenatal DEX increased TpH2 mRNA selectively in the female caudal DRN at P7, whereas it decreased TpH2 mRNA selectively in the female caudal DRN in adulthood. In animals challenged with restraint stress in adulthood, TpH2 mRNA was significantly lower in rostral DRN of prenatal DEX-treated females compared to vehicle-treated females. These data demonstrated that prenatal overexposure to GC alters the development of TpH2 gene expression and these alterations correlated with lasting behavioral changes found in adult female offspring.
Collapse
Affiliation(s)
- R Hiroi
- Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, 550 E. Van Buren Street, Phoenix, AZ 85004, USA; Department of Psychology, Arizona State University, 950 S. McAllister Avenue, Tempe, AZ 85287, USA.
| | - D L Carbone
- Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, 550 E. Van Buren Street, Phoenix, AZ 85004, USA.
| | - D G Zuloaga
- Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, 550 E. Van Buren Street, Phoenix, AZ 85004, USA.
| | - H A Bimonte-Nelson
- Department of Psychology, Arizona State University, 950 S. McAllister Avenue, Tempe, AZ 85287, USA.
| | - R J Handa
- Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, 550 E. Van Buren Street, Phoenix, AZ 85004, USA.
| |
Collapse
|
25
|
Cartier J, Zeng Y, Drake AJ. Glucocorticoids and the prenatal programming of neurodevelopmental disorders. Curr Opin Behav Sci 2016. [DOI: 10.1016/j.cobeha.2015.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
26
|
Virdee K, Kentrop J, Jupp B, Venus B, Hensman D, McArthur S, Wilkinson J, Robbins TW, Gillies G, Dalley JW. Counteractive effects of antenatal glucocorticoid treatment on D1 receptor modulation of spatial working memory. Psychopharmacology (Berl) 2016; 233:3751-3761. [PMID: 27553822 PMCID: PMC5063912 DOI: 10.1007/s00213-016-4405-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/08/2016] [Indexed: 12/11/2022]
Abstract
RATIONALE Antenatal exposure to the glucocorticoid dexamethasone dramatically increases the number of mesencephalic dopaminergic neurons in rat offspring. However, the consequences of this expansion in midbrain dopamine (DA) neurons for behavioural processes in adulthood are poorly understood, including working memory that depends on DA transmission in the prefrontal cortex (PFC). OBJECTIVES We therefore investigated the influence of antenatal glucocorticoid treatment (AGT) on the modulation of spatial working memory by a D1 receptor agonist and on D1 receptor binding and DA content in the PFC and striatum. METHODS Pregnant rats received AGT on gestational days 16-19 by adding dexamethasone to their drinking water. Male offspring reared to adulthood were trained on a delayed alternation spatial working memory task and administered the partial D1 agonist SKF38393 (0.3-3 mg/kg) by systemic injection. In separate groups of control and AGT animals, D1 receptor binding and DA content were measured post-mortem in the PFC and striatum. RESULTS SKF38393 impaired spatial working memory performance in control rats but had no effect in AGT rats. D1 binding was significantly reduced in the anterior cingulate cortex, prelimbic cortex, dorsal striatum and ventral pallidum of AGT rats compared with control animals. However, AGT had no significant effect on brain monoamine levels. CONCLUSIONS These findings demonstrate that D1 receptors in corticostriatal circuitry down-regulate in response to AGT. This compensatory effect in D1 receptors may result from increased DA-ergic tone in AGT rats and underlie the resilience of these animals to the disruptive effects of D1 receptor activation on spatial working memory.
Collapse
Affiliation(s)
- Kanwar Virdee
- Department of Psychology, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK ,Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK
| | - Jiska Kentrop
- Department of Psychology, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK ,Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK
| | - Bianca Jupp
- Department of Psychology, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK ,Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK
| | - Bethany Venus
- Department of Psychology, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK
| | - Daniel Hensman
- Department of Psychology, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK
| | - Simon McArthur
- Department of Biomedical Sciences, University of Westminster, New Cavendish Street, London, W1W 6UW UK
| | - James Wilkinson
- Department of Psychology, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK ,Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK
| | - Trevor W. Robbins
- Department of Psychology, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK ,Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK
| | - Glenda Gillies
- Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Jeffrey W. Dalley
- Department of Psychology, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK ,Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK ,Department of Psychiatry, University of Cambridge, Cambridge, CB2 2QQ UK
| |
Collapse
|
27
|
Jia H, François F, Bourien J, Eybalin M, Lloyd RV, Van De Water TR, Puel JL, Venail F. Prevention of trauma-induced cochlear fibrosis using intracochlear application of anti-inflammatory and antiproliferative drugs. Neuroscience 2015; 316:261-78. [PMID: 26718602 DOI: 10.1016/j.neuroscience.2015.12.031] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 12/14/2015] [Accepted: 12/16/2015] [Indexed: 12/20/2022]
Abstract
Cochlear fibrosis is a common finding following cochlear implantation. Evidence suggests that cochlear fibrosis could be triggered by inflammation and epithelial-to-mesenchymal cell transition (EMT). In this study, we investigate the mechanisms of cochlear fibrosis and the risk/benefit ratio of local administration of the anti-inflammatory drug dexamethasone (DEX) and antimitotic drug aracytine (Ara-C). Cochlear fibrosis was evaluated in cochlear fibrosis models of rat cochlear slices in vitro and in KLH-induced immune labyrinthitis and platinum wire cochlear implantation-induced fibrosis in vivo. Cochleae were invaded with tissue containing fibroblastic cells expressing α-SMA (alpha smooth muscle actin), which along with collagen I, fibronectin, and laminin in the extracellular matrix, suggests the involvement of a fibrotic process triggered by EMT in vitro and in vivo. After perilymphatic injection of an adenoviral vector expressing GFP in vivo, we demonstrated that the fibroblastic cells derived from the mesothelial cells of the scalae tympani and vestibuli. Activation of inflammatory and EMT pathways was further assessed by ELISA analysis of the expression of IL-1β and TGF-β1. Both markers were elevated in vitro and in vivo, and DEX and Ara-C were able to reduce IL-1β and TGF-β1 production. After 5days of culture in vitro, quantification of calcein-positive cells revealed that Ara-C was 30-fold more efficient in preventing fibrosis, and provoked less sensory hair cell loss, than DEX. In KLH-induced immune labyrinthitis and platinum wire-implanted models, Ara-C was more efficient in preventing proliferation of fibrosis with less side effects on hair cells and neurons than DEX. In conclusion, DEX and Ara-C both prevent fibrosis in the cochlea. Analysis of the risk/benefit ratio favors the use of Ara-C for preventing cochlear fibrosis.
Collapse
Affiliation(s)
- H Jia
- INSERM - UMR 1051, Institute for Neurosciences of Montpellier, Montpellier, France; University Montpellier 1 & 2, Montpellier, France; Department of ORL H&N Surgery, Xinhua Hospital - Ear Institute, Shanghai Jiaotong University School of Medicine, China.
| | - F François
- INSERM - UMR 1051, Institute for Neurosciences of Montpellier, Montpellier, France; University Montpellier 1 & 2, Montpellier, France.
| | - J Bourien
- INSERM - UMR 1051, Institute for Neurosciences of Montpellier, Montpellier, France; University Montpellier 1 & 2, Montpellier, France.
| | - M Eybalin
- INSERM - UMR 1051, Institute for Neurosciences of Montpellier, Montpellier, France; University Montpellier 1 & 2, Montpellier, France.
| | - R V Lloyd
- ENT Department, The Tunbridge Wells Hospital, Tunbridge Wells, UK.
| | - T R Van De Water
- Department of Otolaryngology, University of Miami Ear Institute, Miami, FL, USA.
| | - J-L Puel
- INSERM - UMR 1051, Institute for Neurosciences of Montpellier, Montpellier, France; University Montpellier 1 & 2, Montpellier, France.
| | - F Venail
- INSERM - UMR 1051, Institute for Neurosciences of Montpellier, Montpellier, France; University Montpellier 1 & 2, Montpellier, France; ENT Department, University Hospital Gui de Chauliac, Montpellier, France.
| |
Collapse
|
28
|
Kemp MW, Newnham JP, Challis JG, Jobe AH, Stock SJ. The clinical use of corticosteroids in pregnancy. Hum Reprod Update 2015; 22:240-59. [PMID: 26590298 DOI: 10.1093/humupd/dmv047] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 10/13/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The use of antenatal steroid therapy is common in pregnancy. In early pregnancy, steroids may be used in women for the treatment of recurrent miscarriage or fetal abnormalities such as congenital adrenal hyperplasia. In mid-late pregnancy, the antenatal administration of corticosteroids to expectant mothers in anticipation of preterm birth is one of the most important advances in perinatal medicine; antenatal corticosteroids are now standard care for pregnancies at risk of premature delivery in high- and middle-income countries. The widespread uptake of this therapy is due to a compelling body of evidence demonstrating improved neonatal outcomes following antenatal corticosteroid exposure, stemming most notably from corticosteroid-driven maturation of fetal pulmonary function. As we approach the 50th anniversary of landmark work in this area by Liggins and Howie, it is apparent that much remains to be understood with regards to how we might best apply antenatal corticosteroid therapy to improve pregnancy outcomes at both early and mid to late gestation. METHODS Drawing on advances in laboratory science, pre-clinical and clinical studies, we performed a narrative review of the scientific literature to provide a timely update on the benefits, risks and uncertainties regarding antenatal corticosteroid use in pregnancy. Three, well-established therapeutic uses of antenatal steroids, namely recurrent miscarriage, congenital adrenal hyperplasia and preterm birth, were selected to frame the review. RESULTS Even the most well-established antenatal steroid therapies lack the comprehensive pharmacokinetic and dose-response data necessary to optimize dosing regimens. New insights into complex, tissue-specific corticosteroid signalling by genomic-dependent and independent mechanisms have not been used to inform corticosteroid treatment strategies. There is growing evidence that some fetal corticosteroid treatments are either ineffective, or may result in adverse outcomes, in addition to lasting epigenetic changes in a variety of homeostatic mechanisms. Nowhere is the need to better understand the intricacies of corticosteroid therapy better conveyed than in the findings of Althabe and colleagues who recently reported an increase in overall neonatal mortality and maternal morbidity in association with antenatal corticosteroid administration in low-resource settings. CONCLUSIONS New research to clarify the benefits and potential risks of antenatal corticosteroid therapy is urgently needed, especially with regard to corticosteroid use in low-resource environments. We conclude that there is both significant scope and an urgent need for further research-informed refinement to the use of antenatal corticosteroids in pregnancy.
Collapse
Affiliation(s)
- M W Kemp
- School of Women's and Infants' Health, The University of Western Australia, Perth, Western Australia, Australia
| | - J P Newnham
- School of Women's and Infants' Health, The University of Western Australia, Perth, Western Australia, Australia
| | - J G Challis
- Office of the Pro Vice-Chancellor (Health and Medical Research), The University of Western Australia, Perth, Western Australia, Australia
| | - A H Jobe
- School of Women's and Infants' Health, The University of Western Australia, Perth, Western Australia, Australia Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Centre, Cincinnati, OH, USA
| | - S J Stock
- Tommy's Centre for Maternal and Fetal Health, MRC Centre for Reproductive Health, Queen's Medical Research Institute, Edinburgh, UK
| |
Collapse
|
29
|
Goldstein JM, Lancaster K, Longenecker JM, Abbs B, Holsen LM, Cherkerzian S, Whitfield-Gabrieli S, Makris N, Tsuang MT, Buka SL, Seidman LJ, Klibanski A. Sex differences, hormones, and fMRI stress response circuitry deficits in psychoses. Psychiatry Res 2015; 232:226-36. [PMID: 25914141 PMCID: PMC4439265 DOI: 10.1016/j.pscychresns.2015.03.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 02/05/2015] [Accepted: 03/23/2015] [Indexed: 11/20/2022]
Abstract
Response to stress is dysregulated in psychosis (PSY). fMRI studies showed hyperactivity in hypothalamus (HYPO), hippocampus (HIPP), amygdala (AMYG), anterior cingulate (ACC), orbital and medial prefrontal (OFC; mPFC) cortices, with some studies reporting sex differences. We predicted abnormal steroid hormone levels in PSY would be associated with sex differences in hyperactivity in HYPO, AMYG, and HIPP, and hypoactivity in PFC and ACC, with more severe deficits in men. We studied 32 PSY cases (50.0% women) and 39 controls (43.6% women) using a novel visual stress challenge while collecting blood. PSY males showed BOLD hyperactivity across all hypothesized regions, including HYPO and ACC by FWE-correction. Females showed hyperactivity in HIPP and AMYG and hypoactivity in OFC and mPFC, the latter FWE-corrected. Interaction of group by sex was significant in mPFC (F = 7.00, p = 0.01), with PSY females exhibiting the lowest activity. Male hyperactivity in HYPO and ACC was significantly associated with hypercortisolemia post-stress challenge, and mPFC with low androgens. Steroid hormones and neural activity were dissociated in PSY women. Findings suggest disruptions in neural circuitry-hormone associations in response to stress are sex-dependent in psychosis, particularly in prefrontal cortex.
Collapse
Affiliation(s)
- Jill M Goldstein
- Connors Center for Women׳s Health and Gender Biology, Division of Women׳s Health, Brigham and Women׳s Hospital, Boston, MA, USA; Departments of Psychiatry and Medicine, Harvard Medical School, Boston, MA, USA; Division of Psychiatric Neuroscience, Athinoula A. Martinos Center, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.
| | - Katie Lancaster
- Connors Center for Women׳s Health and Gender Biology, Division of Women׳s Health, Brigham and Women׳s Hospital, Boston, MA, USA.
| | - Julia M Longenecker
- Connors Center for Women׳s Health and Gender Biology, Division of Women׳s Health, Brigham and Women׳s Hospital, Boston, MA, USA.
| | - Brandon Abbs
- Connors Center for Women׳s Health and Gender Biology, Division of Women׳s Health, Brigham and Women׳s Hospital, Boston, MA, USA.
| | - Laura M Holsen
- Connors Center for Women׳s Health and Gender Biology, Division of Women׳s Health, Brigham and Women׳s Hospital, Boston, MA, USA; Departments of Psychiatry and Medicine, Harvard Medical School, Boston, MA, USA; Division of Psychiatric Neuroscience, Athinoula A. Martinos Center, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.
| | - Sara Cherkerzian
- Connors Center for Women׳s Health and Gender Biology, Division of Women׳s Health, Brigham and Women׳s Hospital, Boston, MA, USA.
| | - Susan Whitfield-Gabrieli
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Nicolas Makris
- Departments of Psychiatry and Medicine, Harvard Medical School, Boston, MA, USA; Division of Psychiatric Neuroscience, Athinoula A. Martinos Center, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.
| | - Ming T Tsuang
- Center for Behavior Genomics, Department of Psychiatry, University of California at San Diego, San Diego, CA, USA.
| | - Stephen L Buka
- Department of Community Health, Brown University, Providence, RI, USA.
| | - Larry J Seidman
- Division of Psychiatric Neuroscience, Athinoula A. Martinos Center, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Beth Israel Deaconess Medical Center, Division of Public Psychiatry, Massachusetts Mental Health Center and Harvard Medical School, Boston, MA, USA.
| | - Anne Klibanski
- Department of Medicine, Harvard Medical School, Boston, MA, USA; Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
30
|
Neonatal dexamethasone accelerates spreading depression in the rat, and antioxidant vitamins counteract this effect. Brain Res 2014; 1591:93-101. [DOI: 10.1016/j.brainres.2014.09.075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/29/2014] [Accepted: 09/30/2014] [Indexed: 12/27/2022]
|
31
|
Zhang R, Bo T, Shen L, Luo S, Li J. Effect of dexamethasone on intelligence and hearing in preterm infants: a meta-analysis. Neural Regen Res 2014; 9:637-45. [PMID: 25206867 PMCID: PMC4146231 DOI: 10.4103/1673-5374.130085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2014] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE A meta-analysis of published randomized controlled trials investigating the long-term effect of dexamethasone on the nervous system of preterm infants. DATA SOURCES Online literature retrieval was conducted using The Cochrane Library (from January 1993 to June 2013), EMBASE (from January 1980 to June 2013), MEDLINE (from January 1963 to June 2013), OVID (from January 1993 to June 2013), Springer (from January 1994 to June 2013) and Chinese Academic Journal Full-text Database (from January 1994 to June 2013). Key words were preterm infants and dexamethasone in English and Chinese. STUDY SELECTION Selected studies were randomized controlled trials assessing the effect of intravenous dexamethasone in preterm infants. The quality of the included papers was evaluated and those without the development of the nervous system and animal experiments were excluded. Quality assessment was performed through bias risk evaluation in accordance with Cochrane Handbook 5.1.0 software in the Cochrane Collaboration. The homogeneous studies were analyzed and compared using Revman 5.2.6 software, and then effect model was selected and analyzed. Those papers failed to be included in the meta-analysis were subjected to descriptive analysis. MAIN OUTCOME MEASURES Nervous system injury in preterm infants. RESULTS Ten randomized controlled trials were screened, involving 1,038 subjects. Among them 512 cases received dexamethasone treatment while 526 cases served as placebo control group and blank control group. Meta-analysis results showed that the incidence of cerebral palsy, visual impairment and hearing loss in preterm infants after dexamethasone treatment within 7 days after birth was similar to that in the control group (RR = 1.47, 95%CI: 0.97-2.21; RR = 1.46, 95%CI: 0.97-2.20; RR = 0.80, 95%CI: 0.54-1.18; P > 0.05), but intelligence quotient was significantly decreased compared with the control group (MD = -3.55, 95%CI: -6.59 to -0.51; P = 0.02). Preterm infants treated with dexamethasone 7 days after birth demonstrated an incidence of cerebral palsy and visual impairment, and changes in intelligence quotient similar to those in the control group (RR = 1.26, 95%CI: 0.89-1.79; RR = 1.37, 95%CI: 0.73-2.59; RR = 0.53, 95%CI: 0.32-0.89; RR = 1.66, 95%CI: -4.7 to 8.01; P > 0.05). However, the incidence of hearing loss was significantly increased compared with that in the control group (RR = 0.53, 95%CI: 0.32-0.89; P = 0.02). CONCLUSION Dexamethasone may affect the intelligence of preterm infants in the early stages after birth, but may lead to hearing impairment at later stages after birth. More reliable conclusions should be made through large-size, multi-center, well-designed randomized controlled trials.
Collapse
Affiliation(s)
- Ruolin Zhang
- Department of Pediatrics, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Tao Bo
- Department of Pediatrics, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Li Shen
- Department of Pediatrics, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Senlin Luo
- Department of Pediatrics, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jian Li
- Department of Pediatrics, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
32
|
Goldstein JM, Holsen L, Handa R, Tobet S. Fetal hormonal programming of sex differences in depression: linking women's mental health with sex differences in the brain across the lifespan. Front Neurosci 2014; 8:247. [PMID: 25249929 PMCID: PMC4157606 DOI: 10.3389/fnins.2014.00247] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/24/2014] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jill M Goldstein
- Division of Women's Health, Departments of Psychiatry and Medicine, Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital Boston, MA, USA ; Departments of Psychiatry and Medicine, Harvard Medical School Boston, MA, USA ; Division of Psychiatric Neuroscience, Department of Psychiatry, Massachusetts General Hospital Boston, MA, USA
| | - Laura Holsen
- Division of Women's Health, Departments of Psychiatry and Medicine, Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital Boston, MA, USA ; Departments of Psychiatry and Medicine, Harvard Medical School Boston, MA, USA
| | - Robert Handa
- Department of Basic Medical Sciences, University of Arizona College of Medicine Phoenix, AZ, USA
| | - Stuart Tobet
- Department of Biomedical Sciences and School of Biomedical Engineering, College of Veterinary Medicine and Biomedical Sciences, Colorado State University Fort Collins, CO, USA
| |
Collapse
|
33
|
Maternal treatment with glucocorticoids modulates gap junction protein expression in the ovine fetal brain. Neuroscience 2014; 275:248-58. [PMID: 24929069 DOI: 10.1016/j.neuroscience.2014.05.066] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 04/28/2014] [Accepted: 05/14/2014] [Indexed: 12/31/2022]
Abstract
Gap junctions facilitate intercellular communication and are important in brain development. Connexins (Cx) comprise a transmembrane protein family that forms gap junctions. Cx-32 is expressed in oligodendrocytes and neurons, Cx-36 in neurons, and Cx-43 in astrocytes. Although single antenatal steroid courses are recommended for fetal lung maturation, multiple courses can be given to women at recurrent risk for premature delivery. We examined the effects of single and multiple glucocorticoid courses on Cx-32, Cx-36, and Cx-43 protein expressions in the fetal cerebral cortex, cerebellum, and spinal cord, and differences in Cx expression among brain regions under basal conditions. In the single-course groups, the ewes received dexamethasone (6 mg) or placebo as four intramuscular injections every 12h over 48 h. In the multiple-course groups, the ewes received the same treatment, once a week for 5 weeks starting at 76-78 days of gestation. Cx were measured by Western immunoblot on brain samples from 105 to 108-day gestation fetuses. A single dexamethasone course was associated with increases (P<0.05) in cerebral cortical and spinal cord Cx-36 and Cx-43 and multiple courses with increases in cerebellar and spinal cord Cx-36, and cerebral cortical and cerebellar Cx-43. Cx-32 did not change. Cx-32 was higher in the cerebellum than cerebral cortex and spinal cord, Cx-36 higher in the spinal cord than cerebellum, and Cx-43 higher in the cerebellum and spinal cord than cerebral cortex during basal conditions. In conclusion, maternal glucocorticoid therapy increases specific Cx, responses to different maternal courses vary among Cx and brain regions, and Cx expression differs among brain regions under basal conditions. Maternal treatment with glucocorticoids differentially modulates Cx in the fetal brain.
Collapse
|
34
|
Menshanov PN, Bannova AV, Dygalo NN. Dexamethasone suppresses the locomotor response of neonatal rats to novel environment. Behav Brain Res 2014; 271:43-50. [PMID: 24886779 DOI: 10.1016/j.bbr.2014.05.053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 05/23/2014] [Accepted: 05/24/2014] [Indexed: 11/18/2022]
Abstract
Locomotion of animals in the novel environment is determined by two main factors-the intrinsic motor activity and the specific locomotor response to novelty. Glucocorticoids alter neurobehavioral development of mammals and its locomotor manifestations. However, it remains unclear whether the intrinsic and/or the novelty-induced activity are affected by glucocorticoids during early life. Here, the principal component analysis was used to determine the main factors that underlie alterations in locomotion of rat pups treated with dexamethasone. It was shown that neonatal rats exhibited an enhanced locomotion in the novel environment beginning from postnatal day (PD) 5. We found for the first time that this reaction was significantly suppressed by dexamethasone. The effect was specific to the novelty-induced component of behavior, while the intrinsic locomotor activity was not affected by glucocorticoid treatment. The suppression of the behavioral response to novelty was maximal at PD7 and vanquished at PD10-11. In parallel with the hormonal effect on the behavior, dexamethasone upregulated the main cell death executor-active caspase-3 in the prefrontal cortex of 7-day old rats. Thus, dexamethasone-induced alterations in the novelty-related behavior may be the earliest visible signs of the brain damage that could lead to forthcoming depressive state or schizophrenia, emerging as a result of neonatal stress or glucocorticoid treatment.
Collapse
Affiliation(s)
- Petr N Menshanov
- Functional Neurogenomics Laboratory, Institute of Cytology and Genetics SBRAS, Russian Academy of Science, Lavrentyev av. 10., Novosibirsk 630090, Russian Federation; Department of Natural Sciences, Novosibirsk State University, Pirogova st. 2, Novosibirsk 630090, Russian Federation.
| | - Anita V Bannova
- Functional Neurogenomics Laboratory, Institute of Cytology and Genetics SBRAS, Russian Academy of Science, Lavrentyev av. 10., Novosibirsk 630090, Russian Federation
| | - Nikolay N Dygalo
- Functional Neurogenomics Laboratory, Institute of Cytology and Genetics SBRAS, Russian Academy of Science, Lavrentyev av. 10., Novosibirsk 630090, Russian Federation; Department of Natural Sciences, Novosibirsk State University, Pirogova st. 2, Novosibirsk 630090, Russian Federation
| |
Collapse
|
35
|
Prenatal stress and inhibitory neuron systems: implications for neuropsychiatric disorders. Mol Psychiatry 2014; 19:641-51. [PMID: 24751963 PMCID: PMC4031286 DOI: 10.1038/mp.2014.35] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 03/03/2014] [Accepted: 03/10/2014] [Indexed: 01/09/2023]
Abstract
Prenatal stress is a risk factor for several psychiatric disorders in which inhibitory neuron pathology is implicated. A growing body of research demonstrates that inhibitory circuitry in the brain is directly and persistently affected by prenatal stress. This review synthesizes research that explores how this early developmental risk factor impacts inhibitory neurons and how these findings intersect with research on risk factors and inhibitory neuron pathophysiology in schizophrenia, anxiety, autism and Tourette syndrome. The specific impact of prenatal stress on inhibitory neurons, particularly developmental mechanisms, may elucidate further the pathophysiology of these disorders.
Collapse
|
36
|
Chen CV, Brummet JL, Lonstein JS, Jordan CL, Breedlove SM. New knockout model confirms a role for androgen receptors in regulating anxiety-like behaviors and HPA response in mice. Horm Behav 2014; 65:211-8. [PMID: 24440052 PMCID: PMC4295784 DOI: 10.1016/j.yhbeh.2014.01.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 01/02/2014] [Accepted: 01/08/2014] [Indexed: 12/30/2022]
Abstract
Men are less likely than women to suffer from anxiety disorders. Because gonadal hormones play a crucial role in many behavioral sex differences, they may underlie sex differences in human anxiety. In rodents, testosterone (T) exerts anxiolytic effects via the androgen receptor (AR): we found that male mice with a naturally-occurring mutation rendering the AR dysfunctional, referred to as spontaneous testicular feminization mutation (sTfm), showed more anxiety-like behaviors than wildtype (WT) males. Here, we used Cre-lox recombination technology to create another dysfunctional allele for AR. These induced Tfm (iTfm) animals also displayed more anxiety-like behaviors than WTs. We further found that AR-modulation of these behaviors interacts with circadian phase. When tested in the resting phase, iTfms appeared more anxious than WTs in the open field, novel object and elevated plus maze tests, but not the light/dark box. However, when tested during the active phase (lights off), iTfms showed more anxiety-related behavior than WTs in all four tests. Finally, we confirmed a role of T acting via AR in regulating HPA axis activity, as WT males with T showed a lower baseline and overall corticosterone response, and a faster return to baseline following mild stress than did WT males without T or iTfms. These findings demonstrate that this recombined AR allele is a valuable model for studying androgenic modulation of anxiety, that the anxiolytic effects of AR in mice are more prominent in the active phase, and that HPA axis modulation by T is AR dependent.
Collapse
MESH Headings
- Androgen-Insensitivity Syndrome/genetics
- Androgen-Insensitivity Syndrome/physiopathology
- Animals
- Anxiety/metabolism
- Anxiety/physiopathology
- Behavior, Animal/physiology
- Corticosterone/blood
- Disease Models, Animal
- Female
- Hypothalamo-Hypophyseal System/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Animal
- Photoperiod
- Pituitary-Adrenal System/metabolism
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Receptors, Androgen/physiology
- Stress, Psychological/metabolism
- Stress, Psychological/physiopathology
- Testosterone/physiology
Collapse
Affiliation(s)
- Chieh V Chen
- Michigan State University, Psychology Department, 293 Farm Lane, Giltner Room 108, East Lansing, MI 48824, USA.
| | - Jennifer L Brummet
- Michigan State University, Psychology Department, 293 Farm Lane, Giltner Room 108, East Lansing, MI 48824, USA
| | - Joseph S Lonstein
- Michigan State University, Psychology Department, 293 Farm Lane, Giltner Room 108, East Lansing, MI 48824, USA; Michigan State University, Neuroscience Program, 293 Farm Lane, Giltner Room 108, East Lansing, MI 48824, USA
| | - Cynthia L Jordan
- Michigan State University, Psychology Department, 293 Farm Lane, Giltner Room 108, East Lansing, MI 48824, USA; Michigan State University, Neuroscience Program, 293 Farm Lane, Giltner Room 108, East Lansing, MI 48824, USA
| | - S Marc Breedlove
- Michigan State University, Psychology Department, 293 Farm Lane, Giltner Room 108, East Lansing, MI 48824, USA; Michigan State University, Neuroscience Program, 293 Farm Lane, Giltner Room 108, East Lansing, MI 48824, USA
| |
Collapse
|
37
|
Goldstein JM, Handa RJ, Tobet SA. Disruption of fetal hormonal programming (prenatal stress) implicates shared risk for sex differences in depression and cardiovascular disease. Front Neuroendocrinol 2014; 35:140-58. [PMID: 24355523 PMCID: PMC3917309 DOI: 10.1016/j.yfrne.2013.12.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 10/31/2013] [Accepted: 12/04/2013] [Indexed: 12/19/2022]
Abstract
Comorbidity of major depressive disorder (MDD) and cardiovascular disease (CVD) represents the fourth leading cause of morbidity and mortality worldwide, and women have a two times greater risk than men. Thus understanding the pathophysiology has widespread implications for attenuation and prevention of disease burden. We suggest that sex-dependent MDD-CVD comorbidity may result from alterations in fetal programming consequent to the prenatal maternal environments that produce excess glucocorticoids, which then drive sex-dependent developmental alterations of the fetal hypothalamic-pituitary-adrenal (HPA) axis circuitry impacting mood, stress regulation, autonomic nervous system (ANS), and the vasculature in adulthood. Evidence is consistent with the hypothesis that disruptions of pathways associated with gamma aminobutyric acid (GABA) in neuronal and vascular development and growth factors have critical roles in key developmental periods and adult responses to injury in heart and brain. Understanding the potential fetal origins of these sex differences will contribute to development of novel sex-dependent therapeutics.
Collapse
Affiliation(s)
- J M Goldstein
- Departments of Psychiatry and Medicine, Harvard Medical School, Boston, MA, USA; Brigham and Women's Hospital (BWH), Connors Center for Women's Health & Gender Biology, 1620 Tremont St. BC-3-34, Boston, MA 02120, USA; BWH, Departments of Psychiatry and Medicine, 1620 Tremont St. BC-3-34, Boston, MA 02120, USA.
| | - R J Handa
- Department of Basic Medical Sciences, University of Arizona College of Medicine, 425 N. Fifth Street, Phoenix, AZ 85004, USA
| | - S A Tobet
- Department of Biomedical Sciences and School of Biomedical Engineering, Colorado State University, 1617 Campus Delivery, Fort Collins, CO 80523, USA
| |
Collapse
|
38
|
Ahern TH, Krug S, Carr AV, Murray EK, Fitzpatrick E, Bengston L, McCutcheon J, De Vries GJ, Forger NG. Cell death atlas of the postnatal mouse ventral forebrain and hypothalamus: effects of age and sex. J Comp Neurol 2013; 521:2551-69. [PMID: 23296992 PMCID: PMC4968939 DOI: 10.1002/cne.23298] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 11/28/2012] [Accepted: 12/26/2012] [Indexed: 01/21/2023]
Abstract
Naturally occurring cell death is essential to the development of the mammalian nervous system. Although the importance of developmental cell death has been appreciated for decades, there is no comprehensive account of cell death across brain areas in the mouse. Moreover, several regional sex differences in cell death have been described for the ventral forebrain and hypothalamus, but it is not known how widespread the phenomenon is. We used immunohistochemical detection of activated caspase-3 to identify dying cells in the brains of male and female mice from postnatal day (P) 1 to P11. Cell death density, total number of dying cells, and regional volume were determined in 16 regions of the hypothalamus and ventral forebrain (the anterior hypothalamus, arcuate nucleus, anteroventral periventricular nucleus, medial preoptic nucleus, paraventricular nucleus, suprachiasmatic nucleus, and ventromedial nucleus of the hypothalamus; the basolateral, central, and medial amygdala; the lateral and principal nuclei of the bed nuclei of the stria terminalis; the caudate-putamen; the globus pallidus; the lateral septum; and the islands of Calleja). All regions showed a significant effect of age on cell death. The timing of peak cell death varied between P1 to P7, and the average rate of cell death varied tenfold among regions. Several significant sex differences in cell death and/or regional volume were detected. These data address large gaps in the developmental literature and suggest interesting region-specific differences in the prevalence and timing of cell death in the hypothalamus and ventral forebrain.
Collapse
Affiliation(s)
- Todd H. Ahern
- Center for Behavioral Neuroscience, Department of Psychology, Quinnipiac University, Hamden, Connecticut 06518
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Stefanie Krug
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Audrey V. Carr
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Elaine K. Murray
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Emmett Fitzpatrick
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Lynn Bengston
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Jill McCutcheon
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Geert J. De Vries
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303
| | - Nancy G. Forger
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303
| |
Collapse
|
39
|
Zuloaga DG, Siegel JA, Acevedo SF, Agam M, Raber J. Developmental methamphetamine exposure results in short- and long-term alterations in hypothalamic-pituitary-adrenal-axis-associated proteins. Dev Neurosci 2013; 35:338-46. [PMID: 23860125 DOI: 10.1159/000351278] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 04/11/2013] [Indexed: 11/19/2022] Open
Abstract
Developmental exposure to methamphetamine (MA) causes long-term behavioral and cognitive deficits. One pathway through which MA might induce these deficits is by elevating glucocorticoid levels. Glucocorticoid overexposure during brain development can lead to long-term disruptions in the hypothalamic-pituitary-adrenal (HPA) axis. These disruptions affect the regulation of stress responses and may contribute to behavioral and cognitive deficits reported following developmental MA exposure. Furthermore, alterations in proteins associated with the HPA axis, including vasopressin, oxytocin, and glucocorticoid receptors (GR), are correlated with disruptions in mood and cognition. We therefore hypothesized that early MA exposure will result in short- and long-term alterations in the expression of HPA axis-associated proteins. Male mice were treated with MA (5 mg/kg daily) or saline from postnatal day (P) 11 to P20. At P20 and P90, mice were perfused and their brains processed for vasopressin, oxytocin, and GR immunoreactivity within HPA axis-associated regions. At P20, there was a significant decrease in the number of vasopressin-immunoreactive cells and the area occupied by vasopressin immunoreactivity in the paraventricular nucleus (PVN) of MA-treated mice, but no difference in oxytocin immunoreactivity in the PVN, or GR immunoreactivity in the hippocampus or PVN. In the central nucleus of the amygdala, the area occupied by GR immunoreactivity was decreased by MA. At P90, the number of vasopressin-immunoreactive cells was still decreased, but the area occupied by vasopressin immunoreactivity no longer differed from saline controls. No effects of MA were found on oxytocin or GR immunoreactivity at P90. Thus developmental MA exposure has short- and long-term effects on vasopressin immunoreactivity and short-term effects on GR immunoreactivity.
Collapse
Affiliation(s)
- Damian G Zuloaga
- Department of Behavioral Neuroscience, Oregon Health and Science University Portland, Portland, OR 97239, USA.
| | | | | | | | | |
Collapse
|
40
|
Grey KR, Davis EP, Sandman CA, Glynn LM. Human milk cortisol is associated with infant temperament. Psychoneuroendocrinology 2013; 38:1178-85. [PMID: 23265309 PMCID: PMC4777694 DOI: 10.1016/j.psyneuen.2012.11.002] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 11/05/2012] [Accepted: 11/05/2012] [Indexed: 10/27/2022]
Abstract
The implications of the biologically active elements in milk for the mammalian infant are largely unknown. Animal models demonstrate that transmission of glucocorticoids through milk influences behavior and modifies brain development in offspring. The aim of this study was to determine the relation between human milk cortisol levels and temperament of the breastfed infant. Fifty-two mother and infant pairs participated when the infants were three-months old. Milk cortisol levels were assessed and each mother completed the Infant Behavior Questionnaire (IBQ), a widely used parent-report measure of infant temperament. Analyses revealed a positive association between milk cortisol and the negative affectivity dimension of the IBQ (partial r=.37, p<.01). No correlation was found between elevated cortisol levels and the surgency/extraversion or the orienting/regulation dimensions. Further, the positive association between increased maternal milk cortisol and negative affectivity was present among girls (β=.59, p<.01), but not among boys. (Although, the sex by milk cortisol interaction term was not statistically significant, suggesting that these results require replication.) Environmental factors such as maternal demographics and negative maternal affect (depression and perceived stress) at the time of assessment did not account for the positive association. The findings support the proposal that exposure to elevated levels of cortisol in human milk influences infant temperament. The findings further suggest that mothers have the ability to shape offspring phenotype through the transmission of biologically active components in milk.
Collapse
Affiliation(s)
- Katherine R. Grey
- Crean School of Health and Life Sciences, Chapman University, One University Drive, Orange, CA 92866, United States
| | - Elysia Poggi Davis
- Department of Psychiatry and Human Behavior, University of California, Irvine, United States,Department of Pediatrics, University of California, Irvine, United States
| | - Curt A. Sandman
- Department of Psychiatry and Human Behavior, University of California, Irvine, United States
| | - Laura M. Glynn
- Crean School of Health and Life Sciences, Chapman University, One University Drive, Orange, CA 92866, United States,Department of Psychiatry and Human Behavior, University of California, Irvine, United States
| |
Collapse
|
41
|
Saul ML, Helmreich DL, Callahan LM, Fudge JL. Differences in amygdala cell proliferation between adolescent and young adult rats. Dev Psychobiol 2013; 56:517-28. [PMID: 23775606 DOI: 10.1002/dev.21115] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 03/12/2013] [Indexed: 01/10/2023]
Abstract
Adolescence is characterized by changes in both behavior and neural organization. During this period, the amygdala, a structure that mediates social and emotional behaviors, is changing in terms of neural and glia density. We examined cell proliferation within the amygdala of adolescent (post natal day (PND) 31) and adult (PND 70) male Sprague-Dawley rats using BrdU (bromodeoxyuridine) to label dividing cells. BrdU-labeled cells were distributed throughout the amygdala, often found in fibers surrounding major nuclei. Using two independent cell counting strategies under light and confocal microcopy, respectively, we found significantly more labeled cells in the amygdala in adolescent compared to adult animals (239.3 ± 87.18 vs. 44.75 ± 13.68; n=4/group; p<.05). BrdU/doublecortin (DCX) positive cells constitute approximately 30% of all dividing cells in the amygdala in both adolescents and adults. These data suggest that compared to young adulthood, adolescence is a relatively active period of cell proliferation in the amygdala. Moreover, the normal decline in dividing cells with age does not preferentially affect cells co-containing DCX-immunoreactivity.
Collapse
Affiliation(s)
- Michele L Saul
- Department of Neurobiology and Anatomy, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642
| | | | | | | |
Collapse
|
42
|
Ichinohashi Y, Sato Y, Saito A, Ito M, Watanabe K, Hayakawa M, Nakanishi K, Wakatsuki A, Oohira A. Dexamethasone administration to the neonatal rat results in neurological dysfunction at the juvenile stage even at low doses. Early Hum Dev 2013; 89:283-8. [PMID: 23153570 DOI: 10.1016/j.earlhumdev.2012.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Revised: 10/07/2012] [Accepted: 10/16/2012] [Indexed: 10/27/2022]
Abstract
Dexamethasone (DEX), a synthetic glucocorticoid, has been widely used to prevent the development of a variety of poor health conditions in premature infants including chronic lung disease, inflammation, circulatory failure, and shock. Although there are some reports of neurologic complications related to DEX exposure, its full effects on the premature brain have not been examined in detail. To investigate the effects of DEX on neural development, we first administered low doses (0.2 mg/kg bodyweight or less) of the glucocorticoid to neonatal rats on a daily basis during the first postnatal week and examined subsequent behavioral alterations at the juvenile stage. DEX-treated rats exhibited not only a significant reduction in both somatic and brain weights but also learning disabilities as revealed in the shuttle avoidance test. The hippocampi of DEX-treated rats displayed a high apoptotic and a low mitotic cell density compared to control rats on day 7 after birth. In a subsequent experiment, neural stem/progenitor cells were cultured in the presence of DEX for 6 days. The glucocorticoid inhibited cell growth without an increase in cell death. These results suggest that administration of DEX to premature infants induces neurological dysfunction via inhibition of the proliferation of neural stem/progenitor cells.
Collapse
Affiliation(s)
- Yuko Ichinohashi
- Division of Obstetrics and Gynecology, Aichi Medical University Graduate School of Medicine, Nagakute, Aichi 480-1195, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Volumetric parcellation methodology of the human hypothalamus in neuroimaging: normative data and sex differences. Neuroimage 2012; 69:1-10. [PMID: 23247186 DOI: 10.1016/j.neuroimage.2012.12.008] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 11/30/2012] [Accepted: 12/01/2012] [Indexed: 11/22/2022] Open
Abstract
There is increasing evidence regarding the importance of the hypothalamus for understanding sex differences in relation to neurological, psychiatric, endocrine and sleep disorders. Although different in histology, physiology, connections and function, multiple hypothalamic nuclei subserve non-voluntary functions and are nodal points for the purpose of maintaining homeostasis of the organism. Thus, given the critical importance of hypothalamic nuclei and their key multiple roles in regulating basic functions, it is important to develop the ability to conduct in vivo human studies of anatomic structure, volume, connectivity, and function of hypothalamic regions represented at the level of its nuclei. The goals of the present study were to develop a novel method of semi-automated volumetric parcellation for the human hypothalamus that could be used to investigate clinical conditions using MRI and to demonstrate its applicability. The proposed new method subdivides the hypothalamus into five parcels based on visible anatomic landmarks associated with specific nuclear groupings and was confirmed using two ex vivo hypothalami that were imaged in a 7 T (7 T) scanner and processed histologically. Imaging results were compared with histology from the same brain. Further, the method was applied to 44 healthy adults (26 men; 18 women, comparable on age, handedness, ethnicity, SES) to derive normative volumes and assess sex differences in hypothalamic regions using 1.5 T MRI. Men compared to women had a significantly larger total hypothalamus, relative to cerebrum size, similar for both hemispheres, a difference that was primarily driven by the tuberal region, with the sex effect size being largest in the superior tuberal region and, to a lesser extent, inferior tuberal region. Given the critical role of hypothalamic nuclei in multiple chronic diseases and the importance of sex differences, we argue that the use of the novel methodology presented here will allow for critical investigations of these disorders and further delineation of potential treatments, particularly sex-specific approaches to gene and drug discoveries that involve hypothalamic nuclei.
Collapse
|
44
|
Effects of low-intensity electromagnetic fields on the proliferation and differentiation of cultured mouse bone marrow stromal cells. Phys Ther 2012; 92:1208-19. [PMID: 22577063 DOI: 10.2522/ptj.20110224] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Electromagnetic fields (EMFs) used in stem-cell tissue engineering can help elucidate their biological principles. OBJECTIVE The aim of this study was to investigate the effects of low-intensity EMFs on cell proliferation, differentiation, and cycle in mouse bone marrow stromal cells (BMSCs) and the in vivo effects of EMFs on BMSC. METHODS Harvested BMSCs were cultured for 3 generations and divided into 4 groups. The methylthiotetrazole (MTT) assay was used to evaluate cell proliferation, and alkaline phosphatase activity was measured via a colorimetric assay on the 3rd, 7th, and 10th days. Changes in cell cycle also were analyzed on the 7th day, and bone nodule formation was analyzed on the 12th day. Additionally, the expression of the collagen I gene was examined by reverse transcription-polymerase chain reaction (RT-PCR) on the 10th day. The BMSCs of the irradiated group and the control group were transplanted into cortical bone of different mice femurs separately, with poly(lactic-co-glycolic acid) (PLGA) serving as a scaffold. After 4 and 8 weeks, bone the bone specimens of mice were sliced and stained by hematoxylin and eosin separately. RESULTS The results showed that EMFs (0.5 mT, 50 Hz) accelerated cellular proliferation, enhanced cellular differentiation, and increased the percentage of cells in the G(2)/M+S (postsynthetic gap 2 period/mitotic phase + S phase) of the stimulation. The EMF-exposed groups had significantly higher collagen I messenger RNA levels than the control group. The EMF + osteogenic medium-treated group readily formed bone nodules. Hematoxylin and eosin staining showed a clear flaking of bone tissue in the irradiated group. CONCLUSION Irradiation of BMSCs with low-intensity EMFs (0.5 mT, 50 Hz) increased cell proliferation and induced cell differentiation. The results of this study did not establish a stricter animal model for studying osteogenesis, and only short-term results were investigated. Further study of the mechanism of EMF is needed.
Collapse
|
45
|
Bennet L, Davidson JO, Koome M, Gunn AJ. Glucocorticoids and preterm hypoxic-ischemic brain injury: the good and the bad. J Pregnancy 2012; 2012:751694. [PMID: 22970371 PMCID: PMC3431094 DOI: 10.1155/2012/751694] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 07/13/2012] [Indexed: 12/20/2022] Open
Abstract
Fetuses at risk of premature delivery are now routinely exposed to maternal treatment with synthetic glucocorticoids. In randomized clinical trials, these substantially reduce acute neonatal systemic morbidity, and mortality, after premature birth and reduce intraventricular hemorrhage. However, the overall neurodevelopmental impact is surprisingly unclear; worryingly, postnatal glucocorticoids are consistently associated with impaired brain development. We review the clinical and experimental evidence on how glucocorticoids may affect the developing brain and highlight the need for systematic research.
Collapse
Affiliation(s)
- Laura Bennet
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland 1142, New Zealand.
| | | | | | | |
Collapse
|
46
|
Carbone DL, Zuloaga DG, Lacagnina AF, McGivern RF, Handa RJ. Exposure to dexamethasone during late gestation causes female-specific decreases in core body temperature and prepro-thyrotropin-releasing hormone expression in the paraventricular nucleus of the hypothalamus in rats. Physiol Behav 2012; 108:6-12. [PMID: 22884559 DOI: 10.1016/j.physbeh.2012.07.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 07/13/2012] [Accepted: 07/26/2012] [Indexed: 12/20/2022]
Abstract
Synthetic glucocorticoids (GC) have been used to promote lung development in preterm infants, thereby decreasing respiratory distress syndrome and mortality, yet, concern has arisen from reports that such treatment predisposes individuals to disease in adulthood. Given the variety of preclinical studies that show metabolic and behavioral abnormalities in adulthood following fetal exposure to synthetic GC, we examined the effect of in utero exposure to the synthetic GC, dexamethasone (DEX), on hypothalamic expression of thyrotropin-releasing hormone (TRH) a central neuropeptide involved in mediating behavior and metabolic balance. Pregnant Sprague-Dawley rats were administered 0.4mg/kg DEX on gestational days 18-21. As adults (postnatal day (PD) 60), the offspring were fitted with temperature sensing transmitters allowing real-time monitoring of core body temperature (CBT) across the 24h light dark period. This revealed a significant decrease in CBT throughout the day in prenatal DEX-treated females on estrus and diestrus, but not in male offspring. The reduction in CBT by prenatal DEX exposure was accompanied by a significant decrease in the expression of Trh transcript in the paraventricular nucleus of the hypothalamus (PVN) of female rats at PD 60 and this effect was also present on PD7. There was also a female-specific reduction in the number of preproTRH-immunoreactive (ir) neurons in the PVN, with ppTRH-ir nerve fibers decreases that were present in both male and female offspring. No changes in thyroid hormone (triiodothyronine, T3; thyroxine, T4) were observed in adult offspring, but during development, both males and females (PD14) had lower T3 and T4 levels. These data indicate abnormal expression of TRH results from fetal DEX exposure during late gestation, possibly explaining the decreased CBT observed in the female offspring.
Collapse
Affiliation(s)
- David L Carbone
- University of Arizona College of Medicine-Phoenix, Department of Basic Medical Sciences, Phoenix, AZ 85004-2157, United States.
| | | | | | | | | |
Collapse
|
47
|
Fernández-Guasti A, Fiedler JL, Herrera L, Handa RJ. Sex, stress, and mood disorders: at the intersection of adrenal and gonadal hormones. Horm Metab Res 2012; 44:607-18. [PMID: 22581646 PMCID: PMC3584173 DOI: 10.1055/s-0032-1312592] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The risk for neuropsychiatric illnesses has a strong sex bias, and for major depressive disorder (MDD), females show a more than 2-fold greater risk compared to males. Such mood disorders are commonly associated with a dysregulation of the hypothalamo-pituitary-adrenal (HPA) axis. Thus, sex differences in the incidence of MDD may be related with the levels of gonadal steroid hormone in adulthood or during early development as well as with the sex differences in HPA axis function. In rodents, organizational and activational effects of gonadal steroid hormones have been described for the regulation of HPA axis function and, if consistent with humans, this may underlie the increased risk of mood disorders in women. Other developmental factors, such as prenatal stress and prenatal overexposure to glucocorticoids can also impact behaviors and neuroendocrine responses to stress in adulthood and these effects are also reported to occur with sex differences. Similarly, in humans, the clinical benefits of antidepressants are associated with the normalization of the dysregulated HPA axis, and genetic polymorphisms have been found in some genes involved in controlling the stress response. This review examines some potential factors contributing to the sex difference in the risk of affective disorders with a focus on adrenal and gonadal hormones as potential modulators. Genetic and environmental factors that contribute to individual risk for affective disorders are also described. Ultimately, future treatment strategies for depression should consider all of these biological elements in their design.
Collapse
Affiliation(s)
| | - J. L. Fiedler
- Laboratory of Neuroplasticity and Neurogenetics, Department of Biochemistry and Molecular Biology, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
| | - L. Herrera
- Human Genetics Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - R. J. Handa
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona, USA
| |
Collapse
|
48
|
Zuloaga DG, Carbone DL, Handa RJ. Prenatal dexamethasone selectively decreases calretinin expression in the adult female lateral amygdala. Neurosci Lett 2012; 521:109-14. [PMID: 22668856 DOI: 10.1016/j.neulet.2012.05.058] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 04/06/2012] [Accepted: 05/18/2012] [Indexed: 01/07/2023]
Abstract
Exposure to high levels of glucocorticoids (GCs) during early development results in lasting disturbances in emotional behavior in rodents. Inhibitory GABAergic neurons, classified by their expression of calcium binding proteins (CBPs), also contribute to stress-related behaviors and may be GC sensitive during development. Therefore, in the present study we investigated the effects of prenatal treatment with the glucocorticoid receptor agonist dexamethasone (DEX) on expression of calbindin and calretinin in brain areas critical to emotional regulation (basolateral/lateral amygdala and hippocampal CA1 and CA3 regions). Late gestational treatment with DEX (gestational days 18-22) significantly decreased the density of calretinin immunoreactive cells in the lateral amygdala of adult female offspring with no differences in the basolateral amygdala, hippocampal CA1, or CA3 regions. Moreover, there were no effects of gestational DEX treatment on calretinin expression in males. Calbindin expression in adulthood was unaltered within either amygdala or hippocampal subregion of either sex following prenatal DEX treatment. Together these findings indicate that late gestational DEX treatment causes a targeted reduction of calretinin within the lateral amygdala of females and this may be one mechanism through which developmental glucocorticoid exposure contributes to lasting alterations in emotional behavior.
Collapse
Affiliation(s)
- Damian G Zuloaga
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, United States.
| | | | | |
Collapse
|