1
|
Hernandez M, Ghislin S, Lalonde R, Strazielle C. Corticosterone effects on postnatal cerebellar development in mice. Neurochem Int 2023; 171:105611. [PMID: 37704081 DOI: 10.1016/j.neuint.2023.105611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 08/23/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Glucocorticoids administered early in infancy can affect the architectonic organization of brain structures, particularly those with a postnatal development and resulting in long-term deficits of neuromotor function and cognition. The present study was undertaken to study the effects of daily corticosterone (CORT) injections at a pharmacological dose from postnatal days 8-15 on cerebellar and hippocampal development in mouse pups. Gene expression status for trophic factors involved in synaptic development and function as well as measures of layer thickness associated with cytochrome oxidase labelling were analyzed in the hippocampus, hypothalamus, and specific cerebellar lobules involved in motor control. Repeated CORT injections dysregulated the HPA axis with increased Crh and Nr3c1 mRNA levels in the hypothalamus and a resulting higher serum corticosterone level. The CORT treatment altered the morphology of the hippocampus and down-regulated gene transcription for corticotropin-releasing hormone (Crh) and its type-1 receptor (Crhr1), glucocorticoid receptor (Nr3c1), and brain-derived neurotrophic factor Bdnf and its receptor Ntrk2 (neurotrophic receptor tyrosine kinase 2). Similar mRNA expression decreases were found in the cerebellum for Crhr1, Crhr2, Nr3c1, and Grid2 (glutamatergic δ2 receptor). Morphological alterations and metabolic activity variations were observed in specific cerebellar lobules involved in motor control. The paramedian lobule, normally characterized by mitotic activity in the external germinative layer during the second postnatal week, was atrophic but metabolically hyperactive in its granule cell and molecular layers. On the contrary, lobules with an earlier cell proliferation displayed neurogenesis but a hypoactivated granule cell layer, suggesting a developmental delay in synaptogenesis. The results indicate that glucocorticoid, administered daily during the second postnatal week modulated the developmental programming of the hippocampus and cerebellum. These growth and metabolic alterations may lead possibly to morphological and functional changes later in life.
Collapse
Affiliation(s)
- M Hernandez
- Laboratory of Stress, Immunity, Pathogens (EA 7300), Medical School, University of Lorraine, 54500 Vandœuvre-les-Nancy, France; CHRU Nancy, Vandœuvre-les-Nancy, France
| | - S Ghislin
- Laboratory of Stress, Immunity, Pathogens (EA 7300), Medical School, University of Lorraine, 54500 Vandœuvre-les-Nancy, France
| | - R Lalonde
- Laboratory of Stress, Immunity, Pathogens (EA 7300), Medical School, University of Lorraine, 54500 Vandœuvre-les-Nancy, France
| | - C Strazielle
- Laboratory of Stress, Immunity, Pathogens (EA 7300), Medical School, University of Lorraine, 54500 Vandœuvre-les-Nancy, France; CHRU Nancy, Vandœuvre-les-Nancy, France.
| |
Collapse
|
2
|
Warmerdam LA, van Wezel-Meijler G, de Vries LS, Groenendaal F, Steggerda SJ. The Association of Dexamethasone and Hydrocortisone with Cerebellar Growth in Premature Infants. Neonatology 2023; 120:615-623. [PMID: 37379806 DOI: 10.1159/000531075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 04/28/2023] [Indexed: 06/30/2023]
Abstract
OBJECTIVES Corticosteroids are used to prevent or treat lung disease of prematurity. While neurological side effects have been reported, detailed effects on cerebellar growth are unknown. This study aimed to compare cerebellar growth in premature infants who received dexamethasone or hydrocortisone to premature infants who did not receive postnatal corticosteroids. STUDY DESIGN Retrospective case-control study in infants born at a gestational age of <29 weeks and admitted to two level 3 neonatal intensive care units. Exclusion criteria were severe congenital anomalies and cerebellar or severe supratentorial lesions. Infants were treated with dexamethasone (unit 1) or hydrocortisone (unit 2) for chronic lung disease. Controls (unit 1) did not receive postnatal corticosteroids. Sequential head circumference (HC) and ultrasound measurements of transcerebellar diameter (TCD), biparietal diameter (BPD), and corpus callosum-fastigium length (CCFL) were performed until 40 weeks' postmenstrual age (PMA). Growth was assessed using linear mixed models correcting for PMA at measurement, sex, HC z-score at birth, and a propensity score indicating illness severity. Group differences before treatment were assessed using linear regression. RESULTS 346 infants were included (68 dexamethasone, 37 hydrocortisone, 241 controls). Before starting corticosteroids, TCD, BPD, and HC measurements did not differ between patients and controls at a comparable PMA. After starting treatment, both types of corticosteroid had a negative association with TCD growth. BPD, CCFL, and HC growth were not negatively affected. CONCLUSION Administration of dexamethasone and hydrocortisone are both associated with impaired cerebellar growth in premature infants without evident negative associations with cerebral growth.
Collapse
Affiliation(s)
- Laura A Warmerdam
- Department of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Linda S de Vries
- Department of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Floris Groenendaal
- Department of Neonatology, Wilhelmina Children's Hospital, and Brain Center, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Sylke J Steggerda
- Department of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
3
|
Spoto G, Amore G, Vetri L, Quatrosi G, Cafeo A, Gitto E, Nicotera AG, Di Rosa G. Cerebellum and Prematurity: A Complex Interplay Between Disruptive and Dysmaturational Events. Front Syst Neurosci 2021; 15:655164. [PMID: 34177475 PMCID: PMC8222913 DOI: 10.3389/fnsys.2021.655164] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/17/2021] [Indexed: 12/14/2022] Open
Abstract
The cerebellum plays a critical regulatory role in motor coordination, cognition, behavior, language, memory, and learning, hence overseeing a multiplicity of functions. Cerebellar development begins during early embryonic development, lasting until the first postnatal years. Particularly, the greatest increase of its volume occurs during the third trimester of pregnancy, which represents a critical period for cerebellar maturation. Preterm birth and all the related prenatal and perinatal contingencies may determine both dysmaturative and lesional events, potentially involving the developing cerebellum, and contributing to the constellation of the neuropsychiatric outcomes with several implications in setting-up clinical follow-up and early intervention.
Collapse
Affiliation(s)
- Giulia Spoto
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - Greta Amore
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - Luigi Vetri
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Palermo, Italy
| | - Giuseppe Quatrosi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Palermo, Italy
| | - Anna Cafeo
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - Eloisa Gitto
- Neonatal Intensive Care Unit, Department of Human Pathology of the Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - Antonio Gennaro Nicotera
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - Gabriella Di Rosa
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| |
Collapse
|
4
|
Gheorghe DA, Panouillères MTN, Walsh ND. Investigating the effects of cerebellar transcranial direct current stimulation on saccadic adaptation and cortisol response. CEREBELLUM & ATAXIAS 2021; 8:1. [PMID: 33397502 PMCID: PMC7784285 DOI: 10.1186/s40673-020-00124-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Transcranial Direct Current Stimulation (tDCS) over the prefrontal cortex has been shown to modulate subjective, neuronal and neuroendocrine responses, particularly in the context of stress processing. However, it is currently unknown whether tDCS stimulation over other brain regions, such as the cerebellum, can similarly affect the stress response. Despite increasing evidence linking the cerebellum to stress-related processing, no studies have investigated the hormonal and behavioural effects of cerebellar tDCS. METHODS This study tested the hypothesis of a cerebellar tDCS effect on mood, behaviour and cortisol. To do this we employed a single-blind, sham-controlled design to measure performance on a cerebellar-dependent saccadic adaptation task, together with changes in cortisol output and mood, during online anodal and cathodal stimulation. Forty-five participants were included in the analysis. Stimulation groups were matched on demographic variables, potential confounding factors known to affect cortisol levels, mood and a number of personality characteristics. RESULTS Results showed that tDCS polarity did not affect cortisol levels or subjective mood, but did affect behaviour. Participants receiving anodal stimulation showed an 8.4% increase in saccadic adaptation, which was significantly larger compared to the cathodal group (1.6%). CONCLUSION The stimulation effect on saccadic adaptation contributes to the current body of literature examining the mechanisms of cerebellar stimulation on associated function. We conclude that further studies are needed to understand whether and how cerebellar tDCS may module stress reactivity under challenge conditions.
Collapse
Affiliation(s)
- Delia A. Gheorghe
- School of Psychology, University of East Anglia, Norwich, UK
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Muriel T. N. Panouillères
- Department of Experimental Psychology, University of Oxford, Oxford, UK
- CIAMS, Université Paris-Saclay, 91405 Orsay Cedex, France
- CIAMS, Université d’Orléans, 45067 Orléans, France
| | | |
Collapse
|
5
|
Zhang Y, Zhang H, Wang J, Zhang X, Bu S, Liu X, Wang Q, Lin H. Molecular characterization and expression patterns of glucocorticoid receptor (GR) genes in protandrous hermaphroditic yellowtail clownfish, Amphiprion clarkii. Gene 2020; 745:144651. [PMID: 32259633 DOI: 10.1016/j.gene.2020.144651] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 11/27/2022]
Abstract
Sexual differentiation and ovotestis development are closely associated with cortisol levels, the principal indicator of stress, via the glucocorticoid receptor (GR) in teleosts. Thus, GR is regarded as a mediator to expound the relationship between social stress and gonad development. In the present study, two gr genes (gr1 and gr2) were cloned and analyzed from a protandrous hermaphroditic teleost, the yellowtail clownfish (Amphiprion clarkii). GR1 was found to display a conserved nine-amino-acid insert, WRARQNTDG, between two zinc finger domains. The phylogenetic tree of GR showed that yellowtail clownfish GR1 and GR2 are clustered to teleost GR1 and teleost GR2 separately, and differ from tetrapod GR. The result of real-time PCR revealed that high-level gr1 was mainly distributed in the cerebellum, hypothalamus and heart. The gr2 gene was abundant in the pituitary and liver of females and nonbreeders, while gr2 was mainly detected in the medulla oblongata and middle kidney of males. Moreover, GRs can be expressed in cultured eukaryotic cells and functionally interact with dexamethasone (exogenous glucocorticoid), thereby triggering downstream signaling pathways of different potentials. GR1 and GR2 can be activated by 10 nM dexamethasone treatment in HEK-293T cells. Notably, real-time PCR analysis among three social status groups demonstrated that gr2 expression was the highest in the hypothalamus of nonbreeders, but gr1 was no difference. We speculate that social stress would increase the expression of gr2 gene expression in the hypothalamus to inhibit sexual development. These data provide evidence of social stress involving reproductive regulation, which may help to elucidate the underlying mechanism of sex differentiation and change.
Collapse
Affiliation(s)
- Yanyu Zhang
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan University, Haikou, Hainan 570228, China
| | - Hao Zhang
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan University, Haikou, Hainan 570228, China
| | - Jun Wang
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan University, Haikou, Hainan 570228, China
| | - Xian Zhang
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan University, Haikou, Hainan 570228, China
| | - Shaoyang Bu
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan University, Haikou, Hainan 570228, China
| | - Xiaochun Liu
- Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Qian Wang
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan University, Haikou, Hainan 570228, China.
| | - Haoran Lin
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan University, Haikou, Hainan 570228, China; Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| |
Collapse
|
6
|
Wolford E, Lahti-Pulkkinen M, Girchenko P, Lipsanen J, Tuovinen S, Lahti J, Heinonen K, Hämäläinen E, Kajantie E, Pesonen AK, Villa PM, Laivuori H, Reynolds RM, Räikkönen K. Associations of antenatal glucocorticoid exposure with mental health in children. Psychol Med 2020; 50:247-257. [PMID: 30688183 DOI: 10.1017/s0033291718004129] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Synthetic glucocorticoids, to enhance fetal maturation, are a standard treatment when preterm birth before 34 gestational weeks is imminent. While morbidity- and mortality-related benefits may outweigh potential neurodevelopmental harms in children born preterm (<37 gestational weeks), this may not hold true when pregnancy continues to term (⩾37 gestational weeks). We studied the association of antenatal betamethasone exposure on child mental health in preterm and term children. METHODS We included 4708 women and their children, born 2006-2010, from the Prediction and Prevention of Pre-eclampsia and Intrauterine Growth Restriction Study with information on both antenatal betamethasone treatment and child mental and behavioral disorders from the Finnish Hospital Discharge Register from the child's birth to 31 December 2016. Additional follow-up data on mother-reported psychiatric problems and developmental milestones were available for 2640 children at 3.5 (s.d. = 0.07) years-of-age. RESULTS Of the children, 187 were born preterm (61 betamethasone-exposed) and 4521 at term (56 betamethasone-exposed). The prevalence of any mental and behavioral, psychological development, emotional and behavioral, and comorbid disorders was higher in the betamethasone-exposed, compared to non-exposed children [odds ratio 2.76 (95% confidence interval 1.76-4.32), 3.61 (2.19-5.95), 3.29 (1.86-5.82), and 6.04 (3.25-11.27), respectively]. Levels of psychiatric problems and prevalence of failure to meet the age-appropriate development in personal-social skills were also higher in mother-reports of betamethasone-exposed children. These associations did not vary significantly between preterm and term children. CONCLUSIONS Antenatal betamethasone exposure may be associated with mental health problems in children born preterm and in those who end up being born at term.
Collapse
Affiliation(s)
- Elina Wolford
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Marius Lahti-Pulkkinen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Polina Girchenko
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jari Lipsanen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Soile Tuovinen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jari Lahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Helsinki Collegium for Advanced Studies, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Centre, Helsinki, Finland
| | - Kati Heinonen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Esa Hämäläinen
- Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland
| | - Eero Kajantie
- National Institute for Health and Welfare, Helsinki, Finland
- Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Anu-Katriina Pesonen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pia M Villa
- Obstetrics and Gynaecology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Hannele Laivuori
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Rebecca M Reynolds
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
7
|
Matthews LG, Inder TE, Pascoe L, Kapur K, Lee KJ, Monson BB, Doyle LW, Thompson DK, Anderson PJ. Longitudinal Preterm Cerebellar Volume: Perinatal and Neurodevelopmental Outcome Associations. CEREBELLUM (LONDON, ENGLAND) 2018; 17:610-627. [PMID: 29949094 PMCID: PMC6126980 DOI: 10.1007/s12311-018-0946-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Impaired cerebellar development is an important determinant of adverse motor and cognitive outcomes in very preterm (VPT) infants. However, longitudinal MRI studies investigating cerebellar maturation from birth through childhood and associated neurodevelopmental outcomes are lacking. We aimed to compare cerebellar volume and growth from term-equivalent age (TEA) to 7 years between VPT (< 30 weeks' gestation or < 1250 g) and full-term children; and to assess the association between these measures, perinatal factors, and 7-year outcomes in VPT children, and whether these relationships varied by sex. In a prospective cohort study of 224 VPT and 46 full-term infants, cerebellar volumes were measured on MRI at TEA and 7 years. Useable data at either time-point were collected for 207 VPT and 43 full-term children. Cerebellar growth from TEA to 7 years was compared between VPT and full-term children. Associations with perinatal factors and 7-year outcomes were investigated in VPT children. VPT children had smaller TEA and 7-year volumes and reduced growth. Perinatal factors were associated with smaller cerebellar volume and growth between TEA and 7 years, namely, postnatal corticosteroids for TEA volume, and female sex, earlier birth gestation, white and deep nuclear gray matter injury for 7-year volume and growth. Smaller TEA and 7-year volumes, and reduced growth were associated with poorer 7-year IQ, language, and motor function, with differential relationships observed for male and female children. Our findings indicate that cerebellar growth from TEA to 7 years is impaired in VPT children and relates to early perinatal factors and 7-year outcomes.
Collapse
Affiliation(s)
- Lillian G Matthews
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Ave, Boston, MA, 02115, USA.
- Murdoch Children's Research Institute, Melbourne, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, Australia.
| | - T E Inder
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Ave, Boston, MA, 02115, USA
| | - L Pascoe
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - K Kapur
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - K J Lee
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - B B Monson
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Ave, Boston, MA, 02115, USA
- Department of Speech and Hearing Science, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - L W Doyle
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Royal Women's Hospital, Melbourne, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Australia
| | - D K Thompson
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - P J Anderson
- Murdoch Children's Research Institute, Melbourne, Australia
- Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Melbourne, Australia
| |
Collapse
|
8
|
Webb EA, Elliott L, Carlin D, Wilson M, Hall K, Netherton J, Reed J, Barrett TG, Salwani V, Clayden JD, Arlt W, Krone N, Peet AC, Wood AG. Quantitative Brain MRI in Congenital Adrenal Hyperplasia: In Vivo Assessment of the Cognitive and Structural Impact of Steroid Hormones. J Clin Endocrinol Metab 2018; 103:1330-1341. [PMID: 29165577 PMCID: PMC6018658 DOI: 10.1210/jc.2017-01481] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/09/2017] [Indexed: 11/19/2022]
Abstract
Context Brain white matter hyperintensities are seen on routine clinical imaging in 46% of adults with congenital adrenal hyperplasia (CAH). The extent and functional relevance of these abnormalities have not been studied with quantitative magnetic resonance imaging (MRI) analysis. Objective To examine white matter microstructure, neural volumes, and central nervous system (CNS) metabolites in CAH due to 21-hydroxylase deficiency (21OHD) and to determine whether identified abnormalities are associated with cognition, glucocorticoid, and androgen exposure. Design, Setting, and Participants A cross-sectional study at a tertiary hospital including 19 women (18 to 50 years) with 21OHD and 19 age-matched healthy women. Main Outcome Measure Recruits underwent cognitive assessment and brain imaging, including diffusion weighted imaging of white matter, T1-weighted volumetry, and magnetic resonance spectroscopy for neural metabolites. We evaluated white matter microstructure by using tract-based spatial statistics. We compared cognitive scores, neural volumes, and metabolites between groups and relationships between glucocorticoid exposure, MRI, and neurologic outcomes. Results Patients with 21OHD had widespread reductions in white matter structural integrity, reduced volumes of right hippocampus, bilateral thalami, cerebellum, and brainstem, and reduced mesial temporal lobe total choline content. Working memory, processing speed, and digit span and matrix reasoning scores were reduced in patients with 21OHD, despite similar education and intelligence to controls. Patients with 21OHD exposed to higher glucocorticoid doses had greater abnormalities in white matter microstructure and cognitive performance. Conclusion We demonstrate that 21OHD and current glucocorticoid replacement regimens have a profound impact on brain morphology and function. If reversible, these CNS markers are a potential target for treatment.
Collapse
Affiliation(s)
- Emma A Webb
- Department of Endocrinology & Diabetes, Birmingham Children’s Hospital, Birmingham, United Kingdom
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
- University of East Anglia, Faculty of Medical and Health Sciences, Norwich, Norfolk, United Kingdom
| | - Lucy Elliott
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Dominic Carlin
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Martin Wilson
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham, United Kingdom
| | - Kirsty Hall
- School of Psychology, University of Birmingham, Birmingham, United Kingdom
| | - Jennifer Netherton
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Department of Psychology, Birmingham Children’s Hospital, Birmingham, United Kingdom
| | - Julie Reed
- Department of Psychology, Birmingham Children’s Hospital, Birmingham, United Kingdom
| | - Tim G Barrett
- Department of Endocrinology & Diabetes, Birmingham Children’s Hospital, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Vijay Salwani
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Department of Radiology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Jon D Clayden
- Developmental Imaging & Biophysics Section, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Nils Krone
- Academic Unit of Child Health, Department of Oncology & Metabolism, University of Sheffield, Sheffield Children’s Hospital, Sheffield, United Kingdom
| | - Andrew C Peet
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Amanda G Wood
- School Life and Health Sciences & Aston Brain Centre, Aston University, Birmingham, United Kingdom
- Child Neuropsychology, Clinical Sciences, Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
| |
Collapse
|
9
|
Rensel MA, Ding JA, Pradhan DS, Schlinger BA. 11β-HSD Types 1 and 2 in the Songbird Brain. Front Endocrinol (Lausanne) 2018; 9:86. [PMID: 29593652 PMCID: PMC5857549 DOI: 10.3389/fendo.2018.00086] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/23/2018] [Indexed: 12/29/2022] Open
Abstract
Glucocorticoid (GC) hormones act on the brain to regulate diverse functions, from behavior and homeostasis to the activity of the hypothalamic-pituitary-adrenal axis. Local regeneration and metabolism of GCs can occur in target tissues through the actions of the 11β-hydroxysteroid dehydrogenases [11 beta-hydroxysteroid dehydrogenase type 1 (11β-HSD1) and 11 beta-hydroxysteroid dehydrogenase type 2 (11β-HSD2), respectively] to regulate access to GC receptors. Songbirds have become especially important model organisms for studies of stress hormone action; however, there has been little focus on neural GC metabolism. Therefore, we tested the hypothesis that 11β-HSD1 and 11β-HSD2 are expressed in GC-sensitive regions of the songbird brain. Localization of 11β-HSD expression in these regions could provide precise temporal and spatial control over GC actions. We quantified GC sensitivity in zebra finch (Taeniopygia guttata) brain by measuring glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) expression across six regions, followed by quantification of 11β-HSD1 and 11β-HSD2 expression. We detected GR, MR, and 11β-HSD2 mRNA expression throughout the adult brain. Whereas 11β-HSD1 expression was undetectable in the adult brain, we detected low levels of expression in the brain of developing finches. Across several adult brain regions, expression of 11β-HSD2 covaried with GR and MR, with the exception of the cerebellum and hippocampus. It is possible that receptors in these latter two regions require direct access to systemic GC levels. Overall, these results suggest that 11β-HSD2 expression protects the adult songbird brain by rapid metabolism of GCs in a context and region-specific manner.
Collapse
Affiliation(s)
- Michelle A. Rensel
- The Institute for Society and Genetics, University of California, Los Angeles, Los Angeles, CA, United States
- Laboratory of Neuroendocrinology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jessica A. Ding
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Devaleena S. Pradhan
- Laboratory of Neuroendocrinology, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Barney A. Schlinger
- Laboratory of Neuroendocrinology, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
10
|
Marshall NA, Marusak HA, Sala-Hamrick KJ, Crespo LM, Rabinak CA, Thomason ME. Socioeconomic disadvantage and altered corticostriatal circuitry in urban youth. Hum Brain Mapp 2018; 39:1982-1994. [PMID: 29359526 DOI: 10.1002/hbm.23978] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 01/04/2018] [Accepted: 01/08/2018] [Indexed: 12/13/2022] Open
Abstract
Socioeconomic disadvantage (SED) experienced in early life is linked to a range of risk behaviors and diseases. Neuroimaging research indicates that this association is mediated by functional changes in corticostriatal reward systems that modulate goal-directed behavior, reward evaluation, and affective processing. Existing research has focused largely on adults and within-household measures as an index of SED, despite evidence that broader community-level SED (e.g., neighborhood poverty levels) has significant and sometimes distinct effects on development and health outcomes. Here, we test effects of both household- and community-level SED on resting-state functional connectivity (rsFC) of the ventral striatum (VS) in 100 racially and economically diverse children and adolescents (ages 6-17). We observed unique effects of household income and community SED on VS circuitry such that higher community SED was associated with reduced rsFC between the VS and an anterior region of the medial prefrontal cortex (mPFC), whereas lower household income was associated with increased rsFC between the VS and the cerebellum, inferior temporal lobe, and lateral prefrontal cortex. Lower VS-mPFC rsFC was also associated with higher self-reported anxiety symptomology, and rsFC mediated the link between community SED and anxiety. These results indicate unique effects of community-level SED on corticostriatal reward circuitry that can be detected in early life, which carries implications for future interventions and targeted therapies. In addition, our findings raise intriguing questions about the distinct pathways through which specific sources of SED can affect brain and emotional development.
Collapse
Affiliation(s)
- Narcis A Marshall
- Department of Psychology, University of Southern California, Los Angeles, California
| | - Hilary A Marusak
- Department of Pharmacy Practice, Wayne State University School of Medicine, Detroit, Michigan
| | - Kelsey J Sala-Hamrick
- Department of Psychology, University of Southern California, Los Angeles, California.,Department of Psychology, Wayne State University, Detroit, Michigan
| | - Laura M Crespo
- Department of Psychology, University of Southern California, Los Angeles, California.,Department of Psychology, Wayne State University, Detroit, Michigan
| | - Christine A Rabinak
- Department of Pharmacy Practice, Wayne State University School of Medicine, Detroit, Michigan
| | - Moriah E Thomason
- Department of Pediatrics Wayne State University School of Medicine, Detroit, Michigan.,Merrill Palmer Skillman Institute for Child and Family Development Wayne State University, Detroit, Michigan.,Perinatology Research Branch, NICHD/NIH/DHHS, Detroit, MI, Bethesda, Maryland
| |
Collapse
|
11
|
Sveinsdóttir K, Ley D, Hövel H, Fellman V, Hüppi PS, Smith LEH, Hellström A, Hansen Pupp I. Relation of Retinopathy of Prematurity to Brain Volumes at Term Equivalent Age and Developmental Outcome at 2 Years of Corrected Age in Very Preterm Infants. Neonatology 2018; 114:46-52. [PMID: 29649829 PMCID: PMC5997524 DOI: 10.1159/000487847] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 02/17/2018] [Indexed: 01/28/2023]
Abstract
BACKGROUND Retinopathy of prematurity (ROP) is a major complication of preterm birth and has been associated with later visual and nonvisual impairments. OBJECTIVES To evaluate relationships between any stage of ROP, brain volumes, and developmental outcomes. METHODS This study included 52 very preterm infants (gestational age [mean ± SD]: 26.4 ± 1.9 weeks). Total brain, gray matter, unmyelinated white matter (UWMV), and cerebellar volumes were estimated in 51 out of 52 infants by magnetic resonance imaging at term-equivalent age. Bayley Scales of Infant Development were used to assess developmental outcomes in 49 out of 52 infants at a mean corrected age of 24.6 months. RESULTS Nineteen out of 52 infants developed any stage of ROP. Infants with ROP had a lower median (IQR) UWMV (173 [156-181] vs. 204 [186-216] mL, p < 0.001) and cerebellar volume (18.3 [16.5-20] vs. 22.3 [20.3-24.7] mL, p < 0.001) than infants without ROP. They also had a lower median (IQR) mental developmental index (72 [56-83] vs. 100 [88-104], p < 0.001) and a lower psychomotor developmental index (80 [60-85] vs. 92 [81-103], p = 0.002). Brain volumes and developmental outcomes did not differ among infants with different stages of ROP. CONCLUSION Any stage of ROP in preterm infants was associated with a reduced brain volume and an impaired developmental outcome. These results suggest that common pathways may lead to impaired neural and neurovascular development in the brain and retina and that all stages of ROP may be considered in future studies on ROP and development.
Collapse
Affiliation(s)
- Kristbjörg Sveinsdóttir
- Division of Pediatrics, Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
| | - David Ley
- Division of Pediatrics, Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
| | - Holger Hövel
- Division of Pediatrics, Department of Clinical Sciences, Central Hospital Kristianstad, Lund, Sweden
| | - Vineta Fellman
- Division of Pediatrics, Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden.,Children's Hospital, University of Helsinki, Helsinki, Finland
| | - Petra S Hüppi
- Division of Development and Growth, Department of Pediatrics, University Hospital of Geneva, Geneva, Switzerland
| | - Lois E H Smith
- Department of Opthalmology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ann Hellström
- Sahlgrenska Center for Pediatric Ophthalmology Research, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Ingrid Hansen Pupp
- Division of Pediatrics, Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
12
|
Abstract
Although preterm birth is best known to result in adverse neurodevelopmental outcomes through injury of the supratentorial structures, including intraventricular hemorrhage and periventricular leukomalacia, the cerebellum has become increasingly recognized as an important target for injury and adverse motor and cognitive outcomes. Undergoing the most dramatic growth during the preterm period, the cerebellum is vulnerable to large and small hemorrhages, as well as hypoplasia resulting from a number of potentially modifiable risk factors. These factors include contact with intraventricular blood, crossed cerebrocerebellar diaschisis, postnatal glucocorticoid exposure, pain and opioid exposure, nutrition and somatic growth, cardiorespiratory factors, and socioeconomic status. Strategies targeting these factors may result in prevention of the motor and cognitive deficits seen after cerebellar hemorrhage or hypoplasia.
Collapse
Affiliation(s)
- Emily W Y Tam
- Division of Neurology, Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, Canada.
| |
Collapse
|
13
|
Bennett GA, Palliser HK, Shaw JC, Palazzi KL, Walker DW, Hirst JJ. Maternal stress in pregnancy affects myelination and neurosteroid regulatory pathways in the guinea pig cerebellum. Stress 2017; 20:580-588. [PMID: 28969480 DOI: 10.1080/10253890.2017.1378637] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Prenatal stress predisposes offspring to behavioral pathologies. These may be attributed to effects on cerebellar neurosteroids and GABAergic inhibitory signaling, which can be linked to hyperactivity disorders. The aims were to determine the effect of prenatal stress on markers of cerebellar development, a key enzyme in neurosteroid synthesis and the expression of GABAA receptor (GABAAR) subunits involved in neurosteroid signaling. We used a model of prenatal stress (strobe light exposure, 2 h on gestational day 50, 55, 60 and 65) in guinea pigs, in which we have characterized anxiety and neophobic behavioral outcomes. The cerebellum and plasma were collected from control and prenatally stressed offspring at term (control fetus: n = 9 male, n = 7 female; stressed fetus: n = 7 male, n = 8 female) and postnatal day (PND) 21 (control: n = 8 male, n = 8 female; stressed: n = 9 male, n = 6 female). We found that term female offspring exposed to prenatal stress showed decreased expression of mature oligodendrocytes (∼40% reduction) and these deficits improved to control levels by PND21. Reactive astrocyte expression was lower (∼40% reduction) following prenatal stress. GABAAR subunit (δ and α6) expression and circulating allopregnanolone concentrations were not affected by prenatal stress. Prenatal stress increased expression (∼150-250% increase) of 5α-reductase type-1 mRNA in the cerebellum, which may be a neuroprotective response to promote GABAergic inhibition and aid in repair. These observations indicate that prenatal stress exposure has marked effects on the development of the cerebellum. These findings suggest cerebellar changes after prenatal stress may contribute to adverse behavioral outcomes after exposure to these stresses.
Collapse
Affiliation(s)
- Greer A Bennett
- a Mothers and Babies Research Centre , Hunter Medical Research Institute , Newcastle , New South Wales , Australia
- b School of Biomedical Sciences and Pharmacy , University of Newcastle , New South Wales , Australia
| | - Hannah K Palliser
- a Mothers and Babies Research Centre , Hunter Medical Research Institute , Newcastle , New South Wales , Australia
- b School of Biomedical Sciences and Pharmacy , University of Newcastle , New South Wales , Australia
| | - Julia C Shaw
- a Mothers and Babies Research Centre , Hunter Medical Research Institute , Newcastle , New South Wales , Australia
- b School of Biomedical Sciences and Pharmacy , University of Newcastle , New South Wales , Australia
| | - Kerrin L Palazzi
- c Clinical Research Design , Information Technology and Statistical Support (CReDITSS), Hunter Medical Research Institute (HMRI) , Newcastle , New South Wales , Australia
| | - David W Walker
- d School of Health and Biomedical Sciences , RMIT University , Bundoora , Victoria , Australia
| | - Jonathan J Hirst
- a Mothers and Babies Research Centre , Hunter Medical Research Institute , Newcastle , New South Wales , Australia
- b School of Biomedical Sciences and Pharmacy , University of Newcastle , New South Wales , Australia
| |
Collapse
|
14
|
Austdal LPE, Bjørnstad S, Mathisen GH, Aden PK, Mikkola I, Paulsen RE, Rakkestad KE. Glucocorticoid Effects on Cerebellar Development in a Chicken Embryo Model: Exploring Changes in PAX6 and Metalloproteinase-9 After Exposure to Dexamethasone. J Neuroendocrinol 2016; 28. [PMID: 27791298 DOI: 10.1111/jne.12438] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 10/25/2016] [Accepted: 10/25/2016] [Indexed: 12/22/2022]
Abstract
The developing cerebellum is vulnerable to effects of glucocorticoids and cerebellar dysfunction is associated with neurodevelopmental disorders (e.g. autism). Transcription factor PAX6 and matrix metalloproteinase-9 (MMP-9) are critical for normal cerebellar development and are highly expressed in migrating neurones. Alterations in MMP-9 and PAX6 are associated with altered cerebellar development. In the present study, we characterised the growth rate and development of the cortical layers, and further investigated how the levels of PAX6 and MMP-9, as well as glucocorticoid receptor (GR) and proliferating cell nuclear antigen (PCNA), change in the cerebellum during the foetal period [embryonic day (E)12-21] in chicken, which corresponds to the human perinatal period. Dexamethasone (DEX) was administered in ovo at E13 and E16, aiming to investigate how prenatal exposure to glucocorticoids interferes with normal development. DEX reduced foetal and cerebellar weight at E17 in a dose-dependent manner linked to a reduced level of PCNA and, over time, down-regulation of GR. We report that promoter activity of PAX6 and MMP-9 increased as a result of GR-stimulation in vitro. Prenatal DEX increased the protein level of PAX6 in a transient manner. PAX6 is reduced in mature granule neurones, and this occurred earlier in embryos exposed to DEX than in non-exposed controls. DEX exposure also led to a slow-onset down-regulation of MMP-9. Taken together, these findings indicate that excess prenatal glucocorticoid stimulation disturbs normal development of the cerebellum through mechanisms associated with reduced proliferation and accelerated maturation where PAX6 and MMP-9 play important roles.
Collapse
Affiliation(s)
- L P E Austdal
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - S Bjørnstad
- Department of Pathology, Oslo University Hospital - Ullevål, Oslo, Norway
| | - G H Mathisen
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - P K Aden
- Department of Neurosciences for Children, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - I Mikkola
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - R E Paulsen
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - K E Rakkestad
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| |
Collapse
|
15
|
Teicher MH, Samson JA. Annual Research Review: Enduring neurobiological effects of childhood abuse and neglect. J Child Psychol Psychiatry 2016; 57:241-66. [PMID: 26831814 PMCID: PMC4760853 DOI: 10.1111/jcpp.12507] [Citation(s) in RCA: 650] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/24/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Childhood maltreatment is the most important preventable cause of psychopathology accounting for about 45% of the population attributable risk for childhood onset psychiatric disorders. A key breakthrough has been the discovery that maltreatment alters trajectories of brain development. METHODS This review aims to synthesize neuroimaging findings in children who experienced caregiver neglect as well as from studies in children, adolescents and adults who experienced physical, sexual and emotional abuse. In doing so, we provide preliminary answers to questions regarding the importance of type and timing of exposure, gender differences, reversibility and the relationship between brain changes and psychopathology. We also discuss whether these changes represent adaptive modifications or stress-induced damage. RESULTS Parental verbal abuse, witnessing domestic violence and sexual abuse appear to specifically target brain regions (auditory, visual and somatosensory cortex) and pathways that process and convey the aversive experience. Maltreatment is associated with reliable morphological alterations in anterior cingulate, dorsal lateral prefrontal and orbitofrontal cortex, corpus callosum and adult hippocampus, and with enhanced amygdala response to emotional faces and diminished striatal response to anticipated rewards. Evidence is emerging that these regions and interconnecting pathways have sensitive exposure periods when they are most vulnerable. CONCLUSIONS Early deprivation and later abuse may have opposite effects on amygdala volume. Structural and functional abnormalities initially attributed to psychiatric illness may be a more direct consequence of abuse. Childhood maltreatment exerts a prepotent influence on brain development and has been an unrecognized confound in almost all psychiatric neuroimaging studies. These brain changes may be best understood as adaptive responses to facilitate survival and reproduction in the face of adversity. Their relationship to psychopathology is complex as they are discernible in both susceptible and resilient individuals with maltreatment histories. Mechanisms fostering resilience will need to be a primary focus of future studies.
Collapse
Affiliation(s)
- Martin H. Teicher
- Department of Psychiatry, Harvard Medical School, Boston, MA,Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA
| | - Jacqueline A. Samson
- Department of Psychiatry, Harvard Medical School, Boston, MA,Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA
| |
Collapse
|
16
|
Klein JL, Lemmon ME, Northington FJ, Boltshauser E, Huisman TAGM, Poretti A. Clinical and neuroimaging features as diagnostic guides in neonatal neurology diseases with cerebellar involvement. CEREBELLUM & ATAXIAS 2016; 3:1. [PMID: 26770813 PMCID: PMC4712469 DOI: 10.1186/s40673-016-0039-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 12/10/2015] [Indexed: 02/07/2023]
Abstract
Cerebellar abnormalities are encountered in a high number of neurological diseases that present in the neonatal period. These disorders can be categorized broadly as inherited (e.g. malformations, inborn errors of metabolism) or acquired (e.g. hemorrhages, infections, stroke). In some disorders such as Dandy-Walker malformation or Joubert syndrome, the main abnormalities are located within the cerebellum and brainstem. In other disorders such as Krabbe disease or sulfite oxidase deficiency, the main abnormalities are found within the supratentorial brain, but the cerebellar involvement may be helpful for diagnostic purposes. In In this article, we review neurological disorders with onset in the neonatal period and cerebellar involvement with a focus on how characterization of cerebellar involvement can facilitate accurate diagnosis and improved accuracy of neuro-functional prognosis.
Collapse
Affiliation(s)
- Jessica L Klein
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD USA ; Neurosciences Intensive Care Nursery Program, The Johns Hopkins University School of Medicine, Baltimore, MD USA ; Department of Pediatrics, Medical University of South Carolina, Charleston, SC USA
| | - Monica E Lemmon
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD USA ; Neurosciences Intensive Care Nursery Program, The Johns Hopkins University School of Medicine, Baltimore, MD USA ; Division of Pediatric Neurology, Department of Pediatrics, Duke University School of Medicine, Durham, NC USA
| | - Frances J Northington
- Neurosciences Intensive Care Nursery Program, The Johns Hopkins University School of Medicine, Baltimore, MD USA ; Division of Neonatology, Department of Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Eugen Boltshauser
- Division of Pediatric Neurology, University Children's Hospital, Zurich, Switzerland
| | - Thierry A G M Huisman
- Neurosciences Intensive Care Nursery Program, The Johns Hopkins University School of Medicine, Baltimore, MD USA ; Section of Pediatric Neuroradiology, Division of Pediatric Radiology, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Charlotte R. Bloomberg Children's Center, Sheikh Zayed Tower, Room 4174, 1800 Orleans Street, Baltimore, MD USA
| | - Andrea Poretti
- Neurosciences Intensive Care Nursery Program, The Johns Hopkins University School of Medicine, Baltimore, MD USA ; Division of Pediatric Neurology, University Children's Hospital, Zurich, Switzerland ; Section of Pediatric Neuroradiology, Division of Pediatric Radiology, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Charlotte R. Bloomberg Children's Center, Sheikh Zayed Tower, Room 4174, 1800 Orleans Street, Baltimore, MD USA
| |
Collapse
|
17
|
Elsey J, Coates A, Lacadie CM, McCrory EJ, Sinha R, Mayes LC, Potenza MN. Childhood trauma and neural responses to personalized stress, favorite-food and neutral-relaxing cues in adolescents. Neuropsychopharmacology 2015; 40:1580-9. [PMID: 25567424 PMCID: PMC4915266 DOI: 10.1038/npp.2015.6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 12/30/2014] [Accepted: 12/30/2014] [Indexed: 01/17/2023]
Abstract
Previous studies have found childhood trauma to be associated with functional and structural abnormalities in corticostriatal-limbic brain regions, which may explain the associations between trauma and negative mental and physical health outcomes. However, functional neuroimaging of maltreatment-related trauma has been limited by largely using generic and predominantly aversive stimuli. Personalized stress, favorite-food, and neutral/relaxing cues during functional magnetic resonance imaging were used to probe the neural correlates of emotional/motivational states in adolescents with varying exposure to maltreatment-related trauma. Sixty-four adolescents were stratified into high- or low-trauma-exposed groups. Cue-related measures of subjective anxiety and craving were collected. Relative to the low-trauma-exposed group, high-trauma-exposed adolescents displayed an increased activation of insula, anterior cingulate, and prefrontal cortex in response to stress cues. Activation in subcortical structures, including the hippocampus, was inversely correlated with subjective anxiety in the high- but not the low-trauma-exposed group. The high-trauma-exposed group displayed hypoactivity of cerebellar regions in response to neutral/relaxing cues. No group differences were observed in response to favorite-food cues. The relationship between trauma exposure and altered cortico-limbic circuitry may in part explain the association between childhood trauma and heightened vulnerability to emotional disturbances and risky behaviour. This may be particularly pertinent during adolescence when such difficulties often emerge. Further work is needed to elucidate the mechanism linking trauma to obesity.
Collapse
Affiliation(s)
- James Elsey
- Child Study Center, Yale University, New Haven, CT, USA,Anna Freud Centre, University College London, London, UK
| | - Alice Coates
- Child Study Center, Yale University, New Haven, CT, USA,Anna Freud Centre, University College London, London, UK
| | - Cheryl M Lacadie
- Department of Diagnostic Radiology, Yale University, New Haven, CT, USA
| | - Eamon J McCrory
- Division of Psychology and Language Sciences, University College London, London, UK
| | - Rajita Sinha
- Department of Psychiatry, Yale University, New Haven, CT, USA,Department of Neurobiology, Yale University, New Haven, CT, USA
| | - Linda C Mayes
- Child Study Center, Yale University, New Haven, CT, USA,Anna Freud Centre, University College London, London, UK
| | - Marc N Potenza
- Child Study Center, Yale University, New Haven, CT, USA,Department of Psychiatry, Yale University, New Haven, CT, USA,Department of Neurobiology, Yale University, New Haven, CT, USA,Connecticut Mental Health Center, New Haven, CT, USA,Yale University School of Medicine, Departments of Psychiatry, Child Study, and Neurobiology, 34 Park Street, New Haven, CT, 06519, USA, Tel: +1 203 737 3553, Fax: +1 203 737 3591, E-mail:
| |
Collapse
|
18
|
Noguchi KK. Glucocorticoid Induced Cerebellar Toxicity in the Developing Neonate: Implications for Glucocorticoid Therapy during Bronchopulmonary Dysplasia. Cells 2014; 3:36-52. [PMID: 24501683 PMCID: PMC3910303 DOI: 10.3390/cells3010036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Prematurely born infants commonly suffer respiratory dysfunction due to the immature state of their lungs. As a result, clinicians often administer glucocorticoid (GC) therapy to accelerate lung maturation and reduce inflammation. Unfortunately, several studies have found GC therapy can also produce neuromotor/cognitive deficits and selectively stunt the cerebellum. However, despite its continued use, relatively little is known about how exposure to this hormone might produce neurodevelopmental deficits. In this review, we use rodent and human research to provide evidence that GC therapy may disrupt cerebellar development through the rapid induction of apoptosis in the cerebellar external granule layer (EGL). The EGL is a transient proliferative region responsible for the production of over 90% of the neurons in the cerebellum. During normal development, endogenous GC stimulation is thought to selectively signal the elimination of the EGL once production of new neurons is complete. As a result, GC therapy may precociously eliminate the EGL before it can produce enough neurons for normal cerebellar function. It is hoped that this review may provide information for future clinical research in addition to translational guidance for the safer use of GC therapy.
Collapse
Affiliation(s)
- Kevin K Noguchi
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, 660 South Euclid, Box #8134, St. Louis, MO 63110, USA; Tel.: +1-314-362-7007
| |
Collapse
|
19
|
Cheong JL, Burnett AC, Lee KJ, Roberts G, Thompson DK, Wood SJ, Connelly A, Anderson PJ, Doyle LW. Association between postnatal dexamethasone for treatment of bronchopulmonary dysplasia and brain volumes at adolescence in infants born very preterm. J Pediatr 2014; 164:737-743.e1. [PMID: 24332820 PMCID: PMC4029072 DOI: 10.1016/j.jpeds.2013.10.083] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 09/23/2013] [Accepted: 10/29/2013] [Indexed: 12/03/2022]
Abstract
OBJECTIVES To compare brain volumes in adolescents who were born extremely preterm (<28 weeks gestation) who had received postnatal dexamethasone, and to determine if there was a postnatal dexamethasone dose-response effect on brain volumes. STUDY DESIGN Geographical cohort study of extremely preterm adolescents born in 1991-1992 in Victoria, Australia. T1-weighted magnetic resonance imaging was performed at 18 years of age. Segmented and parcellated brain volumes were calculated using an automated segmentation method (FreeSurfer) and compared between groups, with and without adjustment for potential confounders. The relationships between total postnatal dexamethasone dose and brain volumes were explored using linear regression. RESULTS Of the 148 extremely preterm participants, 55 (37%) had received postnatal dexamethasone, with a cumulative mean dose of 7.7 mg/kg. Compared with participants who did not receive postnatal dexamethasone, those who did had smaller total brain tissue volumes (mean difference -3.6%, 95% CI [-7.0%, -0.3%], P value = .04) and smaller white matter, thalami, and basal ganglia volumes (all P < .05). There was a trend of smaller total brain and white matter volumes with increasing dose of postnatal dexamethasone (regression coefficient -7.7 [95% CI -16.2, 0.8] and -3.2 [-6.6, 0.2], respectively). CONCLUSIONS Extremely preterm adolescents who received postnatal dexamethasone in the newborn period had smaller total brain tissue volumes than those who did not receive postnatal dexamethasone, particularly white matter, thalami, and basal ganglia. Vulnerability of brain tissues or structures associated with postnatal dexamethasone varies by structure and persists into adolescence.
Collapse
Affiliation(s)
- Jeanie L.Y. Cheong
- Neonatal Services, Royal Women's Hospital, Melbourne, Australia,Victorian Infant Brain Studies, Murdoch Childrens Research Institute, Melbourne, Australia,Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Australia,Reprint requests: Jeanie L. Y. Cheong, MD, Level 7, Newborn Research, Royal Women's Hospital, 20 Flemington Road, Parkville 3052, Australia.
| | - Alice C. Burnett
- Victorian Infant Brain Studies, Murdoch Childrens Research Institute, Melbourne, Australia
| | - Katherine J. Lee
- Victorian Infant Brain Studies, Murdoch Childrens Research Institute, Melbourne, Australia,Department of Pediatrics, University of Melbourne, Melbourne, Australia,Clinical Epidemiology and Biostatistics, Murdoch Childrens Research Institute, Melbourne, Australia
| | - Gehan Roberts
- Department of Pediatrics, University of Melbourne, Melbourne, Australia,Royal Children's Hospital, Melbourne, Australia
| | - Deanne K. Thompson
- Victorian Infant Brain Studies, Murdoch Childrens Research Institute, Melbourne, Australia,Developmental Imaging, Murdoch Childrens Research Institute, Melbourne, Australia,Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Stephen J. Wood
- School of Psychology, University of Birmingham, Edgbaston, United Kingdom
| | - Alan Connelly
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Peter J. Anderson
- Victorian Infant Brain Studies, Murdoch Childrens Research Institute, Melbourne, Australia,Department of Pediatrics, University of Melbourne, Melbourne, Australia
| | - Lex W. Doyle
- Neonatal Services, Royal Women's Hospital, Melbourne, Australia,Victorian Infant Brain Studies, Murdoch Childrens Research Institute, Melbourne, Australia,Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Australia
| | | |
Collapse
|
20
|
Walsh ND, Dalgleish T, Lombardo MV, Dunn VJ, Van Harmelen AL, Ban M, Goodyer IM. General and specific effects of early-life psychosocial adversities on adolescent grey matter volume. NEUROIMAGE-CLINICAL 2014; 4:308-18. [PMID: 25061568 PMCID: PMC4107373 DOI: 10.1016/j.nicl.2014.01.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 01/03/2014] [Accepted: 01/03/2014] [Indexed: 12/16/2022]
Abstract
Exposure to childhood adversities (CA) is associated with subsequent alterations in regional brain grey matter volume (GMV). Prior studies have focused mainly on severe neglect and maltreatment. The aim of this study was to determine in currently healthy adolescents if exposure to more common forms of CA results in reduced GMV. Effects on brain structure were investigated using voxel-based morphometry in a cross-sectional study of youth recruited from a population-based longitudinal cohort. 58 participants (mean age = 18.4) with (n = 27) or without (n = 31) CA exposure measured retrospectively from maternal interview were included in the study. Measures of recent negative life events (RNLE) recorded at 14 and 17 years, current depressive symptoms, gender, participant/parental psychiatric history, current family functioning perception and 5-HTTLPR genotype were covariates in analyses. A multivariate analysis of adversities demonstrated a general association with a widespread distributed neural network consisting of cortical midline, lateral frontal, temporal, limbic, and cerebellar regions. Univariate analyses showed more specific associations between adversity measures and regional GMV: CA specifically demonstrated reduced vermis GMV and past psychiatric history with reduced medial temporal lobe volume. In contrast RNLE aged 14 was associated with increased lateral cerebellar and anterior cingulate GMV. We conclude that exposure to moderate levels of childhood adversities occurring during childhood and early adolescence exerts effects on the developing adolescent brain. Reducing exposure to adverse social environments during early life may optimize typical brain development and reduce subsequent mental health risks in adult life. Combined psychosocial factors broadly affect brain grey matter volume (GMV). Specific psychosocial risk factors exert specific effects on brain GMV. Exposure to childhood adversities reduces medial cerebellar and vermal GMV. A subsequent psychiatric history is associated with reduced temporal lobe GMV. Exposure to negative life events aged 14 is associated with increased regional GMV.
Collapse
Affiliation(s)
- Nicholas D Walsh
- Developmental Psychiatry Section, Department of Psychiatry, University of Cambridge, Cambridge, UK ; School of Psychology, Faculty of Social Sciences, University of East Anglia, UK
| | - Tim Dalgleish
- Medical Research Council Cognition and Brain Sciences Unit, Cambridge, UK
| | - Michael V Lombardo
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Valerie J Dunn
- Developmental Psychiatry Section, Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Anne-Laura Van Harmelen
- Developmental Psychiatry Section, Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Maria Ban
- Department of Clinical Neurosciences, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Ian M Goodyer
- Developmental Psychiatry Section, Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
21
|
Tam EWY. Potential mechanisms of cerebellar hypoplasia in prematurity. Neuroradiology 2013; 55 Suppl 2:41-6. [PMID: 23842990 DOI: 10.1007/s00234-013-1230-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 06/27/2013] [Indexed: 01/10/2023]
Abstract
INTRODUCTION The cerebellum undergoes dramatic growth and maturation over the neonatal period after preterm birth and is thus particularly sensitive to impaired development due to various clinical factors. METHODS Impairments in growth can occur independent of cerebellar parenchymal damage, such as from local hemorrhage, resulting from reduced expression of sonic hedgehog signaling to trigger the appropriate expansion of the granule precursor cells. RESULTS The primary risk factors for impaired cerebellar development include postnatal glucocorticoid exposure, which has direct effects on the sonic hedgehog pathway, and supratentorial brain injury, including intraventricular hemorrhage and white matter injury, which may result in crossed cerebellar diaschisis and local toxic effects of blood products on the external granular layer. Other cardiorespiratory and nutritional factors may also exist. Impaired cerebellar development is associated with adverse outcomes in motor and cognitive development. CONCLUSION New approaches to care to counteract these risk factors may help improve long-term outcome after preterm birth.
Collapse
Affiliation(s)
- Emily W Y Tam
- Division of Neurology, Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, Ontario, Canada.
| |
Collapse
|
22
|
Cerebellar granule cells are generated postnatally in humans. Brain Struct Funct 2013; 219:1271-86. [PMID: 23716277 DOI: 10.1007/s00429-013-0565-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 04/22/2013] [Indexed: 12/22/2022]
Abstract
How many cerebellar granule cells are generated pre- or postnatally in human is unknown. Using a rigorous design-based stereologic approach we investigated postmortem cerebella from 14 children who died between the first postnatal day (P1) and 11 months of age (M11). We found a statistically significant (p < 0.05) age-related increase in the total number of granule cells from 5.9 × 10(9) at M1 to 37.6 × 10(9) at M10/11 per cerebellar half but not in the total number of Purkinje cells (12.1 × 10(6) at M1 vs. 13.9 × 10(6) at M10/11 per cerebellar half). Accordingly, approximately 85 % of the cerebellar granule cells are generated postnatally in human, and the number of granule cells per Purkinje cell in the human cerebellum increases from 485 at M1 to 2,700 at M10/11, approximately. These data indicate that the human cerebellum has a much higher functional plasticity during the first year of life than previously thought, and may respond very sensitively to internal and external influences during this time. This has important implications for several neuropsychiatric conditions in which cerebellar involvement has been demonstrated.
Collapse
|
23
|
Miki T, Lee KY, Yokoyama T, Liu JQ, Kusaka T, Suzuki S, Ohta KI, Warita K, Jamal M, Ueki M, Yakura T, Hosomi N, Takeuchi Y. Differential effects of neonatal maternal separation on the expression of neurotrophic factors in rat brain. II: Regional differences in the cerebellum versus the cerebral cortex. Okajimas Folia Anat Jpn 2013; 90:53-8. [PMID: 24670490 DOI: 10.2535/ofaj.90] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
This study was conducted in order to examine the effects of early postnatal maternal separation stress on the age-dependent fluctuations in the expression levels of neurotrophic factor ligands and receptors in the developing cerebellum. Wistar rats were separated from their mothers for 3 h each day during postnatal days (PND) 10 to 15. The expression level of mRNA for brain-derived neurotrophic factor (BDNF), tyrosine kinase receptor B (TrkB), insulin-like growth factor-1 (IGF-1), and type-1 IGF receptor (IGF-1R) were evaluated in the cerebellum on PND16, 20, 30, and 60 with real-time RT-PCR. The mRNA levels of cerebellar BDNF in maternally separated rats were increased on PND16, while the other variables showed no significant alterations at any of the time points examined. However, the effects of an identical maternal separation on the cerebral cortex were previously reported to be completely different. These results indicate regional differences in the responses of neurotrophic factor ligands/receptors between the cerebellum and cerebral cortex. Given that neurotrophic factors play important roles in brain development, alterations in these factors may interrupt normal brain development and ultimately, lead to functional disruptions.
Collapse
Affiliation(s)
- Takanori Miki
- Departments of Anatomy and Neurobiology, Kagawa University
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Premature birth often is associated with neurodevelopmental disabilities. In this issue of Science Translational Medicine, a pair of papers investigate--in patients and in a reliable animal model--the effects of glucocorticoids on cerebellar development after premature birth.
Collapse
Affiliation(s)
- Olivier Baud
- Inserm, U676, Hôpital Robert Debré, Paris 75019, France
| | | |
Collapse
|
25
|
Tam EWY, Chau V, Ferriero DM, Barkovich AJ, Poskitt KJ, Studholme C, Fok EDY, Grunau RE, Glidden DV, Miller SP. Preterm cerebellar growth impairment after postnatal exposure to glucocorticoids. Sci Transl Med 2012; 3:105ra105. [PMID: 22013125 DOI: 10.1126/scitranslmed.3002884] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
As survival rates of preterm newborns improve as a result of better medical management, these children increasingly show impaired cognition. These adverse cognitive outcomes are associated with decreases in the volume of the cerebellum. Because animals exhibit reduced preterm cerebellar growth after perinatal exposure to glucocorticoids, we sought to determine whether glucocorticoid exposure and other modifiable factors increased the risk for these adverse outcomes in human neonates. We studied 172 preterm neonatal infants from two medical centers, the University of British Columbia and the University of California, San Francisco, by performing serial magnetic resonance imaging examinations near birth and again near term-equivalent age. After we adjusted for associated clinical factors, antenatal betamethasone was not associated with changes in cerebellar volume. Postnatal exposure to clinically routine doses of hydrocortisone or dexamethasone was associated with impaired cerebellar, but not cerebral, growth. Alterations in treatment after preterm birth, particularly glucocorticoid exposure, may help to decrease risk for adverse neurological outcome after preterm birth.
Collapse
Affiliation(s)
- Emily W Y Tam
- Departments of Neurology and Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Schutter DJLG, Koolschijn PCMP, Peper JS, Crone EA. The cerebellum link to neuroticism: a volumetric MRI association study in healthy volunteers. PLoS One 2012; 7:e37252. [PMID: 22615955 PMCID: PMC3355107 DOI: 10.1371/journal.pone.0037252] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 04/18/2012] [Indexed: 01/23/2023] Open
Abstract
Prior research suggests an association between reduced cerebellar volumes and symptoms of depression and anxiety in patients with mood disorders. However, whether a smaller volume in itself reflects a neuroanatomical correlate for increased susceptibility to develop mood disorders remains unclear. The aim of the present study was to examine the relationship between cerebellar volume and neurotic personality traits in a non-clinical subject sample. 3T Structural magnetic resonance imaging scans were acquired, and trait depression and anxiety scales of the revised NEO personality inventory were assessed in thirty-eight healthy right-handed volunteers. Results showed that cerebellar volume corrected for total brain volume was inversely associated with depressive and anxiety-related personality traits. Cerebellar gray and white matter contributed equally to the observed associations. Our findings extend earlier clinical observations by showing that cerebellar volume covaries with neurotic personality traits in healthy volunteers. The results may point towards a possible role of the cerebellum in the vulnerability to experience negative affect. In conclusion, cerebellar volumes may constitute a clinico-neuroanatomical correlate for the development of depression- and anxiety-related symptoms.
Collapse
Affiliation(s)
- Dennis J L G Schutter
- Experimental Psychology, Helmholtz Institute, Utrecht University, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
27
|
Perinatal cerebellar injury in human and animal models. Neurol Res Int 2012; 2012:858929. [PMID: 22530126 PMCID: PMC3317029 DOI: 10.1155/2012/858929] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 11/29/2011] [Indexed: 12/20/2022] Open
Abstract
Cerebellar injury is increasingly recognized through advanced neonatal brain imaging as a complication of premature birth. Survivors of preterm birth demonstrate a constellation of long-term neurodevelopmental deficits, many of which are potentially referable to cerebellar injury, including impaired motor functions such as fine motor incoordination, impaired motor sequencing and also cognitive, behavioral dysfunction among older patients. This paper reviews the morphogenesis and histogenesis of the human and rodent developing cerebellum, and its more frequent injuries in preterm. Most cerebellar lesions are cerebellar hemorrhage and infarction usually leading to cerebellar abnormalities and/or atrophy, but the exact pathogenesis of lesions of the cerebellum is unknown. The different mechanisms involved have been investigated with animal models and are primarily hypoxia, ischemia, infection, and inflammation Exposure to drugs and undernutrition can also induce cerebellar abnormalities. Different models are detailed to analyze these various disturbances of cerebellar development around birth.
Collapse
|
28
|
Wada H, Breuner CW. Developmental changes in neural corticosteroid receptor binding capacity in altricial nestlings. Dev Neurobiol 2011; 70:853-61. [PMID: 20629050 DOI: 10.1002/dneu.20819] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Altricial nestlings typically do not show an adrenocortical response during the early post-hatch period. This may be a result of an immature hypothalamic-pituitary-adrenal axis, or an enhanced control of the axis by negative feedback. To examine whether the dampened adrenocortical response is due to higher receptor densities in hypothalamus and hippocampus, the major sites for negative feedback and tonic inhibition, we explored the ontogenetic changes in glucocorticoid (GR) and mineralocorticoid receptor (MR) binding capacities in the brain of white-crowned sparrow nestlings. During the 10-day nestling period, MR binding capacity decreased with age, whereas GR capacity was not affected. In addition, this overall decline in MR levels was driven entirely by a decline in cerebellar MR. No age-related changes were observed in hippocampal or hypothalamic areas. Our findings suggest that enhanced negative feedback does not play a major role in the attenuated adrenocortical responses seen in white-crowned sparrow nestlings.
Collapse
Affiliation(s)
- Haruka Wada
- Section of Integrative Biology, University of Texas at Austin, Austin, Texas 78712, USA.
| | | |
Collapse
|
29
|
Noguchi KK, Lau K, Smith DJ, Swiney BS, Farber NB. Glucocorticoid receptor stimulation and the regulation of neonatal cerebellar neural progenitor cell apoptosis. Neurobiol Dis 2011; 43:356-63. [PMID: 21530661 DOI: 10.1016/j.nbd.2011.04.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 03/14/2011] [Accepted: 04/07/2011] [Indexed: 01/21/2023] Open
Abstract
Glucocorticoids are used to treat respiratory dysfunction associated with premature birth but have been shown to cause neurodevelopmental deficits when used therapeutically. Recently, we established that acute glucocorticoid exposure at clinically relevant doses produces neural progenitor cell apoptosis in the external granule layer of the developing mouse cerebellum and permanent decreases in the number of cerebellar neurons. As the cerebellum naturally matures and neurogenesis is no longer needed, the external granule layer decreases proliferation and permanently disappears during the second week of life. At this same time, corticosterone (the endogenous rodent glucocorticoid) release increases and a glucocorticoid-metabolizing enzyme that protects the external granule layer against glucocorticoid receptor stimulation (11β-Hydroxysteroid-Dehydrogenase-Type 2; HSD2) naturally disappears. Here we show that HSD2 inhibition and raising corticosterone to adult physiological levels both can independently increase neural progenitor cell apoptosis in the neonatal mouse. Conversely, glucocorticoid receptor antagonism decreases natural physiological apoptosis in this same progenitor cell population suggesting that endogenous glucocorticoid stimulation may regulate apoptosis in the external granule layer. We also found that glucocorticoids which HSD2 can effectively metabolize generate less external granule layer apoptosis than glucocorticoids this enzyme is ineffective at breaking down. This finding may explain why glucocorticoids that this enzyme can metabolize are clinically effective at treating respiratory dysfunction yet seem to produce no neurodevelopmental deficits. Finally, we demonstrate that both acute and chronic glucocorticoid exposures produce external granule layer apoptosis but without appropriate control groups this effect becomes masked. These results are discussed in terms of their implications for glucocorticoid therapy and neurodevelopment during the perinatal period.
Collapse
Affiliation(s)
- Kevin K Noguchi
- Washington University in Saint Louis, School of Medicine, Department of Psychiatry, 660 South Euclid, Box #8134, Saint Louis, MO 63110, USA.
| | | | | | | | | |
Collapse
|
30
|
Strøm BO, Aden P, Mathisen GH, Lømo J, Davanger S, Paulsen RE. Transfection of chicken cerebellar granule neurons used to study glucocorticoid receptor regulation by nuclear receptor 4A (NR4A). J Neurosci Methods 2010; 193:39-46. [DOI: 10.1016/j.jneumeth.2010.08.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 07/30/2010] [Accepted: 08/08/2010] [Indexed: 10/19/2022]
|
31
|
Wilber AA, Lin GL, Wellman CL. Glucocorticoid receptor blockade in the posterior interpositus nucleus reverses maternal separation-induced deficits in adult eyeblink conditioning. Neurobiol Learn Mem 2010; 94:263-8. [PMID: 20558309 PMCID: PMC2922459 DOI: 10.1016/j.nlm.2010.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 05/05/2010] [Accepted: 06/08/2010] [Indexed: 11/16/2022]
Abstract
Previously, we showed that neonatal maternal separation impaired eyeblink conditioning in adult rats. This impairment is correlated with increased glucocorticoid receptor (GR) expression in the cerebellar posterior interpositus nucleus, a critical site of learning-related plasticity. To assess whether increased GR expression is responsible for the separation-induced learning impairment, we infused a GR antagonist (mifepristone) or vehicle into the posterior interpositus during eyeblink conditioning in adult male Long-Evans rats that had undergone control rearing or neonatal maternal separation (1h/day, postnatal days 2-14). Rats received standard rearing (control) or neonatal maternal separation (separated; 1h/day on postnatal days 2-14). In adulthood, rats underwent surgery for implantation of recording electrodes in the orbicularis oculi of the left eyelid, a bipolar stimulating electrode dorsocaudal to the left eye, and an infusion guide cannula positioned over the posterior interpositus. Then, rats underwent 10 daily sessions of eyeblink conditioning. Rats in each group received either 0.2microl of mifepristone (2ng in 2% EtOH) or vehicle infusion prior to each eyeblink conditioning session. Mifepristone infusions improved conditioning in separated rats, but impaired control rats' performance. Thus, separation-induced increases in GRs may mediate the learning deficit seen in adult neonatally separated rats.
Collapse
Affiliation(s)
- Aaron A Wilber
- Department of Psychological and Brain Sciences and Program in Neuroscience, Indiana University, Bloomington, IN 47405, United States
| | | | | |
Collapse
|
32
|
Viveros MP, Llorente R, López-Gallardo M, Suarez J, Bermúdez-Silva F, De la Fuente M, Rodriguez de Fonseca F, Garcia-Segura LM. Sex-dependent alterations in response to maternal deprivation in rats. Psychoneuroendocrinology 2009; 34 Suppl 1:S217-26. [PMID: 19553026 DOI: 10.1016/j.psyneuen.2009.05.015] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2009] [Revised: 05/24/2009] [Accepted: 05/25/2009] [Indexed: 12/20/2022]
Abstract
We review here our latest results regarding short- and long-term effects of a neonatal maternal deprivation (MD) stress [24h at postnatal day (PND) 9] on diverse psychoneuroimmunoendocrine parameters, pointing out the existence of numerous sexual dimorphisms. Behavioral changes observed in MD animals might be at least in part attributable to neurodevelopmental effects of MD-induced elevated corticosterone levels. Our findings of short-term effects of MD on hippocampal and cerebellar neurons and glial cells appear to support this hypothesis. However, it is important to note that these cellular effects were more marked in males than in females. Moreover, in analyzing the effects of this neonatal stress on the endocannabinoid system (hippocampal endocannabinoid levels and CB1 receptors) we have also found that males were more affected by MD. Since all these sexual dimorphisms were found at an early neonatal age (PND 13), they are attributable to organizational effects of gonadal steroids. We discuss the potential implications of the elevated corticosterone and decreased leptin levels shown by MD animals in their diverse functional alterations, including the above mentioned neural effects as well as the intriguing persistent deficit in their immunological system. We also emphasize the necessity of analyzing the important influence of sex as regards the specific consequences of early life stress.
Collapse
Affiliation(s)
- M P Viveros
- Departamento de Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense, E-28040 Madrid, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Carrion VG, Weems CF, Watson C, Eliez S, Menon V, Reiss AL. Converging evidence for abnormalities of the prefrontal cortex and evaluation of midsagittal structures in pediatric posttraumatic stress disorder: an MRI study. Psychiatry Res 2009; 172:226-34. [PMID: 19349151 PMCID: PMC2704559 DOI: 10.1016/j.pscychresns.2008.07.008] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Revised: 02/02/2008] [Accepted: 07/06/2008] [Indexed: 10/20/2022]
Abstract
Volumetric imaging research has shown abnormal brain morphology in posttraumatic stress disorder (PTSD) when compared with control subjects. We present results on a study of brain morphology in the prefrontal cortex (PFC) and midline structures, via indices of gray matter volume and density, in pediatric PTSD. We hypothesized that both methods would demonstrate aberrant morphology in the PFC. Further, we hypothesized aberrant brainstem anatomy and reduced corpus callosum volume in children with PTSD. Twenty-four children (aged 7-14) with history of interpersonal trauma and 24 age- and gender-matched controls underwent structural magnetic resonance imaging (sMRI). Images of the PFC and midline brain structures were first analyzed using volumetric image analysis. The PFC data were then compared with whole brain voxel-based techniques using statistical parametric mapping (SPM). The PTSD group showed significantly increased gray matter volume in the right and left inferior and superior quadrants of the PFC and smaller gray matter volume in the pons and posterior vermis areas by volumetric image analysis. The voxel-by-voxel group comparisons demonstrated increased gray matter density mostly localized to ventral PFC as compared with the control group. Abnormal frontal lobe morphology, as revealed by separate-complementary image analysis methods, and reduced pons and posterior vermis areas are associated with pediatric PTSD. Voxel-based morphometry may help to corroborate and further localize data obtained by volume of interest methods in PTSD.
Collapse
Affiliation(s)
- Victor G. Carrion
- Stanford University School of Medicine Division of Child & Adolescent Psychiatry and Child Development,Corresponding author: Victor G. Carrion, M.D.Division of Child & Adolescent Psychiatry and Child Development Stanford University Stanford, CA 94305- 5719 Phone (650) 498-5164 Fax (650) 723-5531 Email
| | | | | | - Stephan Eliez
- Geneva University School of Medicine Division of Child and Adolescent Psychiatry
| | - Vinod Menon
- Stanford University School of Medicine Division of Child & Adolescent Psychiatry and Child Development
| | - Allan L. Reiss
- Stanford University School of Medicine Division of Child & Adolescent Psychiatry and Child Development
| |
Collapse
|
34
|
Early maternal deprivation in rats induces gender‐dependent effects on developing hippocampal and cerebellar cells. Int J Dev Neurosci 2009; 27:233-41. [DOI: 10.1016/j.ijdevneu.2009.01.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2008] [Revised: 12/31/2008] [Accepted: 01/13/2009] [Indexed: 11/19/2022] Open
|
35
|
López-Gallardo M, Llorente R, Llorente-Berzal A, Marco E, Prada C, Di Marzo V, Viveros M. Neuronal and glial alterations in the cerebellar cortex of maternally deprived rats: Gender differences and modulatory effects of two inhibitors of endocannabinoid inactivation. Dev Neurobiol 2008; 68:1429-40. [DOI: 10.1002/dneu.20672] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
36
|
Noguchi KK, Walls KC, Wozniak DF, Olney JW, Roth KA, Farber NB. Acute neonatal glucocorticoid exposure produces selective and rapid cerebellar neural progenitor cell apoptotic death. Cell Death Differ 2008; 15:1582-92. [PMID: 18600230 DOI: 10.1038/cdd.2008.97] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
There has been a growing controversy regarding the continued use of glucocorticoid therapy to treat respiratory dysfunction associated with prematurity, as mounting clinical evidence has shown neonatal exposure produces permanent neuromotor and cognitive deficits. Here we report that, during a selective neonatal window of vulnerability, a single glucocorticoid injection in the mouse produces rapid and selective apoptotic cell death of the proliferating neural progenitor cells in the cerebellar external granule layer and permanent reductions in neuronal cell counts of their progeny, the cerebellar internal granule layer neurons. Our estimates suggest that this mouse window of vulnerability would correspond in the human to a period extending from approximately 20 weeks gestation to 6.5 weeks after birth. This death pathway is critically regulated by the proapoptotic Bcl-2 family member Puma and is independent of p53 expression. These rodent data indicate that there exists a previously unknown window of vulnerability during which a single glucocorticoid exposure at clinically relevant doses can produce neural progenitor cell apoptosis and permanent cerebellar pathology that may be responsible for some of the iatrogenically induced neurodevelopmental abnormalities seen in children exposed to this drug. This vulnerability may be related to the physiological role of glucocorticoids in regulating programmed cell death in the mammalian cerebellum.
Collapse
Affiliation(s)
- K K Noguchi
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO 63110-1093, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Scheepens A, van de Waarenburg M, van den Hove D, Blanco CE. A single course of prenatal betamethasone in the rat alters postnatal brain cell proliferation but not apoptosis. J Physiol 2003; 552:163-75. [PMID: 12909684 PMCID: PMC2343326 DOI: 10.1113/jphysiol.2003.043414] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to determine the effects of a clinically relevant single course of prenatal betamethasone in the rat on growth parameters with particular reference to brain cell proliferation and apoptosis. We report that administration of 170 microg kg-1 betamethasone twice within 4 h to E20 pregnant rats conveys moderate somatic growth retardation. Further, using a measure of brain cell proliferation independent of blood-brain barrier (BBB) permeability, we demonstrate for the first time that betamethasone is chronically anti-proliferative to brain cells without inducing caspase-3-mediated apoptosis. More importantly we show that there is a significant and sexually divergent rebound of neural proliferation which occurs earlier in males than in females and continues until at least 21 days of postnatal life. BBB permeability to [3H]thymidine was significantly increased by steroid treatment re-iterating the fact that tracer studies not correcting for BBB permeability, such as bromodeoxyuridine (BrdU), may be questionable in this type of study. Further, prenatal steroid treatment did not alter postnatal corticosterone levels. In summary we show that prenatal betamethasone conveys significant and long-lasting side effects and that its human clinical application in preterm labour needs more careful consideration as compared to the relative ease with which it is prescribed today.
Collapse
Affiliation(s)
- Arjan Scheepens
- Department of Pediatrics, GROW Research Institute, University of Maastricht, 6202 AZ Maastricht, The Netherlands
| | | | | | | |
Collapse
|
38
|
Teicher MH, Andersen SL, Polcari A, Anderson CM, Navalta CP, Kim DM. The neurobiological consequences of early stress and childhood maltreatment. Neurosci Biobehav Rev 2003; 27:33-44. [PMID: 12732221 DOI: 10.1016/s0149-7634(03)00007-1] [Citation(s) in RCA: 834] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Early severe stress and maltreatment produces a cascade of neurobiological events that have the potential to cause enduring changes in brain development. These changes occur on multiple levels, from neurohumoral (especially the hypothalamic-pituitary-adrenal [HPA] axis) to structural and functional. The major structural consequences of early stress include reduced size of the mid-portions of the corpus callosum and attenuated development of the left neocortex, hippocampus, and amygdala. Major functional consequences include increased electrical irritability in limbic structures and reduced functional activity of the cerebellar vermis. There are also gender differences in vulnerability and functional consequences. The neurobiological sequelae of early stress and maltreatment may play a significant role in the emergence of psychiatric disorders during development.
Collapse
Affiliation(s)
- Martin H Teicher
- Department of Psychiatry, Harvard Medical School, and Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA 02478, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Teicher MH, Andersen SL, Polcari A, Anderson CM, Navalta CP. Developmental neurobiology of childhood stress and trauma. Psychiatr Clin North Am 2002; 25:397-426, vii-viii. [PMID: 12136507 DOI: 10.1016/s0193-953x(01)00003-x] [Citation(s) in RCA: 319] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Severe early stress and maltreatment produces a cascade of events that have the potential to alter brain development. The first stage of the cascade involves the stress-induced programming of the glucocorticoid, noradrenergic, and vasopressin-oxytocin stress response systems to augment stress responses. These neurohumors then produce effects on neurogenesis, synaptic overproduction and pruning, and myelination during specific sensitive periods. Major consequences include reduced size of the mid-portions of the corpus callosum; attenuated development of the left neocortex, hippocampus, and amygdala along with abnormal frontotemporal electrical activity; and reduced functional activity of the cerebellar vermis. These alterations, in turn, provide the neurobiological framework through which early abuse increases the risk of developing post-traumatic stress disorder (PTSD), depression, symptoms of attention-deficit/hyperactivity, borderline personality disorder, dissociative identity disorder, and substance abuse.
Collapse
Affiliation(s)
- Martin H Teicher
- Department of Psychiatry, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA.
| | | | | | | | | |
Collapse
|
40
|
Anderson CM, Teicher MH, Polcari A, Renshaw PF. Abnormal T2 relaxation time in the cerebellar vermis of adults sexually abused in childhood: potential role of the vermis in stress-enhanced risk for drug abuse. Psychoneuroendocrinology 2002; 27:231-44. [PMID: 11750781 DOI: 10.1016/s0306-4530(01)00047-6] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Recent studies suggest that childhood sexual abuse (CSA) elicits a cascade of neurohumoral events that affect brain development and is also a risk factor for the later development of substance abuse. We hypothesize that the cerebellar vermis may be a key region linking these observations. The vermis has a protracted ontogeny and a high density of glucocorticoid receptors, rendering it highly susceptible to early stress. The vermis modulates dopamine turnover in the accumbens and receives direct dopamine input through fibers with dopamine transporters. To test this hypothesis, steady-state functional magnetic resonance imaging (fMRI) (T2 relaxometry) was performed to assess resting blood flow in the vermis of 24 young adults (18-22 years) selected by screening from a large community sample. Eight subjects had a history of repeated CSA but were unmedicated and not under psychiatric care. Sixteen subjects were age-matched controls who had no personal or family history of Axis I psychiatric disorders. All subjects were screened to exclude known abnormalities affecting brain development, and any history of drug or alcohol abuse. CSA subjects had higher T2 relaxation time (T2-RT) than controls in the vermis but not in cerebral or cerebellar hemispheres. Vermal T2-RT correlated strongly with Limbic System Checklist (LSCL-33) ratings of temporal lobe epilepsy (TLE)-like symptomatology. From 537 prescreened young adults we found that their frequency of substance use was associated with a monotonic increase in LSCL-33 ratings and depression scores. Together these findings suggest that early trauma may interfere with the development of the vermis, and produce neuropsychiatric symptoms associated with drug use.
Collapse
Affiliation(s)
- Carl M Anderson
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| | | | | | | |
Collapse
|
41
|
Ferguson SA, Holson RR. Neonatal dexamethasone on day 7 causes mild hyperactivity and cerebellar stunting. Neurotoxicol Teratol 1999; 21:71-6. [PMID: 10023803 DOI: 10.1016/s0892-0362(98)00029-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
To investigate the effects of glucocorticoid treatment on central nervous system development, rats were injected with dexamethasone (DEX) (1-3 mg/kg) on postnatal day (PND) 3 or 7. DNA and protein content and concentration were measured in the cerebellum and hippocampus on PND 28 and 112. Whole and regional brain weights were measured at PND 28, 84, and 112. Nest odor preference (PND 10-11), open field activity (PND 18-21), running wheel activity (PND 50-56), and complex maze performance (PND 60-63) were measured in rats treated twice with 1.5 mg/kg DEX on PND 7. DEX treatment on PND 7 resulted in reductions in PND 28 whole brain and regional weights (frontal cortex, cerebellum, and brain stem) and, by PND 112, all except whole brain and cerebellar weights had recovered. A mild syndrome of hyperactivity (increased open field rearing and activity) was apparent in rats treated with DEX on PND 7. These results are discussed in terms of the developmental stage specificity in production of brain, and, specifically, cerebellar insults and their resulting effects.
Collapse
Affiliation(s)
- S A Ferguson
- Division of Reproductive & Developmental Toxicology, National Center for Toxicological Research/FDA, Jefferson, AR 72079, USA.
| | | |
Collapse
|
42
|
Robson AC, Leckie CM, Seckl JR, Holmes MC. 11 Beta-hydroxysteroid dehydrogenase type 2 in the postnatal and adult rat brain. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1998; 61:1-10. [PMID: 9795098 DOI: 10.1016/s0169-328x(98)00161-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
11 Beta-hydroxysteroid dehydrogenase (11 beta-HSD) catalyses the interconversion of active corticosterone and inert 11-dehydrocorticosterone. The recently discovered type 2 isozyme (11 beta-HSD-2) is a high affinity, NAD-dependent, exclusive 11 beta-dehydrogenase, which rapidly inactivates glucocorticoids. Thus the enzyme generates aldosterone-selectivity for intrinsically non-selective mineralocorticoid receptors in vivo as well as excluding glucocorticoids from glucocorticoid receptors, the latter being particularly important during development. Aldosterone exerts selective central effects upon salt appetite and blood pressure whilst glucocorticoids have potent effects upon postnatal neurogenesis and brain remodelling. We examined 11 beta-HSD-2 expression during postnatal ontogeny and in adult rat brain. High 11 beta-HSD-2 mRNA expression was found specifically in the postnatal thalamus and the external granule cell layer of the cerebellum. Expression peaked at the end of the first postnatal week and declined rapidly thereafter. Postnatal brain showed considerable activity of high affinity 11 beta-HSD-2 which paralleled expression of 11 beta-HSD-2 messenger ribonucleic acid (mRNA). Adult brain showed high 11 beta-HSD-2 mRNA expression limited to the subcommissural organ, with lower expression in the ventromedial nucleus of the hypothalamus, amygdala, locus coeruleus and nucleus tractus solitarius. These discrete areas are compatible with proposed selective central actions of aldosterone on blood pressure (subcommissural organ, nucleus tractus solitarius) and salt appetite (ventromedial nucleus, amygdala). In contrast, early postnatal 11 beta-HSD-2 coincides with glucocorticoid receptor rather than mineralocorticoid receptor expression, and areas of expression are among the regions where glucocorticoids have been demonstrated to have profound effects upon neuronal division, growth and maturation.
Collapse
Affiliation(s)
- A C Robson
- Molecular Endocrinology, Molecular Medicine Centre, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, Scotland, UK
| | | | | | | |
Collapse
|
43
|
Morimoto M, Morita N, Ozawa H, Yokoyama K, Kawata M. Distribution of glucocorticoid receptor immunoreactivity and mRNA in the rat brain: an immunohistochemical and in situ hybridization study. Neurosci Res 1996; 26:235-69. [PMID: 9121734 DOI: 10.1016/s0168-0102(96)01105-4] [Citation(s) in RCA: 365] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The localization of glucocorticoid receptor (GR) immunoreactivity and mRNA in the adult rat brain was examined by light microscopic and electron microscopic immunohistochemistries, and in situ hybridization. For the purpose of detailed investigation of the distribution and comparison of GR immunoreactivities and mRNAs, specific polyclonal antibodies against a part of the transcription modulation (TR) domain of rat GR were used in the immunohistochemistry, whereas fluorescein-labeled RNA probes, complementary to the TR domain in the GR cDNA were used in the in situ hybridization. In the rat brain, GR immunoreactivity was predominantly localized in the cell nucleus, and the expression of GR mRNA was detected in the cytoplasm. GR-immunoreactive and GR mRNA-containing cells were widely distributed from the olfactory bulb of the forebrain to the gracile-cuneate nuclei of the medulla oblongata. The highest densities of GR-immunoreactive and mRNA-containing cells were observed in the subfields of cerebral cortex, olfactory cortex, hippocampal formation, amygdala, septal region, dorsal thalamus, hypothalamus, trapezoid body, cerebellar cortex, locus coeruleus and dorsal nucleus raphe. The distributional pattern of GR immunoreactivity in many regions was well-correlated with that of GR mRNA, but in the CA3 and CA4 pyramidal layers of the hippocampus, different localization was noted. The present study provides the groundwork for elucidating the role of GRs in brain function.
Collapse
Affiliation(s)
- M Morimoto
- Department of Anatomy and Neurobiology, Kyoto Prefectural University of Medicine, Japan
| | | | | | | | | |
Collapse
|
44
|
Viau V, Sharma S, Meaney MJ. Changes in plasma adrenocorticotropin, corticosterone, corticosteroid-binding globulin, and hippocampal glucocorticoid receptor occupancy/translocation in rat pups in response to stress. J Neuroendocrinol 1996; 8:1-8. [PMID: 8932731 DOI: 10.1111/j.1365-2826.1996.tb00680.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Pituitary-adrenal responses to stress in the neonatal rat have been reported to be substantially reduced compared to older animals (i.e. a stress hyporesponsive period). This supposed period of endocrine quiescence is characterized by reduced stress-induced increases in both plasma ACTH and corticosterone. At the same time a number of authors have noted the decreased plasma corticosteroid-binding globulin (CBG) levels of the neonate, and there is evidence for an increased percentage of free corticosterone as well as age-related changes in the volume of distribution for corticosterone. These findings suggest that the reduced CBG levels might enhance the biological significance of existing glucocorticoid levels, beyond that assumed on the basis of plasma total corticosterone levels. We examined this question by estimating hippocampal glucocorticoid receptor occupancy and 'translocation' in Day 6, Day 15, and adult animals under basal and stressful conditions. The results showed that: 1) plasma ACTH levels were elevated in Day 6 animals in response to acute exposure to ether, maternal separation, and maternal separation + ether, however, ACTH responses were substantially lower than in Day 15 or adult animals; 2) Plasma total corticosterone levels followed a similar pattern; most noteworthy was the potent glucocorticoid response in Day 15 animals to the combination of maternal separation + ether; 3) Plasma CBG levels in Day 6 animals were extremely low (< 3% adult values); by Day 15 CBG levels were about 25% of adult levels. Interestingly, maternal separation was associated with a substantial decrease in plasma CBG levels; 4) Hippocampal glucocorticoid receptor occupancy/translocation was similar at all ages under both basal and stress conditions. The only notable exception occurred during maternal separation in Day 15 animals, where the percentage of hippocampal glucocorticoid receptor occupancy/translocation was higher than that observed at any time in either Day 6 or adult animals. This finding is likely related to the decrease in plasma CBG that occurs following separation of Day 15 pups from the dam. Thus, despite the higher corticosterone level in the adult, the increase in glucocorticoid receptor occupancy/translocation was generally comparable across all ages either under basal conditions, or following stress. These receptor data underscore the importance of developmental changes in plasma CBG levels.
Collapse
Affiliation(s)
- V Viau
- Developmental Neuroendocrinology Laboratory, Douglas Hospital Research Center, Montreal, Canada
| | | | | |
Collapse
|
45
|
Kawata M. Roles of steroid hormones and their receptors in structural organization in the nervous system. Neurosci Res 1995; 24:1-46. [PMID: 8848287 DOI: 10.1016/0168-0102(96)81278-8] [Citation(s) in RCA: 225] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Due to their chemical properties, steroid hormones cross the blood-brain barrier where they have profound effects on neuronal development and reorganization both in invertebrates and vertebrates, including humans mediated through their receptors. Steroids play a crucial role in the organizational actions of cellular differentiation representing sexual dimorphism and apoptosis, and in the activational effects of phenotypic changes in association with structural plasticity. Their sites of action are primarily the genes themselves but some are coupled with membrane-bound receptor/ion channels. The effects of steroid hormones on gene transcription are not direct, and other cellular components interfere with their receptors through cross-talk and convergence of the signaling pathways in neurons. These genomic and non-genomic actions account for the divergent effects of steroid hormones on brain function as well as on their structure. This review looks again at and updates the tremendous advances made in recent decades on the study of the role of steroid (gonadal and adrenal) hormones and their receptors on developmental processes and plastic changes in the nervous system.
Collapse
Affiliation(s)
- M Kawata
- Department of Anatomy and Neurobiology, Kyoto Prefectural University of Medicine, Japan
| |
Collapse
|
46
|
Abstract
We have previously demonstrated nitric oxide (NO)-dependent cyclic GMP (cGMP) formation in response to noradrenaline (NA) and glutamate (GLU) in astrocyte-enriched cultures from rat cerebrum. In the present work we show heterogeneity in agonist responses in astrocyte cultures from cerebellum, hippocampus and cortex. The response to NA was higher in cells from cerebellum, intermediate in cultures from hippocampus and low in cortical astrocytes. GLU had no significant effect in cortical and cerebellar cultures and presented lower effects than NA in cells from hippocampus. The NO donor sodium nitroprusside (SNP) produced much higher cGMP levels than agonists and the order of efficacies was cerebellum > cortex > hippocampus. Responses to NA and SNP in cerebellar astrocytes were sensitive to culture conditions decreasing when cells were seeded at low density or subcultured. Microglial cells were the main contaminants of the cerebellar astrocyte cultures but did not contribute to the NA or the SNP responses. No soluble guanylyl cyclase or calcium-dependent NO synthase (cNOS) activities were detected in microglial cultures. The effect of NA in cerebellar astrocytes was blocked by L-arginine analogues and by the alpha 1-adrenoceptor antagonist prazosin. The calcium ionophore A23187 mimicked the effect of NA and omission of calcium from the medium prevented both responses. NA did not elicit cGMP formation in granule cell cultures. These results support an astroglial location of the alpha 1-adrenoceptors and the cNOS that mediate NA stimulation of cGMP formation in cerebellum.
Collapse
Affiliation(s)
- L Agulló
- Instituto de Biología Fundamental V. Villar Palasí, Universidad Autónoma de Barcelona, Bellaterra, Spain
| | | | | |
Collapse
|
47
|
Rosenfeld P, van Eekelen JA, Levine S, de Kloet ER. Ontogeny of corticosteroid receptors in the brain. Cell Mol Neurobiol 1993; 13:295-319. [PMID: 8252605 DOI: 10.1007/bf00711575] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
1. In the brain, glucocorticoids bind to both the glucocorticoid receptor (GR) and the mineralocorticoid receptor (MR). These receptors show clearly distinct developmental patterns in the infant rat. 2. Low levels of GRs are present around the time of birth throughout the brain. Concentrations rise slowly, and do not achieve adult levels until the third week of life, approximately. GR affinity for corticosterone is higher perinatally than at later ages. Receptor microdistribution changes dramatically during ontogeny. In particular, certain regions, such as the suprachiasmatic nucleus of the hypothalamus, express high levels of receptor only during the first week of life. GRs may show impaired capacity to undergo transformation and/or nuclear translocation during the second postnatal week. Environmental manipulations during early ontogeny (e.g., early handling) may have permanent effects on GR capacity. 3. MRs are present at very low concentrations in the first days of life. Binding capacity rises rapidly thereafter and resembles that found in the adult by the end of 1 week. Neither binding affinity in vitro nor overall distribution changes with age. As in the adult, low doses of corticosterone, in vivo, bind mainly to the MRs. Levels of corticosterone are low and relatively unperturbable in the intact infant rat. It is likely, therefore, that most of the physiological actions of this hormone during this period are mediated by the MR.
Collapse
Affiliation(s)
- P Rosenfeld
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford University, California 94305
| | | | | | | |
Collapse
|
48
|
Abstract
Several laboratories have reported that chronic exposure to delta-9-tetrahydrocannabinol (THC) or marijuana extracts persistently altered the structure and function of the rat hippocampus, a paleocortical brain region involved with learning and memory processes in both rats and humans. Certain choices must be made in designing experiments to evaluate cannabis neurotoxicity, such as dose, route of administration, duration of exposure, age at onset of exposure, species of subjects, whether or how long to allow withdrawal, and which endpoints or biomarkers of neurotoxicity to measure. A review of the literature suggests that both age during exposure and duration of exposure may be critical determinants of neurotoxicity. Cannabinoid administration for at least three months (8-10% of a rat's lifespan) was required to produce neurotoxic effects in peripubertal rodents, which would be comparable to about three years exposure in rhesus monkeys and seven to ten years in humans. Studies of monkeys after up to 12 months of daily exposure have not consistently reported neurotoxicity, and the results of longer exposures have not yet been studied.
Collapse
Affiliation(s)
- A C Scallet
- Division of Reproductive and Developmental Toxicology, National Center for Toxicological Research, Jefferson, AR 72079
| |
Collapse
|
49
|
Daw NW, Sato H, Fox K, Carmichael T, Gingerich R. Cortisol reduces plasticity in the kitten visual cortex. JOURNAL OF NEUROBIOLOGY 1991; 22:158-68. [PMID: 1674285 DOI: 10.1002/neu.480220206] [Citation(s) in RCA: 30] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We investigated the effect of elevated levels of cortisol on plasticity in the visual cortex of the cat. Animals were given daily injections of cortisol i.m. for 20 days starting around 35 days of age. After 10 days they were monocularly deprived, and after an additional 10 days recordings were made from the visual cortex to construct an ocular dominance histogram. The results were compared with those from normal animals of the same age, and with animals monocularly deprived for the same period but not treated with cortisol. Cortisol reduced the ocular dominance shift in a dose-dependent manner, but did not totally abolish it even at the highest doses used. Two other series of animals were recorded, one slightly later in the critical period and one slightly earlier, with care taken to give cortisol before the animals were exposed to light in the morning. In both cases, cortisol reduced the ocular dominance shift but did not abolish it. To interpret these results, we measured levels of plasma cortisol in normal cats of various ages. Average levels were fairly constant between birth and 12 months of age (0.5-1 microgram/dl), and increased slightly after that, but there was a large variation between animals. Thus elevated levels of cortisol can have a substantial effect on plasticity in the visual cortex of the cat, but the decline of the critical period for plasticity between 6 weeks and 3-5 months of age does not seem to be due to a rise in cortisol levels during this time.
Collapse
Affiliation(s)
- N W Daw
- Department of Cell Biology, Washington University School of Medicine, St. Louis, Missouri 63110
| | | | | | | | | |
Collapse
|
50
|
Kovács KJ, Westphal HM, Péczely P. Distribution of glucocorticoid receptor-like immunoreactivity in the brain, and its relation to CRF and ACTH immunoreactivity in the hypothalamus of the japanese quail, Coturnix coturnix japonica. Brain Res 1989; 505:239-45. [PMID: 2557133 DOI: 10.1016/0006-8993(89)91449-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Monoclonal antibody (MAb49) against the rat liver glucocorticoid receptor was used to evaluate glucocorticoid receptor (GR) immunoreactive structures in the brain of the japanese quail, Coturnix coturnix japonica. Using the avidin-biotin technique, the immunoreaction was present in the nerve cell nuclei in intact male birds. High density of glucocorticoid receptor-like immunoreactivity was found in the tubero-infundibular area, nucleus paraventricularis, posteromedialis and lateralis hypothalami, lateral septum and in some brainstem nuclei. Cerebellar cortex was also immunopositive. In contrast to mammals, no immunoreactive cell nuclei were found in the hippocampal region. The glucocorticoid receptors were colocalized with adrenocorticotropin (ACTH) immunoreactivity in the tubero-infundibular region, while corticotropin releasing factor (CRF) positive cells in the paraventricular nucleus did not contain glucocorticoid receptor-like immunoreactivity in their cell nuclei.
Collapse
Affiliation(s)
- K J Kovács
- Institute of Experimental Medicine, Budapest, Hungary
| | | | | |
Collapse
|