1
|
Schkoda S, Horman B, Witchey S, St Armour G, Nelson M, Gaeta E, Scott M, Patisaul HB. Sex-specific effects on elements of the social brain neural network in Wistar rats from perinatal exposure to FireMaster 550 or its components. Neurotoxicology 2024; 105:111-120. [PMID: 39241866 DOI: 10.1016/j.neuro.2024.09.001] [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: 07/04/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Developmental exposure to chemical flame retardants (FRs) has been linked to a variety of neurodevelopmental disorders and abnormal socioemotional behaviors in human and laboratory animal studies. We have previously shown in Wistar rats that gestational and lactational exposure to the FR mixture Firemaster 550 (FM 550) or its brominated or organophosphate ester (OPFR) components (at 2000 µg, 1000 µg, and 1000 µg oral to the dam respectively (absolute and not by bodyweight)) results in increased anxiety-like behaviors in females and decreased sociality in both sexes. Using their siblings, this study characterized sex and chemical specific targets of disruption in brain regions underlying each behavioral phenotype. Offspring were exposed across gestation and lactation then prepared for either immunohistochemistry or autoradiography at postnatal day 90 to quantify expression of serotonin, estrogen receptor α (ERα), and oxytocin receptor (OTR) in multiple brain regions. No effect of exposure was found in males for any biological target. In females, serotonin innervation was increased in the medial amygdala of FM 550 exposed animals while ERα expression in the bed nucleus of the stria terminalis (BNST) was reduced by FM 550 and OPFR. Evidence of disrupted OTR was observed in males, particularly the BNST but considered an exploratory finding given the small sample size. These results begin to shed light on the mechanisms by which developmental FR exposure alters socioemotional behaviors of relevance to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Stacy Schkoda
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, United States
| | - Brian Horman
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, United States
| | - Shannah Witchey
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, United States
| | - Genevieve St Armour
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, United States; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, United States
| | - Mason Nelson
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, United States
| | - Emily Gaeta
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, United States
| | - Madeline Scott
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, United States
| | - Heather B Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, United States; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, United States.
| |
Collapse
|
2
|
Geuens S, Van Dessel J, Kan HE, Govaarts R, Niks EH, Goemans N, Lemiere J, Doorenweerd N, De Waele L. Genotype and corticosteroid treatment are distinctively associated with gray matter characteristics in patients with Duchenne muscular dystrophy. Neuromuscul Disord 2024; 45:105238. [PMID: 39522443 DOI: 10.1016/j.nmd.2024.105238] [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: 08/05/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
This study investigated if structural variation in specific gray matter areas is associated with corticosteroid treatment or genotype, and if cerebral morphological variations are related to neuropsychological and behavioral outcomes. The CAT12 toolbox in SPM was used for MRI segmentations, assessing subcortical structures, cortical thickness, gyrification, and sulci depths for DMD patients (n = 40; 9-18 years) and age-matched controls (n = 40). Comparisons were made between DMD vs. controls, daily vs. intermittent corticosteroid treatment (n = 20 each), and Dp140+ vs. Dp140- gene mutations (n = 15 vs. 25). MANCOVA, CAT12 3D statistics and Pearson correlations were conducted. DMD patients showed differences in volumes of distinct subcortical structures, left hemisphere cortical thickness, and gyrification in multiple brain areas compared with healthy controls. The daily treated DMD group exhibited differences in subcortical volumes and different patterns of cortical thickness, sulci depth, and gyrification compared to the intermittent treated DMD group. DMD Dp140+ patients displayed altered gyrification and sulci depth compared to DMD Dp140- patients. Finally, we found correlations between neurobehavioral outcomes and brain areas that showed differences in cortical morphology associated with corticosteroid treatment. Both genotype and corticosteroid treatment are associated with variations in subcortical volumes and cortical morphology, albeit in different ways. Corticosteroid treatment appears to have a more profound association with differences in gray matter characteristics of brain regions that are associated with functional outcomes.
Collapse
Affiliation(s)
- Sam Geuens
- University Hospitals Leuven, Child Neurology, Leuven, Belgium; KU Leuven, Department of Development and Regeneration, Leuven, Belgium.
| | - Jeroen Van Dessel
- Center for Developmental Psychiatry, Department of Neurosciences, UPC-KU Leuven, Belgium
| | - Hermien E Kan
- Leiden University Medical Center, C.J. Gorter MRI Center, Department of Radiology, Netherlands; Duchenne Center Netherlands
| | - Rosanne Govaarts
- Leiden University Medical Center, C.J. Gorter MRI Center, Department of Radiology, Netherlands; Duchenne Center Netherlands
| | - Erik H Niks
- Duchenne Center Netherlands; Leiden University Medical Center, Department of Neurology, Netherlands
| | | | - Jurgen Lemiere
- University Hospitals Leuven, Pediatric Hemato-Oncology, Belgium; KU Leuven, Department Oncology, Pediatric Oncology, Belgium
| | - Nathalie Doorenweerd
- Leiden University Medical Center, C.J. Gorter MRI Center, Department of Radiology, Netherlands
| | - Liesbeth De Waele
- University Hospitals Leuven, Child Neurology, Leuven, Belgium; KU Leuven, Department of Development and Regeneration, Leuven, Belgium
| |
Collapse
|
3
|
Sofía-Avendaño-Lopez S, Rodríguez-Marín AJ, Lara-Castillo M, Agresott-Carrillo J, Lara-Cortés LE, Sánchez-Almanzar JF, Villamil-Cruz S, Rojas-Rodríguez LC, Ariza-Salamanca DF, Gaviria-Carrillo M, Calderon-Ospina CA, Rodríguez-Quintana J. Molecular, Pathophysiological, and Clinical Aspects of Corticosteroid-Induced Neuropsychiatric Effects: From Bench to Bedside. Biomedicines 2024; 12:2131. [PMID: 39335644 PMCID: PMC11429036 DOI: 10.3390/biomedicines12092131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/08/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Corticosteroids are frequently prescribed across medical disciplines, yet they are associated with various adverse effects, including neuropsychiatric symptoms, documented since their introduction over 60 years ago. The cellular mechanisms underlying neuropsychiatric symptoms are complex and somewhat obscure, involving multiple pathways. Notably, they include changes in excitability, cellular death of hippocampal and striatal neurons, and increased inflammation and oxidative stress. Clinical presentation varies, encompassing affective disorders (anxiety, euphoria, depression), psychotic episodes, and cognitive deficits. It is crucial to note that these manifestations often go unnoticed by treating physicians, leading to delayed detection of severe symptoms, complications, and underreporting. Discontinuation of corticosteroids constitutes the cornerstone of treatment, resolving symptoms in up to 80% of cases. Although the literature on this topic is scant, isolated cases and limited studies have explored the efficacy of psychotropic medications for symptomatic control and prophylaxis. Pharmacological intervention may be warranted in situations where corticosteroid reduction or withdrawal is not feasible or beneficial for the patient.
Collapse
Affiliation(s)
- Sara Sofía-Avendaño-Lopez
- Social Epidemiology Research Team, Institut Pierre Louis d'Epidémiologie et de Santé Publique, INSERM, Sorbonne Université, F 75012 Paris, France
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Angela Johanna Rodríguez-Marín
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Mateo Lara-Castillo
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Juanita Agresott-Carrillo
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Luna Estefanía Lara-Cortés
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Juan Felipe Sánchez-Almanzar
- Center for Research in Genetics and Genomics (CIGGUR), Institute of Translational Medicine (IMT), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Sophya Villamil-Cruz
- Center for Research in Genetics and Genomics (CIGGUR), Institute of Translational Medicine (IMT), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
- Research Group in Applied Biomedical Sciences (UR Biomed), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Luis Carlos Rojas-Rodríguez
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Daniel Felipe Ariza-Salamanca
- Department of Pharmacobiology, Center for Research and Advanced Studies (Cinvestav), National Polytechnic Institute, Mexico City 14300, Mexico
| | - Mariana Gaviria-Carrillo
- Neuroscience Research Group (NeURos), NeuroVitae Center for Neuroscience, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Carlos Alberto Calderon-Ospina
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
- Center for Research in Genetics and Genomics (CIGGUR), Institute of Translational Medicine (IMT), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
- Research Group in Applied Biomedical Sciences (UR Biomed), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Jesús Rodríguez-Quintana
- Fundacion CardioInfantil-Instituto de Cardiología, Bogotá 111156, Colombia
- Hospital Universitario Mayor Mederi, Bogotá 111411, Colombia
| |
Collapse
|
4
|
Lalonde R, Strazielle C. Neurochemical Anatomy of Cushing's Syndrome. Neurochem Res 2024; 49:1945-1964. [PMID: 38833089 DOI: 10.1007/s11064-024-04172-2] [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: 11/17/2023] [Revised: 03/05/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
The neurochemical anatomy underlying Cushing's syndrome is examined for regional brain metabolism as well as neurotransmitter levels and receptor binding of biogenic amines and amino acids. Preliminary studies generally indicate that glucose uptake, blood flow, and activation on fMRI scans decreased in neocortical areas and increased in subcortical areas of patients with Cushing's syndrome or disease. Glucocorticoid-mediated increases in hippocampal metabolism occurred despite in vitro evidence of glucocorticoid-induced decreases in glucose uptake or consumption, indicating that in vivo increases are the result of indirect, compensatory, or preliminary responses. In animal studies, glucocorticoid administration decreased 5HT levels and 5HT1A receptor binding in several brain regions while adrenalectomy increased such binding. Region-specific effects were also obtained in regard to the dopaminergic system, with predominant actions of glucocorticoid-induced potentiation of reuptake blockers and releasing agents. More in-depth neuroanatomical analyses are warranted of these and amino acid-related neurotransmission.
Collapse
Affiliation(s)
- Robert Lalonde
- Laboratory of Stress, Immunity, Pathogens (UR SIMPA), University of Lorraine, Campus Santé, Bât A/B 9, avenue de la Forêt de Haye, Vandoeuvre-les-Nancy, 54500, France.
| | - Catherine Strazielle
- Laboratory of Stress, Immunity, Pathogens (UR SIMPA), University of Lorraine, Campus Santé, Bât A/B 9, avenue de la Forêt de Haye, Vandoeuvre-les-Nancy, 54500, France
- CHRU Nancy, Vandoeuvre-les-Nancy, France
| |
Collapse
|
5
|
Arcego DM, Buschdorf JP, O'Toole N, Wang Z, Barth B, Pokhvisneva I, Rayan NA, Patel S, de Mendonça Filho EJ, Lee P, Tan J, Koh MX, Sim CM, Parent C, de Lima RMS, Clappison A, O'Donnell KJ, Dalmaz C, Arloth J, Provençal N, Binder EB, Diorio J, Silveira PP, Meaney MJ. A Glucocorticoid-Sensitive Hippocampal Gene Network Moderates the Impact of Early-Life Adversity on Mental Health Outcomes. Biol Psychiatry 2024; 95:48-61. [PMID: 37406925 DOI: 10.1016/j.biopsych.2023.06.028] [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: 11/03/2022] [Revised: 04/15/2023] [Accepted: 06/20/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Early stress increases the risk for psychiatric disorders. Glucocorticoids are stress mediators that regulate transcriptional activity and morphology in the hippocampus, which is implicated in the pathophysiology of multiple psychiatric conditions. We aimed to establish the relevance of hippocampal glucocorticoid-induced transcriptional activity as a mediator of the effects of early life on later psychopathology in humans. METHODS RNA sequencing was performed with anterior and posterior hippocampal dentate gyrus from adult female macaques (n = 12/group) that were chronically treated with betamethasone (glucocorticoid receptor agonist) or vehicle. Coexpression network analysis identified a preserved gene network in the posterior hippocampal dentate gyrus that was strongly associated with glucocorticoid exposure. The single nucleotide polymorphisms in the genes in this network were used to create an expression-based polygenic score in humans. RESULTS The expression-based polygenic score significantly moderated the association between early adversity and psychotic disorders in adulthood (UK Biobank, women, n = 44,519) and on child peer relations (ALSPAC [Avon Longitudinal Study of Parents and Children], girls, n = 1666 for 9-year-olds and n = 1594 for 11-year-olds), an endophenotype for later psychosis. Analyses revealed that this network was enriched for glucocorticoid-induced epigenetic remodeling in human hippocampal cells. We also found a significant association between single nucleotide polymorphisms from the expression-based polygenic score and adult brain gray matter density. CONCLUSIONS We provide an approach for the use of transcriptomic data from animal models together with human data to study the impact of environmental influences on mental health. The results are consistent with the hypothesis that hippocampal glucocorticoid-related transcriptional activity mediates the effects of early adversity on neural mechanisms implicated in psychiatric disorders.
Collapse
Affiliation(s)
- Danusa Mar Arcego
- Douglas Research Centre, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada.
| | - Jan-Paul Buschdorf
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Republic of Singapore
| | - Nicholas O'Toole
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | - Zihan Wang
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | - Barbara Barth
- Douglas Research Centre, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Irina Pokhvisneva
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | | | - Sachin Patel
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | | | - Patrick Lee
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Republic of Singapore
| | - Jennifer Tan
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Republic of Singapore
| | - Ming Xuan Koh
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Republic of Singapore
| | - Chu Ming Sim
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Republic of Singapore
| | - Carine Parent
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | | | - Andrew Clappison
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | - Kieran J O'Donnell
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada; Yale Child Study Center, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Carla Dalmaz
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Janine Arloth
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany; Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Nadine Provençal
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada; BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Elisabeth B Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Josie Diorio
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | - Patrícia Pelufo Silveira
- Douglas Research Centre, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Republic of Singapore
| | - Michael J Meaney
- Douglas Research Centre, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Republic of Singapore; Brain Body Initiative, Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Republic of Singapore
| |
Collapse
|
6
|
Geuens S, Van Dessel J, Govaarts R, Ikelaar NA, Meijer OC, Kan HE, Niks EH, Goemans N, Lemiere J, Doorenweerd N, De Waele L. Comparison of two corticosteroid regimens on brain volumetrics in patients with Duchenne muscular dystrophy. Ann Clin Transl Neurol 2023; 10:2324-2333. [PMID: 37822297 PMCID: PMC10723242 DOI: 10.1002/acn3.51922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023] Open
Abstract
OBJECTIVE Duchenne muscular dystrophy (DMD) is a neuromuscular disorder in which many patients also have neurobehavioral problems. Corticosteroids, the primary pharmacological treatment for DMD, have been shown to affect brain morphology in other conditions, but data in DMD are lacking. This study aimed to investigate the impact of two corticosteroid regimens on brain volumetrics in DMD using magnetic resonance imaging (MRI). METHODS In a cross-sectional, two-center study, T1-weighted MRI scans were obtained from three age-matched groups (9-18 years): DMD patients treated daily with deflazacort (DMDd, n = 20, scan site: Leuven), DMD patients treated intermittently with prednisone (DMDi, n = 20, scan site: Leiden), and healthy controls (n = 40, both scan sites). FSL was used to perform voxel-based morphometry analyses and to calculate intracranial, total brain, gray matter, white matter, and cerebrospinal fluid volumes. A MANCOVA was employed to compare global volumetrics between groups, with site as covariate. RESULTS Both patient groups displayed regional differences in gray matter volumes compared to the control group. The DMDd group showed a wider extent of brain regions affected and a greater difference overall. This was substantiated by the global volume quantification: the DMDd group, but not the DMDi group, showed significant differences in gray matter, white matter, and cerebrospinal fluid volumes compared to the control group, after correction for intracranial volume. INTERPRETATION Volumetric differences in the brain are considered part of the DMD phenotype. This study suggests an additional impact of corticosteroid treatment showing a contrast between pronounced alterations seen in patients receiving daily corticosteroid treatment and more subtle differences in those treated intermittently.
Collapse
Affiliation(s)
- Sam Geuens
- Child NeurologyUniversity Hospitals LeuvenLeuvenBelgium
- Department of Development and RegenerationKU LeuvenLeuvenBelgium
| | - Jeroen Van Dessel
- Department of Neurosciences, Center for Developmental PsychiatryUPC‐KU LeuvenLeuvenBelgium
| | - Rosanne Govaarts
- C.J. Gorter MRI Center, RadiologyLeiden University Medical CenterLeidenNetherlands
- Duchenne Center NetherlandsLeidenNetherlands
| | - Nadine A. Ikelaar
- Duchenne Center NetherlandsLeidenNetherlands
- Department of NeurologyLeiden University Medical CenterLeidenNetherlands
| | - Onno C. Meijer
- Department of MedicineLeiden University Medical CenterLeidenNetherlands
| | - Hermien E. Kan
- C.J. Gorter MRI Center, RadiologyLeiden University Medical CenterLeidenNetherlands
- Duchenne Center NetherlandsLeidenNetherlands
| | - Erik H. Niks
- Duchenne Center NetherlandsLeidenNetherlands
- Department of NeurologyLeiden University Medical CenterLeidenNetherlands
| | | | - Jurgen Lemiere
- Pediatric Hemato‐OncologyUniversity Hospitals LeuvenLeuvenBelgium
- Department Oncology, Pediatric OncologyKU LeuvenLeuvenBelgium
| | - Nathalie Doorenweerd
- C.J. Gorter MRI Center, RadiologyLeiden University Medical CenterLeidenNetherlands
| | - Liesbeth De Waele
- Child NeurologyUniversity Hospitals LeuvenLeuvenBelgium
- Department of Development and RegenerationKU LeuvenLeuvenBelgium
| |
Collapse
|
7
|
Lisakovska O, Labudzynskyi D, Khomenko A, Isaev D, Savotchenko A, Kasatkina L, Savosko S, Veliky M, Shymanskyi I. Brain vitamin D3-auto/paracrine system in relation to structural, neurophysiological, and behavioral disturbances associated with glucocorticoid-induced neurotoxicity. Front Cell Neurosci 2023; 17:1133400. [PMID: 37020845 PMCID: PMC10067932 DOI: 10.3389/fncel.2023.1133400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/28/2023] [Indexed: 03/22/2023] Open
Abstract
IntroductionVitamin D3 (VD3) is a potent para/autocrine regulator and neurosteroid that can strongly influence nerve cell function and counteract the negative effects of glucocorticoid (GC) therapy. The aim of the study was to reveal the relationship between VD3 status and behavioral, structural-functional and molecular changes associated with GC-induced neurotoxicity.MethodsFemale Wistar rats received synthetic GC prednisolone (5 mg/kg b.w.) with or without VD3 (1000 IU/kg b.w.) for 30 days. Behavioral, histological, physiological, biochemical, molecular biological (RT-PCR, Western blotting) methods, and ELISA were used.Results and discussionThere was no difference in open field test (OFT), while forced swim test (FST) showed an increase in immobility time and a decrease in active behavior in prednisolone-treated rats, indicative of depressive changes. GC increased the perikaryon area, enlarged the size of the nuclei, and caused a slight reduction of cell density in CA1-CA3 hippocampal sections. We established a GC-induced decrease in the long-term potentiation (LTP) in CA1-CA3 hippocampal synapses, the amplitude of high K+-stimulated exocytosis, and the rate of Ca2+-dependent fusion of synaptic vesicles with synaptic plasma membranes. These changes were accompanied by an increase in nitration and poly(ADP)-ribosylation of cerebral proteins, suggesting the development of oxidative-nitrosative stress. Prednisolone upregulated the expression and phosphorylation of NF-κB p65 subunit at Ser311, whereas downregulating IκB. GC loading depleted the circulating pool of 25OHD3 in serum and CSF, elevated VDR mRNA and protein levels but had an inhibitory effect on CYP24A1 and VDBP expression. Vitamin D3 supplementation had an antidepressant-like effect, decreasing the immobility time and stimulating active behavior. VD3 caused a decrease in the size of the perikaryon and nucleus in CA1 hippocampal area. We found a recovery in depolarization-induced fusion of synaptic vesicles and long-term synaptic plasticity after VD3 treatment. VD3 diminished the intensity of oxidative-nitrosative stress, and suppressed the NF-κB activation. Its ameliorative effect on GC-induced neuroanatomical and behavioral abnormalities was accompanied by the 25OHD3 repletion and partial restoration of the VD3-auto/paracrine system.ConclusionGC-induced neurotoxicity and behavioral disturbances are associated with increased oxidative-nitrosative stress and impairments of VD3 metabolism. Thus, VD3 can be effective in preventing structural and functional abnormalities in the brain and behavior changes caused by long-term GC administration.
Collapse
Affiliation(s)
- Olha Lisakovska
- Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, Kyiv, Ukraine
- *Correspondence: Olha Lisakovska,
| | - Dmytro Labudzynskyi
- Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, Kyiv, Ukraine
| | - Anna Khomenko
- Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, Kyiv, Ukraine
| | - Dmytro Isaev
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, Kyiv, Ukraine
| | - Alina Savotchenko
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, Kyiv, Ukraine
| | - Ludmila Kasatkina
- Research Laboratory for Young Scientists, Palladin Institute of Biochemistry, Kyiv, Ukraine
| | - Serhii Savosko
- Department of Histology and Embryology, Bogomolets National Medical University, Kyiv, Ukraine
| | - Mykola Veliky
- Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, Kyiv, Ukraine
| | - Ihor Shymanskyi
- Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, Kyiv, Ukraine
| |
Collapse
|
8
|
Crawford AH, Hornby NL, de la Fuente AG, Piercy RJ. Brain magnetic resonance imaging in the DE50-MD dog model of Duchenne muscular dystrophy reveals regional reductions in cerebral gray matter. BMC Neurosci 2023; 24:21. [PMID: 36932329 PMCID: PMC10024360 DOI: 10.1186/s12868-023-00788-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 02/24/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy is a X-linked disease characterized by severe and progressive muscle weakness, alongside cognitive impairment and a range of neurobehavioral disorders secondary to brain dystrophin deficiency. Duchenne muscular dystrophy patients have reduced cerebral gray matter and altered white matter ultrastructure (detected by magnetic resonance imaging) compared to age-matched controls. METHODS We studied the DE50-MD canine model of Duchenne muscular dystrophy, which is deficient in full length brain dystrophin (Dp427) isoforms and has a neurocognitive phenotype. Eight DE50-MD and 6 age-matched littermate wild type male dogs underwent serial brain magnetic resonance imaging from 14 to 33 months of age. RESULTS Reduced regional gray matter was detected in DE50-MD dogs compared with wildtype, including the piriform lobe, hippocampus and cingulate gyrus. Lateral ventricle volume was larger in DE50-MD dogs. Differences did not progress over time. White matter volume did not differ between DE50-MD and wildtype dogs. There was no difference in brain nor cranial vault volume between DE50-MD and wildtype dogs. CONCLUSION Dystrophin deficiency in the canine brain results in structural changes that likely contribute to the neurocognitive phenotype.
Collapse
Affiliation(s)
- Abbe H. Crawford
- grid.20931.390000 0004 0425 573XComparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, UK
| | - Natasha L. Hornby
- grid.20931.390000 0004 0425 573XComparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, UK
| | - Alerie G. de la Fuente
- grid.513062.30000 0004 8516 8274Institute of Health and Biomedical Research of Alicante (ISABIAL), Alicante, Spain
- Institute of Neurosciences CSIC-UMH, San Juan de Alicante, Spain
- grid.4777.30000 0004 0374 7521Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast, UK
| | - Richard J. Piercy
- grid.20931.390000 0004 0425 573XComparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, UK
| |
Collapse
|
9
|
Hidden Comorbidities in Asthma: A Perspective for a Personalized Approach. J Clin Med 2023; 12:jcm12062294. [PMID: 36983294 PMCID: PMC10059265 DOI: 10.3390/jcm12062294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/05/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Bronchial asthma is the most frequent inflammatory non-communicable condition affecting the airways worldwide. It is commonly associated with concomitant conditions, which substantially contribute to its burden, whether they involve the lung or other districts. The present review aims at providing an overview of the recent acquisitions in terms of asthma concomitant systemic conditions, besides the commonly known respiratory comorbidities. The most recent research has highlighted a number of pathobiological interactions between asthma and other organs in the view of a shared immunological background underling different diseases. A bi-univocal relationship between asthma and common conditions, including cardiovascular, metabolic or neurodegenerative diseases, as well as rare disorders such as sickle cell disease, α1-Antitrypsin deficiency and immunologic conditions with hyper-eosinophilia, should be considered and explored, in terms of diagnostic work-up and long-term assessment of asthma patients. The relevance of that acquisition is of utmost importance in the management of asthma patients and paves the way to a new approach in the light of a personalized medicine perspective, besides targeted therapies.
Collapse
|
10
|
Wang T, Huang X, Wang J. Asthma's effect on brain connectivity and cognitive decline. Front Neurol 2023; 13:1065942. [PMID: 36818725 PMCID: PMC9936195 DOI: 10.3389/fneur.2022.1065942] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/29/2022] [Indexed: 02/05/2023] Open
Abstract
Objective To investigate the changes in dynamic voxel mirror homotopy connection (dVMHC) between cerebral hemispheres in patients with asthma. Methods Our study was designed using a case-control method. A total of 31 subjects with BA and 31 healthy subjects with matching basic information were examined using rsfMRI. We also calculated and obtained the dVMHC value between the cerebral cortexes. Results Compared with the normal control group, the dVMHC of the lingual gyrus (Ling) and the calcarine sulcus (CAL), which represented the visual network (VN), increased significantly in the asthma group, while the dVMHC of the medial superior frontal gyrus (MSFG), the anterior/middle/posterior cingulate gyrus (A/M/PCG), and the supplementary motor area (SMA) of the sensorimotor network decreased significantly in the asthma group. Conclusion This study showed that the ability of emotion regulation and the efficiency of visual and cognitive information processing in patients with BA was lower than in those in the HC group. The dVMHC analysis can be used to sensitively evaluate oxygen saturation, visual function changes, and attention bias caused by emotional disorders in patients with asthma, as well as to predict airway hyperresponsiveness, inflammatory progression, and dyspnea.
Collapse
Affiliation(s)
- Tao Wang
- Medical College of Nanchang University, Nanchang, China,The Second Department of Respiratory Disease, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Xin Huang
- Department of Ophthalmology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Jun Wang
- The Second Department of Respiratory Disease, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China,*Correspondence: Jun Wang ✉
| |
Collapse
|
11
|
Shorey CL, Mulla RT, Mielke JG. The effects of synthetic glucocorticoid treatment for inflammatory disease on brain structure, function, and dementia outcomes: A systematic review. Brain Res 2022; 1798:148157. [DOI: 10.1016/j.brainres.2022.148157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 08/31/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022]
|
12
|
van der Meulen M, Amaya JM, Dekkers OM, Meijer OC. Association between use of systemic and inhaled glucocorticoids and changes in brain volume and white matter microstructure: a cross-sectional study using data from the UK Biobank. BMJ Open 2022; 12:e062446. [PMID: 36041764 PMCID: PMC9438037 DOI: 10.1136/bmjopen-2022-062446] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE To test the hypothesis that systemic and inhaled glucocorticoid use is associated with changes in grey matter volume (GMV) and white matter microstructure. DESIGN Cross-sectional study. SETTING UK Biobank, a prospective population-based cohort study of adults recruited in the UK between 2006 and 2010. PARTICIPANTS After exclusion based on neurological, psychiatric or endocrinological history, and use of psychotropic medication, 222 systemic glucocorticoid users, 557 inhaled glucocorticoid users and 24 106 controls with available T1 and diffusion MRI data were included. MAIN OUTCOME MEASURES Primary outcomes were differences in 22 volumetric and 14 diffusion imaging parameters between glucocorticoid users and controls, determined using linear regression analyses adjusted for potential confounders. Secondary outcomes included cognitive functioning (six tests) and emotional symptoms (four questions). RESULTS Both systemic and inhaled glucocorticoid use were associated with reduced white matter integrity (lower fractional anisotropy (FA) and higher mean diffusivity (MD)) compared with controls, with larger effect sizes in systemic users (FA: adjusted mean difference (AMD)=-3.7e-3, 95% CI=-6.4e-3 to 1.0e-3; MD: AMD=7.2e-6, 95% CI=3.2e-6 to 1.1e-5) than inhaled users (FA: AMD=-2.3e-3, 95% CI=-4.0e-3 to -5.7e-4; MD: AMD=2.7e-6, 95% CI=1.7e-7 to 5.2e-6). Systemic use was also associated with larger caudate GMV (AMD=178.7 mm3, 95% CI=82.2 to 275.0), while inhaled users had smaller amygdala GMV (AMD=-23.9 mm3, 95% CI=-41.5 to -6.2) than controls. As for secondary outcomes, systemic users performed worse on the symbol digit substitution task (AMD=-0.17 SD, 95% CI=-0.34 to -0.01), and reported more depressive symptoms (OR=1.76, 95% CI=1.25 to 2.43), disinterest (OR=1.84, 95% CI=1.29 to 2.56), tenseness/restlessness (OR=1.78, 95% CI=1.29 to 2.41), and tiredness/lethargy (OR=1.90, 95% CI=1.45 to 2.50) compared with controls. Inhaled users only reported more tiredness/lethargy (OR=1.35, 95% CI=1.14 to 1.60). CONCLUSIONS Both systemic and inhaled glucocorticoid use are associated with decreased white matter integrity and limited changes in GMV. This association may contribute to the neuropsychiatric side effects of glucocorticoid medication, especially with chronic use.
Collapse
Affiliation(s)
- Merel van der Meulen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jorge Miguel Amaya
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Olaf M Dekkers
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Onno C Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
13
|
Dekkers AJ, Amaya JM, van der Meulen M, Biermasz NR, Meijer OC, Pereira AM. Long-term effects of glucocorticoid excess on the brain. J Neuroendocrinol 2022; 34:e13142. [PMID: 35980208 PMCID: PMC9541651 DOI: 10.1111/jne.13142] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 11/27/2022]
Abstract
The metabolic and cardiovascular clinical manifestations in patients with Cushing's syndrome (CS) are generally well known. However, recent studies have broadened the perspective of the effects of hypercortisolism, showing that both endogenous and exogenous glucocorticoid excess alter brain functioning on several time scales. Consequently, cognitive deficits and neuropsychological symptoms are highly prevalent during both active CS and CS in remission, as well as during glucocorticoid treatment. In this review, we discuss the effects of endogenous hypercortisolism and exogenously induced glucocorticoid excess on the brain, as well as the prevalence of cognitive and neuropsychological deficits and their course after biochemical remission. Furthermore, we propose possible mechanisms that may underly neuronal changes, based on experimental models and in vitro studies. Finally, we offer recommendations for future studies.
Collapse
Affiliation(s)
- Alies J. Dekkers
- Department of Medicine, Division of Endocrinology, Pituitary Center and Center for Endocrine TumorsLeiden University Medical CenterLeidenThe Netherlands
- Department of Medicine, Center for Endocrine Tumours LeidenLeiden University Medical CenterLeidenThe Netherlands
| | - Jorge Miguel Amaya
- Department of Medicine, Division of Endocrinology, Pituitary Center and Center for Endocrine TumorsLeiden University Medical CenterLeidenThe Netherlands
| | - Merel van der Meulen
- Department of Medicine, Division of Endocrinology, Pituitary Center and Center for Endocrine TumorsLeiden University Medical CenterLeidenThe Netherlands
- Department of Medicine, Center for Endocrine Tumours LeidenLeiden University Medical CenterLeidenThe Netherlands
| | - Nienke R. Biermasz
- Department of Medicine, Division of Endocrinology, Pituitary Center and Center for Endocrine TumorsLeiden University Medical CenterLeidenThe Netherlands
- Department of Medicine, Center for Endocrine Tumours LeidenLeiden University Medical CenterLeidenThe Netherlands
| | - Onno C. Meijer
- Department of Medicine, Division of Endocrinology, Pituitary Center and Center for Endocrine TumorsLeiden University Medical CenterLeidenThe Netherlands
| | - Alberto M. Pereira
- Department of Endocrinology & MetabolismAmsterdam UMC (AMC)AmsterdamThe Netherlands
| |
Collapse
|
14
|
Rosenkranz MA, Dean DC, Bendlin BB, Jarjour NN, Esnault S, Zetterberg H, Heslegrave A, Evans MD, Davidson RJ, Busse WW. Neuroimaging and biomarker evidence of neurodegeneration in asthma. J Allergy Clin Immunol 2022; 149:589-598.e6. [PMID: 34536414 PMCID: PMC8821112 DOI: 10.1016/j.jaci.2021.09.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/19/2021] [Accepted: 09/07/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Epidemiologic studies have shown that Alzheimer's disease (AD) and related dementias (ADRD) are seen more frequently with asthma, especially with greater asthma severity or exacerbation frequency. OBJECTIVE To examine the changes in brain structure that may underlie this phenomenon, we examined diffusion-weighted magnetic resonance imaging (dMRI) and blood-based biomarkers of AD (phosphorylated tau 181, p-Tau181), neurodegeneration (neurofilament light chain, NfL), and glial activation (glial fibrillary acidic protein, GFAP). METHODS dMRI data were obtained in 111 individuals with asthma, ranging in disease severity from mild to severe, and 135 healthy controls. Regression analyses were used to test the relationships between asthma severity and neuroimaging measures, as well as AD pathology, neurodegeneration, and glial activation, indexed by plasma p-Tau181, NfL, and GFAP, respectively. Additional relationships were tested with cognitive function. RESULTS Asthma participants had widespread and large-magnitude differences in several dMRI metrics, which were indicative of neuroinflammation and neurodegeneration, and which were robustly associated with GFAP and, to a lesser extent, NfL. The AD biomarker p-Tau181 was only minimally associated with neuroimaging outcomes. Further, asthma severity was associated with deleterious changes in neuroimaging outcomes, which in turn were associated with slower processing speed, a test of cognitive performance. CONCLUSIONS Asthma, particularly when severe, is associated with characteristics of neuroinflammation and neurodegeneration, and may be a potential risk factor for neural injury and cognitive dysfunction. There is a need to determine how asthma may affect brain health and whether treatment directed toward characteristics of asthma associated with these risks can mitigate these effects.
Collapse
Affiliation(s)
- Melissa A Rosenkranz
- Department of Psychiatry, University of Wisconsin-Madison, Madison, Wisc; Center for Healthy Minds, University of Wisconsin-Madison, Madison, Wisc.
| | - Douglas C Dean
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisc; Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisc; Waisman Center, University of Wisconsin-Madison, Madison, Wisc
| | - Barbara B Bendlin
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisc; Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, Madison, Wisc
| | - Nizar N Jarjour
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisc
| | - Stephane Esnault
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisc
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom; UK Dementia Research Institute at UCL, London, United Kingdom
| | | | - Michael D Evans
- Biostatistical Design and Analysis Center, Clinical and Translational Science Institute, University of Minnesota, Minneapolis, Minn
| | - Richard J Davidson
- Department of Psychiatry, University of Wisconsin-Madison, Madison, Wisc; Center for Healthy Minds, University of Wisconsin-Madison, Madison, Wisc; Department of Psychology, University of Wisconsin-Madison, Madison, Wisc
| | - William W Busse
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisc
| |
Collapse
|
15
|
Subcortical gray matter volumes in asthma: associations with asthma duration, control, and anxiety. Brain Imaging Behav 2021; 14:2341-2350. [PMID: 31501976 PMCID: PMC8116327 DOI: 10.1007/s11682-019-00188-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Asthma as a chronic inflammatory disease can be expected to affect central nervous system structures but little is known about subcortical structures in asthma and their potential association with illness-specific outcomes and anxiety. A total of 40 young adults (20 with asthma and 20 gender- and age-matched controls) underwent high-resolution T1-weighted MRI scan, viewed short distressing film clips, and filled in questionnaires about anxious and depressed mood, as well as asthma history, control, and catastrophizing thoughts about asthma, for those with asthma. The structural scans were processed in FSL's FIRST program to delineate subcortical structures of interest: amygdala, hippocampus, putamen, pallidum, caudate nucleus, nucleus accumbens, and thalamus. Findings showed no general reduction in subcortical gray matter volumes in asthma compared to controls. Asthma duration, asthma control, and catastrophizing of asthma and asthma attacks were negatively associated with volumes of putamen and pallidum, and to a weaker extent thalamus and amygdala, while controlling for gender, age, and corticosteroid inhaler use. In addition, stronger anxiety in response to distressing films was associated with lower volume of the pallidum, whereas general anxious and depressed mood was unrelated to subcortical structures. Thus, although there are no subcortical structural differences between young adults with asthma and healthy controls, longer asthma history, suboptimal management, and illness-related anxiety are reflected in lower gray matter volumes of subcortical structures, further emphasizing the importance of maintaining optimal asthma control.
Collapse
|
16
|
Mikhailichenko N, Wang YH, Wei JCC, Lai TJ. Association between Anti-inflammatory Drug and Dementia in Patients with Gout: A Nationwide, Population-Based Nested Case-Control Study. Int J Med Sci 2021; 18:2042-2050. [PMID: 33850475 PMCID: PMC8040399 DOI: 10.7150/ijms.55496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/22/2021] [Indexed: 11/25/2022] Open
Abstract
Introduction: The interaction between hyperuricemia and the cognitive system is still under debate, with studies presenting somewhat conflicting results. Objectives: This study aimed to investigate the risk of dementia in patients with gout who are administered anti-inflammatory drug treatment. Methods: Gouty arthritis patients aged 50 years and older, who received at least one of the background therapy drugs (colchicine, corticosteroids, or nonsteroidal anti-inflammatory drugs for 6 months), were divided into the following groups and compared: patients who had dementia over a period of 5 years (n = 2,292) and matched patients without dementia (n = 2,292). Results: We found that the most significant risk factors for dementia were stroke (OR, 2.66; 95% C.I., 2.33-3.03; AOR, 2.39; 95% C.I., 2.08-2.75) and depression (OR, 3.72; 95% C.I., 3.01-4.6; AOR, 3.25; 95% C.I., 2.60-4.05). The results of anti-gout drug administration, which impacted the dementia risk among patients of all ages (but especially in 50-64-year-old patients), demonstrated a higher risk ratio after 90 days of corticosteroid use (OR, 3.39; 95% C.I., 1.15-9.99), which was further increased after 180 days (OR, 3.61; 95% C.I., 1.31-9.94). We revealed that female patients experienced a significant increase in dementia risk after 90 days of corticosteroid administration, whereas male patients experienced a significant increase only after 180 days (OR, 1.52; 95% C.I., 1.06-2.17). Conclusion: We had identified that > 90-day corticosteroid administration is a significant dementia risk factor in both female and male patients of all ages, especially in the 50-60-year-old group.
Collapse
Affiliation(s)
- Natalia Mikhailichenko
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- NEVRON International Medical Center, Vladivostok, 690078, Russia;
| | - Yu-Hsun Wang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - James Cheng-Chung Wei
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Allergy, Immunology & Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Te-Jen Lai
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Psychiatry, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
17
|
Zhang L, Lu L, Bu X, Li H, Tang S, Gao Y, Liang K, Zhang S, Hu X, Wang Y, Li L, Hu X, Lim KO, Gong Q, Huang X. Alterations in hippocampal subfield and amygdala subregion volumes in posttraumatic subjects with and without posttraumatic stress disorder. Hum Brain Mapp 2021; 42:2147-2158. [PMID: 33566375 PMCID: PMC8046112 DOI: 10.1002/hbm.25356] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 12/02/2020] [Accepted: 01/20/2021] [Indexed: 02/05/2023] Open
Abstract
The hippocampus and amygdala are important structures in the posttraumatic stress disorder (PTSD); however, the exact relationship between these structures and stress or PTSD remains unclear. Moreover, they consist of several functionally distinct subfields/subregions that may serve different roles in the neuropathophysiology of PTSD. Here we present a subregional profile of the hippocampus and amygdala in 145 survivors of a major earthquake and 56 non‐traumatized healthy controls (HCs). We found that the bilateral hippocampus and left amygdala were significantly smaller in survivors than in HCs, and there was no difference between survivors with (n = 69) and without PTSD (trauma‐exposed controls [TCs], n = 76). Analyses revealed similar results in most subfields/subregions, except that the right hippocampal body (in a head‐body‐tail segmentation scheme), right presubiculum, and left amygdala medial nuclei (Me) were significantly larger in PTSD patients than in TCs but smaller than in HCs. Larger hippocampal body were associated with the time since trauma in PTSD patients. The volume of the right cortical nucleus (Co) was negatively correlated with the severity of symptoms in the PTSD group but positively correlated with the same measurement in the TC group. This correlation between symptom severity and Co volume was significantly different between the PTSD and TCs. Together, we demonstrated that generalized smaller volumes in the hippocampus and amygdala were more likely to be trauma‐related than PTSD‐specific, and their subfields/subregions were distinctively affected. Notably, larger left Me, right hippocampal body and presubiculum were PTSD‐specific; these could be preexisting factors for PTSD or reflect rapid posttraumatic reshaping.
Collapse
Affiliation(s)
- Lianqing Zhang
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Lu Lu
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Xuan Bu
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Hailong Li
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Shi Tang
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Yingxue Gao
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Kaili Liang
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Suming Zhang
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Xinyue Hu
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Yanlin Wang
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Lei Li
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Xinyu Hu
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Kelvin O Lim
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, and Minneapolis VA Medical Center, Minneapolis, Minnesota, USA
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Xiaoqi Huang
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| |
Collapse
|
18
|
H A, Jj T, Nm S, N H, O R, Ti L, J P, V S, R P, T L, L K, H K. Prenatal maternal depressive symptoms are associated with smaller amygdalar volumes of four-year-old children. Psychiatry Res Neuroimaging 2020; 304:111153. [PMID: 32771833 DOI: 10.1016/j.pscychresns.2020.111153] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/26/2020] [Accepted: 07/27/2020] [Indexed: 10/23/2022]
Abstract
Prenatal maternal depressive symptoms are related to an increased offspring susceptibility to psychiatric disorders over the life course. Alterations in fetal brain development might partly mediate this association. The relation of prenatal depressive symptoms with child's amygdalar volumes is still underexplored, and this study aimed to address this gap. We explored the association of prenatal maternal depressive symptoms with amygdalar volumes in 28 4-year-old children (14 female). Amygdalar volumes were assessed using the volBrain pipeline and manual segmentation. Prenatal depressive symptoms were self-reported by mothers at gestational weeks 14, 24 and 34 (Edinburgh Postnatal Depression Scale). Sex differences were probed, and possible pre- and postnatal confounders, such as maternal general anxiety, were controlled for. We observed that elevated depressive symptoms of the early second trimester, after controlling for prenatal maternal general anxiety, were significantly related to smaller right amygdalar volumes in the whole sample. Higher depressive symptoms of the third trimester were associated with significantly smaller right amygdalar volumes in boys compared to girls. Altogether, our data suggest that offspring limbic brain development might be affected by maternal depressive symptoms in early pregnancy, and might also be more vulnerable to depressive symptoms in late pregnancy in boys compared to girls.
Collapse
Affiliation(s)
- Acosta H
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland; Department of Psychiatry and Psychotherapy, Philipps University of Marburg, Rudolf-Bultmann-St. 8, 35039, Marburg, Germany.
| | - Tuulari Jj
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland; Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland; Turku Collegium for Science and Medicine, University of Turku, Turku, Finland; Department of Psychiatry, University of Oxford, Oxford, UK
| | - Scheinin Nm
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland.
| | - Hashempour N
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Rajasilta O
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Lavonius Ti
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Pelto J
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Saunavaara V
- Department of Medical Physics, Turku University Hospital, Turku, Finland; Turku PET Center, University of Turku and Turku University Hospital, Turku, Finland
| | - Parkkola R
- Department of Radiology, University of Turku and Turku University Hospital, Turku, Finland
| | - Lähdesmäki T
- Department of Pediatric Neurology, University of Turku and Turku University Hospital, Turku, Finland
| | - Karlsson L
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland; Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Karlsson H
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland; Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland; Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
19
|
Acosta H, Kantojärvi K, Hashempour N, Pelto J, Scheinin NM, Lehtola SJ, Lewis JD, Fonov VS, Collins DL, Evans A, Parkkola R, Lähdesmäki T, Saunavaara J, Karlsson L, Merisaari H, Paunio T, Karlsson H, Tuulari JJ. Partial Support for an Interaction Between a Polygenic Risk Score for Major Depressive Disorder and Prenatal Maternal Depressive Symptoms on Infant Right Amygdalar Volumes. Cereb Cortex 2020; 30:6121-6134. [PMID: 32676648 DOI: 10.1093/cercor/bhaa158] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/02/2020] [Accepted: 05/09/2020] [Indexed: 12/22/2022] Open
Abstract
Psychiatric disease susceptibility partly originates prenatally and is shaped by an interplay of genetic and environmental risk factors. A recent study has provided preliminary evidence that an offspring polygenic risk score for major depressive disorder (PRS-MDD), based on European ancestry, interacts with prenatal maternal depressive symptoms (GxE) on neonatal right amygdalar (US and Asian cohort) and hippocampal volumes (Asian cohort). However, to date, this GxE interplay has only been addressed by one study and is yet unknown for a European ancestry sample. We investigated in 105 Finnish mother-infant dyads (44 female, 11-54 days old) how offspring PRS-MDD interacts with prenatal maternal depressive symptoms (Edinburgh Postnatal Depression Scale, gestational weeks 14, 24, 34) on infant amygdalar and hippocampal volumes. We found a GxE effect on right amygdalar volumes, significant in the main analysis, but nonsignificant after multiple comparison correction and some of the control analyses, whose direction paralleled the US cohort findings. Additional exploratory analyses suggested a sex-specific GxE effect on right hippocampal volumes. Our study is the first to provide support, though statistically weak, for an interplay of offspring PRS-MDD and prenatal maternal depressive symptoms on infant limbic brain volumes in a cohort matched to the PRS-MDD discovery sample.
Collapse
Affiliation(s)
- H Acosta
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, 20500 Turku, Finland.,Department of Psychiatry and Psychotherapy, Philipps University of Marburg, 35037 Marburg, Germany
| | - K Kantojärvi
- Finnish Institute for Health and Welfare, Genomics and Biobank Unit, FI-00271 Helsinki, Finland.,Department of Psychiatry and SleepWell Research Program, Faculty of Medicine, Helsinki University Central Hospital, University of Helsinki, 00100 Helsinki, Finland
| | - N Hashempour
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, 20500 Turku, Finland
| | - J Pelto
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, 20500 Turku, Finland
| | - N M Scheinin
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, 20500 Turku, Finland.,Department of Psychiatry, Turku University Hospital, University of Turku, 20500 Turku, Finland
| | - S J Lehtola
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, 20500 Turku, Finland
| | - J D Lewis
- Montreal Neurological Institute, McGill University, Montreal H3A 0G4, Canada
| | - V S Fonov
- Montreal Neurological Institute, McGill University, Montreal H3A 0G4, Canada
| | - D L Collins
- Montreal Neurological Institute, McGill University, Montreal H3A 0G4, Canada
| | - A Evans
- Montreal Neurological Institute, McGill University, Montreal H3A 0G4, Canada
| | - R Parkkola
- Department of Radiology, Turku University Hospital, University of Turku, 20500 Turku, Finland
| | - T Lähdesmäki
- Department of Pediatric Neurology, Turku University Hospital, University of Turku, 20500 Turku, Finland
| | - J Saunavaara
- Department of Medical Physics, Turku University Hospital, 20521 Turku, Finland
| | - L Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, 20500 Turku, Finland.,Department of Child Psychiatry, Turku University Hospital, University of Turku, 20500 Turku, Finland
| | - H Merisaari
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, 20500 Turku, Finland.,Department of Future Technologies, University of Turku, 20500 Turku, Finland.,Center of Computational Imaging and Personalized Diagnostics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - T Paunio
- Finnish Institute for Health and Welfare, Genomics and Biobank Unit, FI-00271 Helsinki, Finland.,Department of Psychiatry and SleepWell Research Program, Faculty of Medicine, Helsinki University Central Hospital, University of Helsinki, 00100 Helsinki, Finland
| | - H Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, 20500 Turku, Finland.,Department of Psychiatry, Turku University Hospital, University of Turku, 20500 Turku, Finland
| | - J J Tuulari
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, 20500 Turku, Finland.,Department of Psychiatry, Turku University Hospital, University of Turku, 20500 Turku, Finland.,Turku Collegium for Science and Medicine, University of Turku, 20500 Turku, Finland.,Department of Psychiatry, University of Oxford, Oxford, OX1 2JD, UK
| |
Collapse
|
20
|
Cole JL. Steroid-Induced Sleep Disturbance and Delirium: A Focused Review for Critically Ill Patients. Fed Pract 2020; 37:260-267. [PMID: 32669778 PMCID: PMC7357890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
OBJECTIVE Insomnia and delirium have gained much attention since the publication of recent guidelines for the management in critically ill adults. Neurologic effects such as sleep disturbance, psychosis, and delirium are commonly cited adverse effects (AEs) of corticosteroids. Steroid use is considered a modifiable risk factor in intensive care unit patients; however, reported mechanisms are often lacking. This focused review will specifically evaluate the effects of steroids on sleep deprivation, psychosis, delirium, and what is known about these effects in a critically ill population. OBSERVATIONS The medical literature proposes 3 pathways primarily responsible for neurocognitive AEs of steroids: behavior changes through modification of the hypothalamic-pituitary-adrenal axis, changes in natural sleep-wake cycles, and hyperarousal caused by modification in neuroinhibitory pathways. Initial search fields produced 285 articles. Case reports, reviews, letters, and articles pertaining to primary care or palliative populations were excluded, leaving 8 relevant articles for inclusion. CONCLUSIONS Although steroid therapy often cannot be altered in the critically ill population, research showed that steroid overuse is common in intensive care units. Minimizing dosage and duration are important ways clinicians can mitigate AEs.
Collapse
Affiliation(s)
- Jennifer L Cole
- is a Critical Care and Internal Medicine Pharmacy Specialist at the Veterans Health Care System of the Ozarks in Fayetteville, Arkansas
| |
Collapse
|
21
|
Labanski A, Langhorst J, Engler H, Elsenbruch S. Stress and the brain-gut axis in functional and chronic-inflammatory gastrointestinal diseases: A transdisciplinary challenge. Psychoneuroendocrinology 2020; 111:104501. [PMID: 31715444 DOI: 10.1016/j.psyneuen.2019.104501] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 10/17/2019] [Accepted: 10/29/2019] [Indexed: 12/18/2022]
Abstract
The broad role of stress in the brain-gut axis is widely acknowledged, with implications for multiple prevalent health conditions that are characterized by chronic gastrointestinal symptoms. These include the functional gastrointestinal disorders (FGID), such as irritable bowel syndrome and functional dyspepsia, as well as inflammatory bowel diseases (IBD) like ulcerative colitis and Crohn's disease. Although the afferent and efferent pathways linking the gut and the brain are modulated by stress, the fields of neurogastroenterology and psychoneuroendocrinology (PNE)/ psychoneuroimmunology (PNI) remain only loosely connected. We aim to contribute to bringing these fields closer together by drawing attention to a fascinating, evolving research area, targeting an audience with a strong interest in the role of stress in health and disease. To this end, this review introduces the concept of the brain-gut axis and its major pathways, and provides a brief introduction to epidemiological and clinical aspects of FGIDs and IBD. From an interdisciplinary PNE/PNI perspective, we then detail current knowledge regarding the role of chronic and acute stress in the pathophysiology of FGID and IBD. We provide an overview of evidence regarding non-pharmacological treatment approaches that target central or peripheral stress mechanisms, and conclude with future directions, particularly those arising from recent advances in the neurosciences and discoveries surrounding the gut microbiota.
Collapse
Affiliation(s)
- Alexandra Labanski
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jost Langhorst
- Chair for Integrative Medicine, University of Duisburg-Essen, Essen, Germany; Clinic for Internal and Integrative Medicine, Klinikum Bamberg, Bamberg, Germany
| | - Harald Engler
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sigrid Elsenbruch
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
22
|
Sigurdardottir HL, Lanzenberger R, Kranz GS. Genetics of sex differences in neuroanatomy and function. HANDBOOK OF CLINICAL NEUROLOGY 2020; 175:179-193. [PMID: 33008524 DOI: 10.1016/b978-0-444-64123-6.00013-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
Sex differences are observed at many distinct biologic levels, such as in the anatomy and functioning of the brain, behavior, and susceptibility to neuropsychiatric disorders. Previously, these differences were believed to entirely result from the secretion of gonadal hormones; however, recent research has demonstrated that differences are also the consequence of direct or nonhormonal effects of genes located on the sex chromosomes. This chapter reviews the four core genotype model that separates the effects of hormones and sex chromosomes and highlights a few genes that are believed to be partly responsible for sex dimorphism of the brain, in particular, the Sry gene. Genetics of the brain's neurochemistry is discussed and the susceptibility to certain neurologic and psychiatric disorders is reviewed. Lastly, we discuss the sex-specific genetic contribution in disorders of sexual development. The precise molecular mechanisms underlying these differences are currently not entirely known. An increased knowledge and understanding of the role of candidate genes will undeniably be of great aid in elucidating the molecular basis of sex-biased disorders and potentially allow for more sex-specific therapies.
Collapse
Affiliation(s)
- Helen L Sigurdardottir
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria.
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Georg S Kranz
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria; Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, People's Republic of China; The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, People's Republic of China
| |
Collapse
|
23
|
Acosta H, Tuulari JJ, Scheinin NM, Hashempour N, Rajasilta O, Lavonius TI, Pelto J, Saunavaara V, Parkkola R, Lähdesmäki T, Karlsson L, Karlsson H. Maternal Pregnancy-Related Anxiety Is Associated With Sexually Dimorphic Alterations in Amygdala Volume in 4-Year-Old Children. Front Behav Neurosci 2019; 13:175. [PMID: 31447658 PMCID: PMC6691065 DOI: 10.3389/fnbeh.2019.00175] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/15/2019] [Indexed: 12/03/2022] Open
Abstract
Prenatal stress is associated with child behavioral outcomes increasing susceptibility for psychiatric disorders in later life. Altered fetal brain development might partly mediate this association, as some studies suggest. With this study, we investigated the relation between prenatal stress, child's brain structure and behavioral problems. The association between self-reported maternal pregnancy-related anxiety (PRAQ-R2 questionnaire, second and third trimester) and brain gray matter volume was probed in 27 4-year-old children (13 female). Voxel based morphometry was applied with an age-matched template in SPM for the whole-brain analyses, and amygdala volume was assessed with manual segmentation. Possible pre- and postnatal confounders, such as maternal depression and anxiety among others, were controlled for. Child behavioral problems were assessed with the Strength and Difficulties Questionnaire by maternal report. We found a significant interaction effect of pregnancy-related anxiety and child's sex on child's amygdala volume, i.e., higher pregnancy-related anxiety in the second trimester was related to significantly greater left relative amygdala volume in girls compared to boys. Further exploratory analyses yielded that both maternal pregnancy-related anxiety and child's amygdala volume are related to child emotional and behavioral difficulties: While higher pregnancy-related anxiety was associated with more emotional symptoms, peer relationship problems and overall child difficulties, greater left amygdala volume was related to less of these child difficulties and might partly mediate sex-specific associations between pregnancy-related anxiety and child behavioral difficulties. Our data suggest that maternal prenatal distress leads to sexually dimorphic structural changes in the offspring's limbic system and that these changes are also linked to behavioral difficulties. Our results provide further support for the notion that prenatal stress impacts child development.
Collapse
Affiliation(s)
- Henriette Acosta
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Jetro J. Tuulari
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
- Department of Psychiatry, Turku University Hospital, University of Turku, Turku, Finland
- Turku Collegium for Science and Medicine, University of Turku, Turku, Finland
| | - Noora M. Scheinin
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Niloofar Hashempour
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Olli Rajasilta
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Tuomas I. Lavonius
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Juho Pelto
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Virva Saunavaara
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | - Riitta Parkkola
- Department of Radiology, Turku University Hospital, University of Turku, Turku, Finland
| | - Tuire Lähdesmäki
- Department of Pediatric Neurology, Turku University Hospital, University of Turku, Turku, Finland
| | - Linnea Karlsson
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
- Department of Child Psychiatry, Turku University Hospital, University of Turku, Turku, Finland
| | - Hasse Karlsson
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
- Department of Psychiatry, Turku University Hospital, University of Turku, Turku, Finland
| |
Collapse
|
24
|
Liebell D. The Science of Auricular Microsystem Acupuncture: Amygdala Function in Psychiatric, Neuromusculoskeletal, and Functional Disorders. Med Acupunct 2019; 31:157-163. [PMID: 31297169 DOI: 10.1089/acu.2019.1339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Addressing dysfunction of the amygdala via the auricular acupuncture microsystem has vast potential. Widespread medical consequences of amygdala dysregulation have been well-defined scientifically. Clinical results and a significant quantity of existing amygdala research support the rationale for addressing amygdala function through auricular-acupuncture techniques. Further scientific inquiry into their mechanisms of action, clinical applications, and expansion of both professional and public education of the merits of these techniques is warranted.
Collapse
Affiliation(s)
- Donald Liebell
- The Liebell Clinic: Chronic Pain & Wellness Solutions, Virginia Beach, VA
| |
Collapse
|
25
|
Holm SK, Madsen KS, Vestergaard M, Born AP, Paulson OB, Siebner HR, Uldall P, Baaré WFC. Previous glucocorticoid treatment in childhood and adolescence is associated with long-term differences in subcortical grey matter volume and microstructure. NEUROIMAGE-CLINICAL 2019; 23:101825. [PMID: 31004915 PMCID: PMC6475768 DOI: 10.1016/j.nicl.2019.101825] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 03/24/2019] [Accepted: 04/10/2019] [Indexed: 11/17/2022]
Abstract
BACKGROUND Glucocorticoids are widely used in the treatment of several pediatric diseases with undisputed disease-related benefits. Perinatal exposure to high levels of glucocorticoids can have long-term adverse cerebral effects. In adults, glucocorticoid treatment has been associated with smaller volumes of subcortical grey matter structures. Recently, we observed smaller total brain volumes in children and adolescents treated with glucocorticoid during childhood compared to healthy controls. The possible long-term effects of glucocorticoid treatment during childhood on subcortical brain volume and microstructure remain unknown. METHOD We examined 30 children and adolescents, who had previously been treated with glucocorticoids for nephrotic syndrome or rheumatic disease, and 30 healthy volunteers. Patients and healthy control groups were matched by age, gender, and level of parent education. Participants underwent 3 T magnetic resonance (MR) brain imaging. T1-weighted and diffusion-weighted images were acquired. Volume and mean diffusivity (MD) measures were extracted for hippocampus, amygdala, nucleus accumbens, caudate nucleus and putamen. Multiple linear regression analyses were used to assess differences between patients and controls, and to evaluate possible dose-response relationships. A priori, we expected patients to display lower hippocampal and amygdala volumes. RESULTS While children previously treated with glucocorticoids displayed smaller right hippocampal volumes than controls, this difference did not survive correction for multiple comparisons. Furthermore, patients as compared to controls showed lower right hippocampal MD, which remained when correcting for global changes in MD. The longer the time between the glucocorticoid treatment termination and MR-scan, the more right hippocampal MD values resembled that of healthy controls. Patient and controls did not differ in amygdala volume or MD. Analyses of the nucleus accumbens, caudate nucleus and putamen only revealed smaller putamen volumes in patients compared to controls, which remained significant when controlling for total brain volume. CONCLUSION The results suggest that extra-cerebral diseases during childhood treated with glucocorticoids may be associated with reduced subcortical grey matter volumes and lower right hippocampal mean diffusivity later in life. Our findings warrant replication and elaboration in larger, preferably prospective and longitudinal studies. Such studies may also allow disentangling disease-specific effects from possible glucocorticoid treatment effects.
Collapse
Affiliation(s)
- Sara Krøis Holm
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Hvidovre, Denmark; Department of Paediatrics and Adolescent Medicine, Neuropaediatric Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Kathrine Skak Madsen
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Hvidovre, Denmark; Radiography, Department of Technology, University College Copenhagen, Copenhagen, Denmark
| | - Martin Vestergaard
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Hvidovre, Denmark
| | - Alfred Peter Born
- Department of Paediatrics and Adolescent Medicine, Neuropaediatric Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Olaf B Paulson
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Hvidovre, Denmark; Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Hartwig Roman Siebner
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Hvidovre, Denmark; Department of Neurology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Peter Uldall
- Department of Paediatrics and Adolescent Medicine, Neuropaediatric Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - William F C Baaré
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Hvidovre, Denmark.
| |
Collapse
|
26
|
Ennis GE, Quintin EM, Saelzler U, Kennedy KM, Hertzog C, Moffat SD. Cortisol relates to regional limbic system structure in older but not younger adults. Psychoneuroendocrinology 2019; 101:111-120. [PMID: 30453123 PMCID: PMC8074622 DOI: 10.1016/j.psyneuen.2018.09.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 12/22/2022]
Abstract
We investigated if the relationship between age and regional limbic system brain structure would be moderated by diurnal cortisol output and diurnal cortisol slope. Participants aged 23-83 years collected seven salivary cortisol samples each day for 10 consecutive days and underwent magnetic resonance imaging. Age, sex, cortisol, and an age x cortisol interaction were tested as predictors of hippocampal and amygdalar volume and caudal and rostral anterior cingulate cortex (ACC) thickness. We found significant interactions between age and cortisol on left and right amygdalar volumes and right caudal ACC thickness. Older adults with higher cortisol output had smaller left and right amygdalar volumes than older adults with lower cortisol output and younger adults with higher cortisol output. Older and younger adults with lower cortisol output had similar amygdalar volumes. Older adults with a steeper decline in diurnal cortisol had a thicker right caudal ACC than younger adults with a similarly shaped cortisol slope. Hippocampal volume was not related to either cortisol slope or output, nor was pallidum volume which was assessed as an extra-limbic control region. Results suggest that subtle differences in cortisol output are related to differences in limbic system structure in older but not younger adults.
Collapse
Affiliation(s)
- Gilda E Ennis
- School of Psychology, Georgia Institute of Technology, 654 Cherry Street, Atlanta, GA 30332-0170, United states.
| | - Eve-Marie Quintin
- School of Psychology, Georgia Institute of Technology, 654 Cherry Street, Atlanta, GA 30332-0170, United states.
| | - Ursula Saelzler
- School of Psychology, Georgia Institute of Technology, 654 Cherry Street, Atlanta, GA 30332-0170, United states.
| | - Kristen M Kennedy
- Center for Vital Longevity, School of Behavioral and Brain Sciences, University of Texas at Dallas, 1600 Viceroy Drive, Suite 800, Dallas, TX, 75235, United states.
| | - Christopher Hertzog
- School of Psychology, Georgia Institute of Technology, 654 Cherry Street, Atlanta, GA 30332-0170, United states.
| | - Scott D Moffat
- School of Psychology, Georgia Institute of Technology, 654 Cherry Street, Atlanta, GA 30332-0170, United states.
| |
Collapse
|
27
|
Kogelman B, Khmelinskii A, Verhaart I, van Vliet L, Bink DI, Aartsma-Rus A, van Putten M, van der Weerd L. Influence of full-length dystrophin on brain volumes in mouse models of Duchenne muscular dystrophy. PLoS One 2018; 13:e0194636. [PMID: 29601589 PMCID: PMC5877835 DOI: 10.1371/journal.pone.0194636] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 03/07/2018] [Indexed: 11/23/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) affects besides muscle also the brain, resulting in memory and behavioral problems. The consequences of dystrophinopathy on gross macroscopic alterations are unclear. To elucidate the effect of full-length dystrophin expression on brain morphology, we used high-resolution post-mortem MRI in mouse models that either express 0% (mdx), 100% (BL10) or a low amount of full-length dystrophin (mdx-XistΔhs). While absence or low amounts of full-length dystrophin did not significantly affect whole brain volume and skull morphology, we found differences in volume of individual brain structures. The results are in line with observations in humans, where whole brain volume was found to be reduced only in patients lacking both full-length dystrophin and the shorter isoform Dp140.
Collapse
Affiliation(s)
- Bauke Kogelman
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Artem Khmelinskii
- Division of Image Processing, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
- Percuros B.V., Enschede, the Netherlands
| | - Ingrid Verhaart
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Laura van Vliet
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Diewertje I. Bink
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Maaike van Putten
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Louise van der Weerd
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
28
|
Low left amygdala volume is associated with a longer duration of unipolar depression. J Neural Transm (Vienna) 2017; 125:229-238. [DOI: 10.1007/s00702-017-1811-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/08/2017] [Indexed: 02/08/2023]
|
29
|
Bramble MS, Lipson A, Vashist N, Vilain E. Effects of chromosomal sex and hormonal influences on shaping sex differences in brain and behavior: Lessons from cases of disorders of sex development. J Neurosci Res 2017; 95:65-74. [PMID: 27841933 DOI: 10.1002/jnr.23832] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/30/2016] [Accepted: 06/20/2016] [Indexed: 01/15/2023]
Abstract
Sex differences in brain development and postnatal behavior are determined largely by genetic sex and in utero gonadal hormone secretions. In humans however, determining the weight that each of these factors contributes remains a challenge because social influences should also be considered. Cases of disorders of sex development (DSD) provide unique insight into how mutations in genes responsible for gonadal formation can perturb the subsequent developmental hormonal milieu and elicit changes in normal human brain maturation. Specific forms of DSDs such as complete androgen insensitivity syndrome (CAIS), congenital adrenal hyperplasia (CAH), and 5α-reductase deficiency syndrome have variable effects between males and females, and the developmental outcomes of such conditions are largely dependent on sex chromosome composition. Medical and psychological works focused on CAH, CAIS, and 5α-reductase deficiency have helped form the foundation for understanding the roles of genetic and hormonal factors necessary for guiding human brain development. Here we highlight how the three aforementioned DSDs contribute to brain and behavioral phenotypes that can uniquely affect 46,XY and 46,XX individuals in dramatically different fashions. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Matthew S Bramble
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Allen Lipson
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Neerja Vashist
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Eric Vilain
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
30
|
Dieleman N, Koek HL, Hendrikse J. Short-term mechanisms influencing volumetric brain dynamics. NEUROIMAGE-CLINICAL 2017; 16:507-513. [PMID: 28971004 PMCID: PMC5609861 DOI: 10.1016/j.nicl.2017.09.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 07/28/2017] [Accepted: 09/04/2017] [Indexed: 12/14/2022]
Abstract
With the use of magnetic resonance imaging (MRI) and brain analysis tools, it has become possible to measure brain volume changes up to around 0.5%. Besides long-term brain changes caused by atrophy in aging or neurodegenerative disease, short-term mechanisms that influence brain volume may exist. When we focus on short-term changes of the brain, changes may be either physiological or pathological. As such determining the cause of volumetric dynamics of the brain is essential. Additionally for an accurate interpretation of longitudinal brain volume measures by means of neurodegeneration, knowledge about the short-term changes is needed. Therefore, in this review, we discuss the possible mechanisms influencing brain volumes on a short-term basis and set-out a framework of MRI techniques to be used for volumetric changes as well as the used analysis tools. 3D T1-weighted images are the images of choice when it comes to MRI of brain volume. These images are excellent to determine brain volume and can be used together with an analysis tool to determine the degree of volume change. Mechanisms that decrease global brain volume are: fluid restriction, evening MRI measurements, corticosteroids, antipsychotics and short-term effects of pathological processes like Alzheimer's disease, hypertension and Diabetes mellitus type II. Mechanisms increasing the brain volume include fluid intake, morning MRI measurements, surgical revascularization and probably medications like anti-inflammatory drugs and anti-hypertensive medication. Exercise was found to have no effect on brain volume on a short-term basis, which may imply that dehydration caused by exercise differs from dehydration by fluid restriction. In the upcoming years, attention should be directed towards studies investigating physiological short-term changes within the light of long-term pathological changes. Ultimately this may lead to a better understanding of the physiological short-term effects of pathological processes and may aid in early detection of these diseases. Fluid-restriction, evening MRI, corticosteroids, & antipsychotics decrease volume Fluid-intake, morning MRI, surgical revascularization & medications increase volume Short-term changes within the light of long-term pathological changes should be investigated Short-term changes may introduce bias in longitudinal data
Collapse
Affiliation(s)
- Nikki Dieleman
- Department of Radiology, University Medical Center Utrecht, The Netherlands
| | - Huiberdina L Koek
- Department of Geriatrics, University Medical Center Utrecht, The Netherlands
| | - Jeroen Hendrikse
- Department of Radiology, University Medical Center Utrecht, The Netherlands
| |
Collapse
|
31
|
Abadie D, Essilini A, Fulda V, Gouraud A, Yéléhé-Okouma M, Micallef J, Montastruc F, Montastruc JL. Drug-induced panic attacks: Analysis of cases registered in the French pharmacovigilance database. J Psychiatr Res 2017; 90:60-66. [PMID: 28231495 DOI: 10.1016/j.jpsychires.2017.02.008] [Citation(s) in RCA: 9] [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] [Received: 06/03/2016] [Revised: 02/03/2017] [Accepted: 02/08/2017] [Indexed: 12/26/2022]
Abstract
BACKGROUND The potential role of drugs in the onset of panic attacks (PAs) is poorly understood. AIM The objective of our study was to characterize drug-induced PAs. METHOD We performed an analysis of PAs registered in the French pharmacovigilance database between 01/01/1985 and 05/11/2014. RESULTS Among the 163 recorded cases, 136 (83.4%) were directly related to drugs, mainly antidepressants (11.3%, mainly serotonin reuptake inhibitors), mefloquine (7.2%), isotretinoin (5.2%), rimonabant (3.6%) and corticosteroids (4.7%). PAs are labelled in the Summary of Product Characteristics (SmPC) for a minority (8.6%) of these drugs. In 31.4% of these cases, withdrawal of the suspected drug was performed more than a week after the onset of PAs. PAs could also be secondary to another adverse drug reaction (ADR; n = 14, 8.6%), mainly an allergy to antineoplastic or immunomodulating agents. In 13 cases (8.0%), PAs occurred during a drug-withdrawal syndrome, mainly after benzodiazepines or opioids. Most cases (73%) involved patients without any previous psychiatric disorder. CONCLUSION This is the first pharmacoepidemiological study about iatrogenic PAs. Beside antidepressants, the most often encountered drugs are not indicated for psychiatric diseases. This study also reveals that iatrogenic PAs mostly occur in patients without any psychiatric medical history and that PAs can be triggered by another ADR. Lastly, the many cases with delayed management underline the need to raise awareness of this relatively unknown ADR among physicians, especially since PAs are generally not labelled in SmPCs of the suspected drugs.
Collapse
Affiliation(s)
- Delphine Abadie
- Department of Medical and Clinical Pharmacology, Toulouse University Hospital, Faculty of Medicine, 37 allées Jules Guesde, 31000 Toulouse, France.
| | - Anaïs Essilini
- Department of Medical and Clinical Pharmacology, Toulouse University Hospital, Faculty of Medicine, 37 allées Jules Guesde, 31000 Toulouse, France
| | - Virginie Fulda
- Regional Pharmacovigilance Center, Hôpital Européen Georges Pompidou, 20-40 rue Leblanc, 75015 Paris, France
| | - Aurore Gouraud
- Regional Pharmacovigilance Center, Hospices Civils de Lyon, 162 avenue Lacassagne, 69424 Lyon, France
| | - Mélissa Yéléhé-Okouma
- Regional Pharmacovigilance Center, Hôpitaux de Nancy, 29 Avenue du Maréchal de Lattre de Tassigny, 54035 Nancy, France
| | - Joëlle Micallef
- Regional Pharmacovigilance Center, Department of Medical and Clinical Pharmacology, Hôpital Sainte-Marguerite AP-HM, 270 boulevard de Saint-Marguerite, 13009 Marseille, France
| | - François Montastruc
- Department of Medical and Clinical Pharmacology, Toulouse University Hospital, Faculty of Medicine, 37 allées Jules Guesde, 31000 Toulouse, France
| | - Jean Louis Montastruc
- Department of Medical and Clinical Pharmacology, Toulouse University Hospital, Faculty of Medicine, 37 allées Jules Guesde, 31000 Toulouse, France
| |
Collapse
|
32
|
Han KM, Won E, Sim Y, Kang J, Han C, Kim YK, Kim SH, Joe SH, Lee MS, Tae WS, Ham BJ. Influence of FKBP5 polymorphism and DNA methylation on structural changes of the brain in major depressive disorder. Sci Rep 2017; 7:42621. [PMID: 28198448 PMCID: PMC5309810 DOI: 10.1038/srep42621] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 01/13/2017] [Indexed: 12/17/2022] Open
Abstract
A single nucleotide polymorphism of rs1360780 in the FKBP5 gene is associated with a predisposition to developing major depressive disorder (MDD). We investigated the interactive effects of FKBP5 rs1360780 allelic variants, DNA methylation, and the diagnosis of MDD on structural changes of the entire brain. One hundred and fourteen patients with MDD and eighty-eight healthy controls underwent T1-weighted structural magnetic resonance imaging and FKBP5 rs1360780 genotyping, including DNA methylation of intron 7. We analyzed the volume of cortical and subcortical regions and cortical thickness using FreeSurfer. Significant genotype-by-diagnosis interactions were observed for volumes of the left pars triangularis, supramarginal gyrus, superior parietal lobule, right frontomarginal, and posterior midcingulate gyrus. The T allele was associated with significant volume reductions in these brain regions only in the MDD group except for the right posterior midcingulate gyrus. FKBP5 DNA methylation showed a positive correlation with the thickness of the right transverse frontopolar gyrus in the C allele homozygote group. Our findings suggest that the FKBP5 gene and its epigenetic changes could have influence on morphologic changes of several brain regions involved in emotion regulation, and that this process may be associated with the development of MDD.
Collapse
Affiliation(s)
- Kyu-Man Han
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Eunsoo Won
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Youngbo Sim
- Brain Convergence Research Center, Korea University Anam Hospital, Seoul, Republic of Korea
| | - June Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Changsu Han
- Department of Psychiatry, Korea University Ansan Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yong-Ku Kim
- Department of Psychiatry, Korea University Ansan Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seung-Hyun Kim
- Department of Psychiatry, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sook-Haeng Joe
- Department of Psychiatry, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Min-Soo Lee
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Woo-Suk Tae
- Brain Convergence Research Center, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Byung-Joo Ham
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea.,Brain Convergence Research Center, Korea University Anam Hospital, Seoul, Republic of Korea
| |
Collapse
|
33
|
Marwha D, Halari M, Eliot L. Meta-analysis reveals a lack of sexual dimorphism in human amygdala volume. Neuroimage 2016; 147:282-294. [PMID: 27956206 DOI: 10.1016/j.neuroimage.2016.12.021] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/27/2016] [Accepted: 12/08/2016] [Indexed: 12/31/2022] Open
Abstract
The amygdala plays a key role in many affective behaviors and psychiatric disorders that differ between men and women. To test whether human amygdala volume (AV) differs reliably between the sexes, we performed a systematic review and meta-analysis of AVs reported in MRI studies of age-matched healthy male and female groups. Using four search strategies, we identified 46 total studies (58 matched samples) from which we extracted effect sizes for the sex difference in AV. All data were converted to Hedges g values and pooled effect sizes were calculated using a random-effects model. Each dataset was further meta-regressed against study year and average participant age. We found that uncorrected amygdala volume is about 10% larger in males, with pooled sex difference effect sizes of g=0.581 for right amygdala (κ=28, n=2022), 0.666 for left amygdala (κ=28, n=2006), and 0.876 for bilateral amygdala (κ=16, n=1585) volumes (all p values < 0.001). However, this difference is comparable to the sex differences in intracranial volume (ICV; g=1.186, p<.001, 11.9% larger in males, κ=11) and total brain volume (TBV; g=1.278, p<0.001, 11.5% larger in males, κ=15) reported in subsets of the same studies, suggesting the sex difference in AV is a product of larger brain size in males. Among studies reporting AVs normalized for ICV or TBV, sex difference effect sizes were small and not statistically significant: g=0.171 for the right amygdala (p=0.206, κ=13, n=1560); 0.233 for the left amygdala (p=0.092, κ=12, n=1512); and 0.257 for bilateral volume (p=0.131, κ=5, n=1629). These values correspond to less than 0.1% larger corrected right AV and 2.5% larger corrected left AV in males compared to females. In summary, AV is not selectively enhanced in human males, as often claimed. Although we cannot rule out subtle male-female group differences, it is not accurate to refer to the human amygdala as "sexually dimorphic."
Collapse
Affiliation(s)
- Dhruv Marwha
- Department of Neuroscience, Chicago Medical School, Rosalind Franklin University of Medicine & Science, United States
| | - Meha Halari
- Department of Neuroscience, Chicago Medical School, Rosalind Franklin University of Medicine & Science, United States
| | - Lise Eliot
- Department of Neuroscience, Chicago Medical School, Rosalind Franklin University of Medicine & Science, United States.
| |
Collapse
|
34
|
Shirayama Y, Takahashi M, Osone F, Hara A, Okubo T. Myo-inositol, Glutamate, and Glutamine in the Prefrontal Cortex, Hippocampus, and Amygdala in Major Depression. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2016; 2:196-204. [PMID: 29560915 DOI: 10.1016/j.bpsc.2016.11.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/13/2016] [Accepted: 11/28/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND The brains of patients with depression exhibit many changes in various regions. Recently, proton magnetic resonance spectroscopy has been used to measure brain metabolites, using saturation bands to shape the volume of interest. Our a priori hypothesis was that myo-inositol and glutamate were downregulated in the hippocampus and amygdala in depression. METHODS We measured brain metabolites from the medial prefrontal cortex, hippocampus, and amygdala of 22 drug-naïve, first-episode patients with major depressive disorder and 27 healthy control subjects using 3T proton magnetic resonance spectroscopy. RESULTS Compared with healthy control subjects, patients showed statistically significant reductions in myo-inositol levels in all three regions and reductions in glutamate levels in the medial prefrontal cortex. Furthermore, we found significant decreases in the ratios of glutamate to creatine plus phosphocreatine in the medial prefrontal cortex and amygdala. Additionally, the ratios of glutamine to creatine plus phosphocreatine were also decreased in all three regions examined, although not all the participants presented reliable data. Finally, glutamate levels in the medial prefrontal cortex and amygdala have significant correlations with executive function and those in the hippocampus with memory function. Hippocampal myo-inositol was significantly related to blood cortisol. CONCLUSIONS Our findings indicated abnormal myo-inositol, glutamate, and glutamine levels in the brains of major depressive disorder patients.
Collapse
Affiliation(s)
- Yukihiko Shirayama
- Department of Psychiatry, Teikyo University Chiba Medical Center, Ichihara, Chiba, Japan.
| | - Michio Takahashi
- Department of Psychiatry, Teikyo University Chiba Medical Center, Ichihara, Chiba, Japan
| | - Fumio Osone
- Department of Radiology, Teikyo University Chiba Medical Center, Ichihara, Chiba, Japan
| | - Akira Hara
- Department of Radiology, Teikyo University Chiba Medical Center, Ichihara, Chiba, Japan
| | - Toshiyuki Okubo
- Department of Radiology, Teikyo University Chiba Medical Center, Ichihara, Chiba, Japan
| |
Collapse
|
35
|
Doorenweerd N, Dumas EM, Ghariq E, Schmid S, Straathof CSM, Roest AAW, Wokke BH, van Zwet EW, Webb AG, Hendriksen JGM, van Buchem MA, Verschuuren JJGM, Asllani I, Niks EH, van Osch MJP, Kan HE. Decreased cerebral perfusion in Duchenne muscular dystrophy patients. Neuromuscul Disord 2016; 27:29-37. [PMID: 27927595 DOI: 10.1016/j.nmd.2016.10.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/17/2016] [Accepted: 10/11/2016] [Indexed: 10/20/2022]
Abstract
Duchenne muscular dystrophy is caused by dystrophin gene mutations which lead to the absence of the protein dystrophin. A significant proportion of patients suffer from learning and behavioural disabilities, in addition to muscle weakness. We have previously shown that these patients have a smaller total brain and grey matter volume, and altered white matter microstructure compared to healthy controls. Patients with more distal gene mutations, predicted to affect dystrophin isoforms Dp140 and Dp427, showed greater grey matter reduction. Now, we studied if cerebral blood flow in Duchenne muscular dystrophy patients is altered, since cerebral expression of dystrophin also occurs in vascular endothelial cells and astrocytes associated with cerebral vasculature. T1-weighted anatomical and pseudo-continuous arterial spin labeling cerebral blood flow images were obtained from 26 patients and 19 age-matched controls (ages 8-18 years) on a 3 tesla MRI scanner. Group comparisons of cerebral blood flow were made with and without correcting for grey matter volume using partial volume correction. Results showed that patients had a lower cerebral blood flow than controls (40.0 ± 6.4 and 47.8 ± 6.3 mL/100 g/min respectively, p = 0.0002). This reduction was independent of grey matter volume, suggesting that they are two different aspects of the pathophysiology. Cerebral blood flow was lowest in patients lacking Dp140. There was no difference in CBF between ambulant and non-ambulant patients. Only three patients showed a reduced left ventricular ejection fraction. No correlation between cerebral blood flow and age was found. Our results indicate that cerebral perfusion is reduced in Duchenne muscular dystrophy patients independent of the reduced grey matter volume.
Collapse
Affiliation(s)
- Nathalie Doorenweerd
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, The Netherlands; Leiden Institute for Brain and Cognition, Leiden, The Netherlands; Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Eve M Dumas
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Eidrees Ghariq
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, The Netherlands; Leiden Institute for Brain and Cognition, Leiden, The Netherlands
| | - Sophie Schmid
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, The Netherlands; Leiden Institute for Brain and Cognition, Leiden, The Netherlands
| | - Chiara S M Straathof
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Arno A W Roest
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Beatrijs H Wokke
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik W van Zwet
- Department of Medical Statistics, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrew G Webb
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, The Netherlands
| | - Jos G M Hendriksen
- Department of Medical Statistics, Leiden University Medical Center, Leiden, The Netherlands; Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Mark A van Buchem
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Iris Asllani
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, USA
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Matthias J P van Osch
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, The Netherlands; Leiden Institute for Brain and Cognition, Leiden, The Netherlands
| | - Hermien E Kan
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, The Netherlands; Leiden Institute for Brain and Cognition, Leiden, The Netherlands
| |
Collapse
|
36
|
Rifkin-Graboi A, Kong L, Sim LW, Sanmugam S, Broekman BFP, Chen H, Wong E, Kwek K, Saw SM, Chong YS, Gluckman PD, Fortier MV, Pederson D, Meaney MJ, Qiu A. Maternal sensitivity, infant limbic structure volume and functional connectivity: a preliminary study. Transl Psychiatry 2015; 5:e668. [PMID: 26506054 PMCID: PMC4930120 DOI: 10.1038/tp.2015.133] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 07/02/2015] [Accepted: 07/22/2015] [Indexed: 11/30/2022] Open
Abstract
Mechanisms underlying the profound parental effects on cognitive, emotional and social development in humans remain poorly understood. Studies with nonhuman models suggest variations in parental care affect the limbic system, influential to learning, autobiography and emotional regulation. In some research, nonoptimal care relates to decreases in neurogenesis, although other work suggests early-postnatal social adversity accelerates the maturation of limbic structures associated with emotional learning. We explored whether maternal sensitivity predicts human limbic system development and functional connectivity patterns in a small sample of human infants. When infants were 6 months of age, 20 mother-infant dyads attended a laboratory-based observational session and the infants underwent neuroimaging at the same age. After considering age at imaging, household income and postnatal maternal anxiety, regression analyses demonstrated significant indirect associations between maternal sensitivity and bilateral hippocampal volume at six months, with the majority of associations between sensitivity and the amygdala demonstrating similar indirect, but not significant results. Moreover, functional analyses revealed direct associations between maternal sensitivity and connectivity between the hippocampus and areas important for emotional regulation and socio-emotional functioning. Sensitivity additionally predicted indirect associations between limbic structures and regions related to autobiographical memory. Our volumetric results are consistent with research indicating accelerated limbic development in response to early social adversity, and in combination with our functional results, if replicated in a larger sample, may suggest that subtle, but important, variations in maternal care influence neuroanatomical trajectories important to future cognitive and emotional functioning.
Collapse
Affiliation(s)
- A Rifkin-Graboi
- Integrative Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Singapore,Integrative Neuroscience Program, Singapore Institute for Clinical Sciences, Brenner Centre for Molecular Medicine 30 Medical Drive, Singapore 117609, Singapore. E-mail:
| | - L Kong
- Department of Biomedical Engineering and Clinical Imaging Research Center, National University of Singapore, Singapore, Singapore
| | - L W Sim
- Integrative Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Singapore
| | - S Sanmugam
- Integrative Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Singapore
| | - B F P Broekman
- Integrative Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Singapore,Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore, Singapore
| | - H Chen
- Department of Psychological Medicine, KK Women's and Children's Hospital, Duke-National University of Singapore, Singapore, Singapore
| | - E Wong
- Integrative Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Singapore
| | - K Kwek
- Department of Maternal Fetal Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - S-M Saw
- Department of Epidemiology, Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Y-S Chong
- Integrative Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Singapore,Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore, Singapore
| | - P D Gluckman
- Human Development, Singapore Institute for Clinical Sciences, Singapore, Singapore,Liggins Institute, University of Auckland, Auckland, New Zealand
| | - M V Fortier
- Department of Diagnostic and Interventional Imaging, KK Women's and Children's Hospital, Singapore, Singapore
| | - D Pederson
- Department of Psychology, University of Western Ontario, London, Ontario, Canada
| | - M J Meaney
- Integrative Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Singapore,Department of Neurosciences, Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada,Sackler Program for Epigenetics and Psychobiology, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
| | - A Qiu
- Integrative Neuroscience Program, Singapore Institute for Clinical Sciences, Singapore, Singapore,Department of Biomedical Engineering and Clinical Imaging Research Center, National University of Singapore, Singapore, Singapore,Department of Biomedical Engineering, National University of Singapore, 9 Engineering Drive 1, Block EA #03-12, Singapore 117576, Singapore. E-mail:
| |
Collapse
|
37
|
Andela CD, van Haalen FM, Ragnarsson O, Papakokkinou E, Johannsson G, Santos A, Webb SM, Biermasz NR, van der Wee NJA, Pereira AM. MECHANISMS IN ENDOCRINOLOGY: Cushing's syndrome causes irreversible effects on the human brain: a systematic review of structural and functional magnetic resonance imaging studies. Eur J Endocrinol 2015; 173:R1-14. [PMID: 25650405 DOI: 10.1530/eje-14-1101] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/03/2015] [Indexed: 11/08/2022]
Abstract
BACKGROUND Cushing's syndrome (CS) is characterized by excessive exposure to cortisol, and is associated with both metabolic and behavioral abnormalities. Symptoms improve substantially after biochemical cure, but may persist during long-term remission. The causes for persistent morbidity are probably multi-factorial, including a profound effect of cortisol excess on the brain, a major target area for glucocorticoids. OBJECTIVE To review publications evaluating brain characteristics in patients with CS using magnetic resonance imaging (MRI). METHODS Systematic review of literature published in PubMed, Embase, Web of Knowledge, and Cochrane databases. RESULTS Nineteen studies using MRI in patients with CS were selected, including studies in patients with active disease, patients in long-term remission, and longitudinal studies, covering a total of 339 unique patients. Patients with active disease showed smaller hippocampal volumes, enlarged ventricles, and cerebral atrophy as well as alterations in neurochemical concentrations and functional activity. After abrogation of cortisol excess, the reversibility of structural and neurochemical alterations was incomplete after long-term remission. MRI findings were related to clinical characteristics (i.e., cortisol levels, duration of exposure to hypercortisolism, current age, age at diagnosis, and triglyceride levels) and behavioral outcome (i.e., cognitive and emotional functioning, mood, and quality of life). CONCLUSION Patients with active CS demonstrate brain abnormalities, which only partly recover after biochemical cure, because these still occur even after long-term remission. CS might be considered as a human model of nature that provides a keyhole perspective of the neurotoxic effects of exogenous glucocorticoids on the brain.
Collapse
Affiliation(s)
- Cornelie D Andela
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The NetherlandsLeiden Institute for Brain and CognitionLeiden, The NetherlandsDepartment of Endocrinology Diabetes and MetabolismSahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenEndocrinology/Medicine DepartmentsHospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), ISCIII, IIB-Sant Pau, Universitat Autònoma de Barcelona (UAB), Barcelona, SpainDepartment of PsychiatryLeiden University Medical Center, Leiden, The Netherlands Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The NetherlandsLeiden Institute for Brain and CognitionLeiden, The NetherlandsDepartment of Endocrinology Diabetes and MetabolismSahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenEndocrinology/Medicine DepartmentsHospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), ISCIII, IIB-Sant Pau, Universitat Autònoma de Barcelona (UAB), Barcelona, SpainDepartment of PsychiatryLeiden University Medical Center, Leiden, The Netherlands
| | - Femke M van Haalen
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The NetherlandsLeiden Institute for Brain and CognitionLeiden, The NetherlandsDepartment of Endocrinology Diabetes and MetabolismSahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenEndocrinology/Medicine DepartmentsHospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), ISCIII, IIB-Sant Pau, Universitat Autònoma de Barcelona (UAB), Barcelona, SpainDepartment of PsychiatryLeiden University Medical Center, Leiden, The Netherlands
| | - Oskar Ragnarsson
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The NetherlandsLeiden Institute for Brain and CognitionLeiden, The NetherlandsDepartment of Endocrinology Diabetes and MetabolismSahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenEndocrinology/Medicine DepartmentsHospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), ISCIII, IIB-Sant Pau, Universitat Autònoma de Barcelona (UAB), Barcelona, SpainDepartment of PsychiatryLeiden University Medical Center, Leiden, The Netherlands
| | - Eleni Papakokkinou
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The NetherlandsLeiden Institute for Brain and CognitionLeiden, The NetherlandsDepartment of Endocrinology Diabetes and MetabolismSahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenEndocrinology/Medicine DepartmentsHospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), ISCIII, IIB-Sant Pau, Universitat Autònoma de Barcelona (UAB), Barcelona, SpainDepartment of PsychiatryLeiden University Medical Center, Leiden, The Netherlands
| | - Gudmundur Johannsson
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The NetherlandsLeiden Institute for Brain and CognitionLeiden, The NetherlandsDepartment of Endocrinology Diabetes and MetabolismSahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenEndocrinology/Medicine DepartmentsHospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), ISCIII, IIB-Sant Pau, Universitat Autònoma de Barcelona (UAB), Barcelona, SpainDepartment of PsychiatryLeiden University Medical Center, Leiden, The Netherlands
| | - Alicia Santos
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The NetherlandsLeiden Institute for Brain and CognitionLeiden, The NetherlandsDepartment of Endocrinology Diabetes and MetabolismSahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenEndocrinology/Medicine DepartmentsHospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), ISCIII, IIB-Sant Pau, Universitat Autònoma de Barcelona (UAB), Barcelona, SpainDepartment of PsychiatryLeiden University Medical Center, Leiden, The Netherlands
| | - Susan M Webb
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The NetherlandsLeiden Institute for Brain and CognitionLeiden, The NetherlandsDepartment of Endocrinology Diabetes and MetabolismSahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenEndocrinology/Medicine DepartmentsHospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), ISCIII, IIB-Sant Pau, Universitat Autònoma de Barcelona (UAB), Barcelona, SpainDepartment of PsychiatryLeiden University Medical Center, Leiden, The Netherlands
| | - Nienke R Biermasz
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The NetherlandsLeiden Institute for Brain and CognitionLeiden, The NetherlandsDepartment of Endocrinology Diabetes and MetabolismSahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenEndocrinology/Medicine DepartmentsHospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), ISCIII, IIB-Sant Pau, Universitat Autònoma de Barcelona (UAB), Barcelona, SpainDepartment of PsychiatryLeiden University Medical Center, Leiden, The Netherlands Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The NetherlandsLeiden Institute for Brain and CognitionLeiden, The NetherlandsDepartment of Endocrinology Diabetes and MetabolismSahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenEndocrinology/Medicine DepartmentsHospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), ISCIII, IIB-Sant Pau, Universitat Autònoma de Barcelona (UAB), Barcelona, SpainDepartment of PsychiatryLeiden University Medical Center, Leiden, The Netherlands
| | - Nic J A van der Wee
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The NetherlandsLeiden Institute for Brain and CognitionLeiden, The NetherlandsDepartment of Endocrinology Diabetes and MetabolismSahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenEndocrinology/Medicine DepartmentsHospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), ISCIII, IIB-Sant Pau, Universitat Autònoma de Barcelona (UAB), Barcelona, SpainDepartment of PsychiatryLeiden University Medical Center, Leiden, The Netherlands Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The NetherlandsLeiden Institute for Brain and CognitionLeiden, The NetherlandsDepartment of Endocrinology Diabetes and MetabolismSahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenEndocrinology/Medicine DepartmentsHospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), ISCIII, IIB-Sant Pau, Universitat Autònoma de Barcelona (UAB), Barcelona, SpainDepartment of PsychiatryLeiden University Medical Center, Leiden, The Netherlands
| | - Alberto M Pereira
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The NetherlandsLeiden Institute for Brain and CognitionLeiden, The NetherlandsDepartment of Endocrinology Diabetes and MetabolismSahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenEndocrinology/Medicine DepartmentsHospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), ISCIII, IIB-Sant Pau, Universitat Autònoma de Barcelona (UAB), Barcelona, SpainDepartment of PsychiatryLeiden University Medical Center, Leiden, The Netherlands Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The NetherlandsLeiden Institute for Brain and CognitionLeiden, The NetherlandsDepartment of Endocrinology Diabetes and MetabolismSahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, SwedenEndocrinology/Medicine DepartmentsHospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), ISCIII, IIB-Sant Pau, Universitat Autònoma de Barcelona (UAB), Barcelona, SpainDepartment of PsychiatryLeiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
38
|
Organic changes in the central nervous system in children on chronic inhaled corticosteroid therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 839:41-5. [PMID: 25298261 DOI: 10.1007/5584_2014_48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Treatment of asthma, the most common chronic respiratory disease in children, includes long-term inhaled corticosteroids (ICS). The purpose of this study was to analyze the impact of chronic inhaled steroids in children with asthma on changes in the central nervous system (CNS). Eleven children (8-17 years) on at least 4 years inhaled corticosteroid therapy were assessed with magnetic resonance imaging (MRI). All participants underwent a pediatric and neurological examination and spirometry. MRI data were obtained using a 1.5 T scanner with parallel imaging capability. Structural images consisted of axial T1, T2 using turbo spin echo, FLAIR and DWI sequences using typical parameters. Images were assessed in three planes (axial, coronal, and sagittal). Pediatric and neurological examination were normal in all children. In six, the MRI studies revealed small subcortical hyperintense foci. Three had more than five lesions, all of which were smaller than 3 mm. Features of mild supratentorial cortical atrophy were apparent in four. The cerebellum was unremarkable in all children imaged. In conclusion, patients receiving chronic inhaled corticosteroids had small subcortical hyperintense foci and features of mild supratentorial cortical atrophy. These findings suggest that ICS exposure may be associated with the development of organic changes in CNS. Further studies are needed to detail the extent of brain ICS-induced changes in children on chronic inhalative corticosteroid therapy as well as delayed impact of these changes on psychomotor functioning in adulthood.
Collapse
|
39
|
Daskalakis NP, Binder EB. Schizophrenia in the spectrum of gene-stress interactions: the FKBP5 example. Schizophr Bull 2015; 41:323-9. [PMID: 25592294 PMCID: PMC4332957 DOI: 10.1093/schbul/sbu189] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Many studies have demonstrated that genotype (G) interacts with adverse life experiences (E) to produce individual differences in vulnerability and resilience to mental disorders, including schizophrenia. Genetic susceptibility to stress and the timing of the environmental exposure(s) are relevant for these interactions and represent common risk factors. We take the example of the FKBP5 gene to illustrate G × E interactions that predict pleiotropic psychiatric outcomes, including schizophrenia.
Collapse
Affiliation(s)
- Nikolaos P. Daskalakis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY;,Mental Health Care Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY;,*To whom correspondence should be addressed; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, US; tel: +1-212-241-0250, fax: +1-212-828-4221, e-mail:
| | - Elisabeth B. Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany;,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
40
|
Bao F, Wang Y, Liu J, Mao C, Ma S, Guo C, Ding H, Zhang M. Structural changes in the CNS of patients with hemifacial spasm. Neuroscience 2015; 289:56-62. [DOI: 10.1016/j.neuroscience.2014.12.070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 11/26/2014] [Accepted: 12/23/2014] [Indexed: 12/20/2022]
|
41
|
Agostini A, Campieri M, Bertani A, Scarcelli A, Ballotta D, Calabrese C, Rizzello F, Gionchetti P, Nichelli P, Benuzzi F. Absence of change in the gray matter volume of patients with ulcerative colitis in remission: a voxel based morphometry study. Biopsychosoc Med 2015; 9:1. [PMID: 25614759 PMCID: PMC4302580 DOI: 10.1186/s13030-014-0028-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 12/19/2014] [Indexed: 12/26/2022] Open
Abstract
Background Recent neuroimaging studies have investigated the brain involvement in patients with Crohn's disease (CD) and Ulcerative Colitis (UC). Functional studies found abnormalities in cognitive and emotional functions in CD and UC, while a voxel based morphometry (VBM) study found morphological changes in CD. We conducted a VBM study to compare the gray matter (GM) volume of UC patients and controls. Methods Eighteen UC patients in remission and eighteen healthy controls underwent structural MRI. VBM is a fully automated technique allowing identification of regional differences in the amount of GM, which enables an objective analysis of the whole brain. VBM was used for comparisons between patients and controls. Results UC patients were all in remission and had a mild clinical course. There were no differences between patients and controls in GM volume. Conclusion The brain morphology of patients with UC in remission is similar to controls. The lack of GM abnormalities in UC patients might reflect the mild clinical course of the inflammatory bowel disorder. Further research involving patients with different degrees of disease severity or during flares could shed more light on potential brain structural changes in UC.
Collapse
Affiliation(s)
- Alessandro Agostini
- Department of Psychology, University of Bologna, Bologna, Italy ; Department of Clinical Medicine, IBD Unit, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Massimo Campieri
- Department of Clinical Medicine, IBD Unit, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Angela Bertani
- Department of Gastroenterology, IBD Unit, Policlinico Hospital, Modena, Italy
| | - Antonella Scarcelli
- Department of Gastroenterology, IBD Unit, Policlinico Hospital, Modena, Italy
| | - Daniela Ballotta
- Department of Biomedical, Metabolic Sciences, and Neurosciences, Nuovo Ospedale Civile S. Agostino-Estense, University of Modena and Reggio Emilia, Modena, Italy
| | - Carlo Calabrese
- Department of Clinical Medicine, IBD Unit, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Fernando Rizzello
- Department of Clinical Medicine, IBD Unit, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Paolo Gionchetti
- Department of Clinical Medicine, IBD Unit, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Paolo Nichelli
- Department of Biomedical, Metabolic Sciences, and Neurosciences, Nuovo Ospedale Civile S. Agostino-Estense, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Benuzzi
- Department of Biomedical, Metabolic Sciences, and Neurosciences, Nuovo Ospedale Civile S. Agostino-Estense, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
42
|
Doorenweerd N, Straathof CS, Dumas EM, Spitali P, Ginjaar IB, Wokke BH, Schrans DG, van den Bergen JC, van Zwet EW, Webb A, van Buchem MA, Verschuuren JJ, Hendriksen JG, Niks EH, Kan HE. Reduced cerebral gray matter and altered white matter in boys with Duchenne muscular dystrophy. Ann Neurol 2014; 76:403-11. [DOI: 10.1002/ana.24222] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 07/08/2014] [Accepted: 07/08/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Nathalie Doorenweerd
- Department of Radiology; C. J. Gorter Center for High Field MRI, Leiden University Medical Center; Leiden
- Leiden Institute for Brain and Cognition; Leiden
- Department of Neurology; Leiden University Medical Center; Leiden
| | | | - Eve M. Dumas
- Department of Neurology; Leiden University Medical Center; Leiden
| | - Pietro Spitali
- Department of Human Genetics; Leiden University Medical Center; Leiden
| | - Ieke B. Ginjaar
- Department of Clinical Genetics; Leiden University Medical Center; Leiden
| | | | - Debby G. Schrans
- Department of Neurological Learning Disabilities; Kempenhaeghe Epilepsy Center; Heeze
| | | | - Erik W. van Zwet
- Department of Medical Statistics; Leiden University Medical Center; Leiden
| | - Andrew Webb
- Department of Radiology; C. J. Gorter Center for High Field MRI, Leiden University Medical Center; Leiden
| | - Mark A. van Buchem
- Department of Radiology; C. J. Gorter Center for High Field MRI, Leiden University Medical Center; Leiden
| | | | - Jos G. Hendriksen
- Department of Neurological Learning Disabilities; Kempenhaeghe Epilepsy Center; Heeze
- Department of Neurology; Maastricht University Medical Center; Maastricht the Netherlands
| | - Erik H. Niks
- Department of Neurology; Leiden University Medical Center; Leiden
| | - Hermien E. Kan
- Department of Radiology; C. J. Gorter Center for High Field MRI, Leiden University Medical Center; Leiden
- Leiden Institute for Brain and Cognition; Leiden
| |
Collapse
|
43
|
Pagliaccio D, Luby JL, Bogdan R, Agrawal A, Gaffrey MS, Belden AC, Botteron KN, Harms MP, Barch DM. Stress-system genes and life stress predict cortisol levels and amygdala and hippocampal volumes in children. Neuropsychopharmacology 2014; 39:1245-53. [PMID: 24304824 PMCID: PMC3957120 DOI: 10.1038/npp.2013.327] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 10/25/2013] [Accepted: 11/14/2013] [Indexed: 02/08/2023]
Abstract
Depression has been linked to increased cortisol reactivity and differences in limbic brain volumes, yet the mechanisms underlying these alterations are unclear. One main hypothesis is that stress causes these effects. This is supported by animal studies showing that chronic stress or glucocorticoid administration can lead to alterations in hippocampal and amygdala structures. Relatedly, life stress is cited as one of the major risk factors for depression and candidate gene studies have related variation in stress-system genes to increased prevalence and severity of depression. The present study tested the hypothesis that genetic profile scores combining variance across 10 single nucleotide polymorphisms from four stress-system genes (CRHR1, NR3C2, NR3C1, and FKBP5) and early life stress would predict increases in cortisol levels during laboratory stressors in 120 preschool-age children (3-5 years old), as well as hippocampal and amygdala volumes assessed with MRI in these same children at school age (7-12 years old). We found that stress-system genetic profile scores positively predicted cortisol levels while the number of stressful/traumatic life events experienced by 3-5 years old negatively predicted cortisol levels. The interaction of genetic profile scores and early life stress predicted left hippocampal and left amygdala volumes. Cortisol partially mediated the effects of genetic variation and life stress on limbic brain volumes, particularly on left amygdala volume. These results suggest that stress-related genetic and early environmental factors contribute to variation in stress cortisol reactivity and limbic brain volumes in children, phenotypes associated with depression in adulthood.
Collapse
Affiliation(s)
- David Pagliaccio
- Program in Neuroscience, Washington University in St Louis, St Louis, MO, USA,The Program in Neuroscience, Washington University in St Louis, Campus Box 1125, One Brookings Drive, St Louis, MO 63130, USA, Tel: +9146450103, Fax: +913149358790, E-mail: or
| | - Joan L Luby
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Ryan Bogdan
- Program in Neuroscience, Washington University in St Louis, St Louis, MO, USA,Department of Psychology, Washington University in St Louis, St Louis, MO, USA
| | - Arpana Agrawal
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Michael S Gaffrey
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Andrew C Belden
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Kelly N Botteron
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA,Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Michael P Harms
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Deanna M Barch
- Program in Neuroscience, Washington University in St Louis, St Louis, MO, USA,Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA,Department of Psychology, Washington University in St Louis, St Louis, MO, USA,Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| |
Collapse
|
44
|
Levita L, Bois C, Healey A, Smyllie E, Papakonstantinou E, Hartley T, Lever C. The Behavioural Inhibition System, anxiety and hippocampal volume in a non-clinical population. BIOLOGY OF MOOD & ANXIETY DISORDERS 2014; 4:4. [PMID: 24607258 PMCID: PMC4007806 DOI: 10.1186/2045-5380-4-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 02/19/2014] [Indexed: 11/16/2022]
Abstract
Background Animal studies have suggested that the hippocampus may play an important role in anxiety as part of the Behavioural Inhibition System (BIS), which mediates reactivity to threat and punishment and can predict an individual’s response to anxiety-relevant cues in a given environment. The aim of the present structural magnetic resonance imaging (MRI) study was to examine the relationship between individual differences in BIS and hippocampal structure, since this has not received sufficient attention in non-clinical populations. Thirty healthy right-handed participants with no history of alcohol or drug abuse, neurological or psychiatric disorders, or traumatic brain injury were recruited (16 male, 14 female, age 18 to 32 years). T1-weighted structural MRI scans were used to derive estimates of total intracranial volume, and hippocampal and amygdala gray matter volume using FreeSurfer. To relate brain structure to Gray’s BIS, participants completed the Sensitivity to Punishment questionnaire. They also completed questionnaires assessing other measures potentially associated with hippocampal volume (Beck Depression Inventory, Negative Life Experience Survey), and two other measures of anxiety (Spielberger Trait Anxiety Inventory and the Beck Anxiety Inventory). Results We found that high scores on the Sensitivity to Punishment scale were positively associated with hippocampal volume, and that this phenomenon was lateralized to the right side. In other words, greater levels of behavioural inhibition (BIS) were positively associated with right hippocampal volume. Conclusions Our data suggest that hippocampal volume is related to the cognitive and affective dimensions of anxiety indexed by the Sensitivity to Punishment, and support the idea that morphological differences in the hippocampal formation may be associated with behavioural inhibition contributions to anxiety.
Collapse
Affiliation(s)
- Liat Levita
- Present address: Department of Psychology, University of Sheffield, Western Bank, Sheffield S10 2TN, UK.
| | | | | | | | | | | | | |
Collapse
|
45
|
Sudheimer KD, Abelson JL, Taylor SF, Martis B, Welsh RC, Warner C, Samet M, Manduzzi A, Liberzon I. Exogenous glucocorticoids decrease subgenual cingulate activity evoked by sadness. Neuropsychopharmacology 2013; 38:826-45. [PMID: 23303057 PMCID: PMC3599059 DOI: 10.1038/npp.2012.249] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 11/02/2012] [Accepted: 11/28/2012] [Indexed: 11/09/2022]
Abstract
The glucocorticoid hormone cortisol is known to have wide-ranging effects on a variety of physiological systems, including the morphology and physiology of the amygdala and hippocampus. Disruptions of cortisol regulation and signaling are also linked with psychiatric disorders involving emotional disturbances. Although there is much evidence to suggest a relationship between cortisol signaling and the brain physiology underlying emotion, few studies have attempted to test for direct effects of cortisol on the neurophysiology of emotion. We administered exogenous synthetic cortisol (hydrocortisone, HCT) using two different dosing regimens (25 mg/day over 4 days, 100 mg single dose), in a double-blind placebo-controlled functional magnetic resonance imaging (fMRI) study. During fMRI scanning, healthy subjects viewed images designed to induce happy, sad, and neutral emotional states. Subjective emotional reactions were collected for each experimental stimulus after fMRI scanning. Mood ratings were also collected throughout the 4 days of the study. Both dose regimens of HCT resulted in decreased subgenual cingulate activation during sadness conditions. The 25 mg/day regimen also resulted in higher arousal ratings of sad stimuli. No effects of HCT were observed on any mood ratings. Few reliable effects of HCT were observed on brain activity patterns or subjective emotional responses to stimuli that were not sad. The inhibitory effects of cortisol on sadness-induced subgenual cingulate activity may have critical relevance to the pathophysiology of major depression, as both subgenual hyperactivity and decreased sensitivity to cortisol signaling have been documented in patients with depression.
Collapse
Affiliation(s)
- Keith D Sudheimer
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Guidetti G. The role of cognitive processes in vestibular disorders. HEARING, BALANCE AND COMMUNICATION 2013. [DOI: 10.3109/21695717.2013.765085] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
47
|
Liu W, Katz DA, Locke C, Daszkowski D, Wang Y, Rieser MJ, Awni W, Marek GJ, Dutta S. Clinical Safety, Pharmacokinetics, and Pharmacodynamics of the 11β-Hydroxysteroid Dehydrogenase Type 1 Inhibitor ABT-384 in Healthy Volunteers and Elderly Adults. Clin Pharmacol Drug Dev 2013; 2:133-51. [PMID: 27121668 DOI: 10.1002/cpdd.5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 10/10/2012] [Indexed: 11/10/2022]
Abstract
ABT-384 is a potent and selective inhibitor of 11β-hydroxysteroid dehydrogenase type 1 (HSD-1), the enzyme that regenerates cortisol in several tissues. Two clinical studies of ABT-384 were undertaken to assess its safety, pharmacokinetics, target engagement, and pharmacologic effects in healthy subjects. Single doses from 1 to 240 mg, and multiple doses from 1 to 100 mg once daily for 7-14 days, were administered to healthy adults. Multiple doses from 10 to 100 mg once daily for 21 days were administered to elderly subjects. A total of 103 subjects received at least 1 dose of ABT-384. A maximum-tolerated dose was not defined in either study. The pharmacokinetic profiles of ABT-384 and its active metabolite support once daily dosing. Analysis of urine cortisol metabolites demonstrated full hepatic HSD-1 inhibition with regimens from 1 mg daily, and confirmed in vitro target selectivity. Pharmacologic effects included increases of adrenocorticotrophic hormone levels, cortisol production and androgen and estradiol levels. ABT-384 has a wide therapeutic index relative to full hepatic target engagement which is relevant for indications such as diabetes and metabolic syndrome. Its therapeutic index for other potential indications such as Alzheimer's disease remains to be established.
Collapse
Affiliation(s)
- Wei Liu
- AbbVie, North Chicago, IL, USA
| | | | | | | | - Yi Wang
- AbbVie, North Chicago, IL, USA
| | | | | | | | | |
Collapse
|
48
|
Agostini A, Benuzzi F, Filippini N, Bertani A, Scarcelli A, Farinelli V, Marchetta C, Calabrese C, Rizzello F, Gionchetti P, Ercolani M, Campieri M, Nichelli P. New insights into the brain involvement in patients with Crohn's disease: a voxel-based morphometry study. Neurogastroenterol Motil 2013; 25:147-e82. [PMID: 22998431 DOI: 10.1111/nmo.12017] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Crohn's disease (CD) is a chronic intestinal disorder characterized by overproduction of inflammatory cytokines and recurrent abdominal pain. Recently, brain morphological abnormalities in the pain matrix were found in patients with chronic pain disorders including irritable bowel syndrome. To investigate potential structural brain changes associated with CD, we used magnetic resonance imaging (MRI). Furthermore, we tested whether in patients gray matter (GM) volumes correlated with disease duration. METHODS Eighteen CD patients in remission and 18 healthy controls underwent structural MRI. Voxel-based morphometry (VBM) is a fully automated technique allowing identification of regional differences in the amount of GM enabling an objective analysis of the whole brain between groups of subjects. VBM was used for comparisons and correlation analysis. KEY RESULTS With respect to controls, CD patients exhibited decreased GM volumes in portion of the frontal cortex and in the anterior midcingulate cortex. Disease duration was negatively correlated with GM volumes of several brain regions including neocortical and limbic areas. CONCLUSIONS & INFERENCES Crohn's disease is associated with brain morphological changes in cortical and subcortical structures involved in nociception, emotional, and cognitive processes. Our findings provide new insight into the brain involvement in chronic inflammatory bowel disorders.
Collapse
Affiliation(s)
- A Agostini
- Department of Psychology, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Alexander N, Klucken T, Koppe G, Osinsky R, Walter B, Vaitl D, Sammer G, Stark R, Hennig J. Interaction of the serotonin transporter-linked polymorphic region and environmental adversity: increased amygdala-hypothalamus connectivity as a potential mechanism linking neural and endocrine hyperreactivity. Biol Psychiatry 2012; 72:49-56. [PMID: 22418015 DOI: 10.1016/j.biopsych.2012.01.030] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 01/10/2012] [Accepted: 01/23/2012] [Indexed: 11/30/2022]
Abstract
BACKGROUND Gene by environment (G×E) interaction between genetic variation in the promoter region of the serotonin transporter gene (serotonin transporter-linked polymorphic region [5-HTTLPR]) and stressful life events (SLEs) has been extensively studied in the context of depression. Recent findings suggest increased neural and endocrine stress sensitivity as a possible mechanism conveying elevated vulnerability to psychopathology. Furthermore, these G×E mediated alterations very likely reflect interrelated biological processes. METHODS In the present functional magnetic resonance imaging study, amygdala reactivity to fearful stimuli was assessed in healthy male adults (n = 44), who were previously found to differ with regard to endocrine stress reactivity as a function of 5-HTTLPR × SLEs. Furthermore, functional connectivity between the amygdala and the hypothalamus was measured as a potential mechanism linking elevated neural and endocrine responses during stressful/threatening situations. The study sample was carefully preselected regarding 5-HTTLPR genotype and SLEs. RESULTS We report significant G×E interaction on neural response patterns and functional amygdala-hypothalamus connectivity. Specifically, homozygous carriers of the 5-HTTLPR S' allele with a history of SLEs (S'S'/high SLEs group) displayed elevated bilateral amygdala activation in response to fearful faces. Within the same sample, a comparable G×E interaction effect has previously been demonstrated regarding increased cortisol reactivity, indicating a cross-validation of heightened biological stress sensitivity. Furthermore, S'S'/high SLEs subjects were characterized by an increased functional coupling between the right amygdala and the hypothalamus, thus indicating a potential link between neural and endocrine hyperreactivity. CONCLUSIONS The present findings contribute to the ongoing debate on 5-HTTLPR × SLEs interaction and are discussed with respect to clinical implications.
Collapse
Affiliation(s)
- Nina Alexander
- Department of Biological Psychology, Technische Universität, Dresden, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Aas M, Navari S, Gibbs A, Mondelli V, Fisher HL, Morgan C, Morgan K, MacCabe J, Reichenberg A, Zanelli J, Fearon P, Jones PB, Murray RM, Pariante CM, Dazzan P. Is there a link between childhood trauma, cognition, and amygdala and hippocampus volume in first-episode psychosis? Schizophr Res 2012; 137:73-9. [PMID: 22353995 DOI: 10.1016/j.schres.2012.01.035] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 01/09/2012] [Accepted: 01/26/2012] [Indexed: 01/15/2023]
Abstract
Patients with psychosis have higher rates of childhood trauma, which is also associated with adverse effects on cognitive functions such as attention, concentration and mental speed, language, and verbal intelligence. Although the pathophysiological substrate for this association remains unclear, these cognitive deficits may represent the functional correlate of changes observed in relation to trauma exposure in structures such as the amygdala and the hippocampus. Interestingly, these structures are often reported as altered in psychosis. This study investigated the association between childhood trauma, cognitive function and amygdala and hippocampus volume, in first-episode psychosis. We investigated 83 patients with first-episode psychosis and 63 healthy controls. All participants underwent an MRI scan acquired with a GE Sigma 1.5-T system, and a standardized neuropsychological assessment of general cognition, memory, processing speed, executive function, visuo-spatial abilities, verbal intelligence, and language. In a subsample of the patients (N=45) information on childhood trauma was collected with the Childhood Experience of Care and Abuse Questionnaire (CECA.Q). We found that amygdala, but not hippocampus, volume was significantly smaller (p=0.001) in patients compared to healthy controls. There was a trend level interaction for hippocampus volume between group and sex (p=0.056). A history of childhood trauma was associated with both worse cognitive performance and smaller amygdala volume. This smaller amygdala appeared to mediate the relationship between childhood trauma and performance on executive function, language and verbal intelligence in patients with psychosis. This points to a complex relationship between childhood trauma exposure, cognitive function and amygdala volume in first-episode psychosis.
Collapse
Affiliation(s)
- Monica Aas
- Department of Psychosis Studies, Institute of Psychiatry, King's College London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|