51
|
Convergence of placenta biology and genetic risk for schizophrenia. Nat Med 2018; 24:792-801. [DOI: 10.1038/s41591-018-0021-y] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 03/16/2018] [Indexed: 01/16/2023]
|
52
|
Giannopoulou I, Pagida MA, Briana DD, Panayotacopoulou MT. Perinatal hypoxia as a risk factor for psychopathology later in life: the role of dopamine and neurotrophins. Hormones (Athens) 2018; 17:25-32. [PMID: 29858855 DOI: 10.1007/s42000-018-0007-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/16/2017] [Indexed: 12/26/2022]
Abstract
Brain development is influenced by various prenatal, intrapartum, and postnatal events which may interact with genotype to affect the neural and psychophysiological systems related to emotions, specific cognitive functions (e.g., attention, memory), and language abilities and thereby heighten the risk for psychopathology later in life. Fetal hypoxia (intrapartum oxygen deprivation), hypoxia-related obstetric complications, and hypoxia during the early neonatal period are major environmental risk factors shown to be associated with an increased risk for later psychopathology. Experimental models of perinatal hypoxia/ischemia (PHI) showed that fetal hypoxia-a consequence common to many birth complications in humans-results in selective long-term disturbances of the dopaminergic systems that persist in adulthood. On the other hand, neurotrophic signaling is critical for pre- and postnatal brain development due to its impact on the process of neuronal development and its reaction to perinatal stress. The aim of this review is (a) to summarize epidemiological data confirming an association of PHI with an increased risk of a range of psychiatric disorders from childhood through adolescence to adulthood, (b) to present immunohistochemical findings on human autopsy material indicating vulnerability of the dopaminergic neurons of the human neonate to PHI that could predispose infant survivors of PHI to dopamine-related neurological and/or cognitive deficits in adulthood, and
Collapse
Affiliation(s)
- Ioanna Giannopoulou
- 2nd Department of Psychiatry, National and Kapodistrian University of Athens, Athens, Greece
| | - Marianna A Pagida
- 1st Department of Psychiatry, National and Kapodistrian University of Athens, Athens, Greece
- Laboratory of Neurobiology and Histochemistry, University Mental Health Research Institute, PO Box 66517, GR-15601, Papagou, Athens, Greece
| | - Despina D Briana
- Neonatal Unit, 1st Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria T Panayotacopoulou
- 1st Department of Psychiatry, National and Kapodistrian University of Athens, Athens, Greece.
- Laboratory of Neurobiology and Histochemistry, University Mental Health Research Institute, PO Box 66517, GR-15601, Papagou, Athens, Greece.
| |
Collapse
|
53
|
Casas BS, Vitória G, do Costa MN, Madeiro da Costa R, Trindade P, Maciel R, Navarrete N, Rehen SK, Palma V. hiPSC-derived neural stem cells from patients with schizophrenia induce an impaired angiogenesis. Transl Psychiatry 2018; 8:48. [PMID: 29467462 PMCID: PMC5821759 DOI: 10.1038/s41398-018-0095-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 12/30/2017] [Indexed: 12/18/2022] Open
Abstract
Schizophrenia is a neurodevelopmental disease characterized by cerebral connectivity impairment and loss of gray matter. It was described in adult schizophrenia patients (SZP) that concentration of VEGFA, a master angiogenic factor, is decreased. Recent evidence suggests cerebral hypoperfusion related to a dysfunctional Blood Brain Barrier (BBB) in SZP. Since neurogenesis and blood-vessel formation occur in a coincident and coordinated fashion, a defect in neurovascular development could result in increased vascular permeability and, therefore, in poor functionality of the SZP's neurons. Here, we characterized the conditioned media (CM) of human induced Pluripotent Stem Cells (hiPSC)-derived Neural Stem Cells of SZP (SZP NSC) versus healthy subjects (Ctrl NSC), and its impact on angiogenesis. Our results reveal that SZP NSC have an imbalance in the secretion and expression of several angiogenic factors, among them non-canonical neuro-angiogenic guidance factors. SZP NSC migrated less and their CM was less effective in inducing migration and angiogenesis both in vitro and in vivo. Since SZP originates during embryonic brain development, our findings suggest a defective crosstalk between NSC and endothelial cells (EC) during the formation of the neuro-angiogenic niche.
Collapse
Affiliation(s)
- Bárbara S Casas
- Laboratory of Stem Cells and Development, Universidad de Chile, Santiago, Chile
| | - Gabriela Vitória
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Marcelo N do Costa
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Pablo Trindade
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Renata Maciel
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Nelson Navarrete
- Universidad de Chile Clinical Hospital, Región Metropolitana, Chile
| | - Stevens K Rehen
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil.
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Verónica Palma
- Laboratory of Stem Cells and Development, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
54
|
Hefter D, Marti HH, Gass P, Inta D. Perinatal Hypoxia and Ischemia in Animal Models of Schizophrenia. Front Psychiatry 2018; 9:106. [PMID: 29651259 PMCID: PMC5884869 DOI: 10.3389/fpsyt.2018.00106] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/16/2018] [Indexed: 12/12/2022] Open
Abstract
Intrauterine or perinatal complications constitute a major risk for psychiatric diseases. Infants who suffered from hypoxia-ischemia (HI) are at twofold risk to develop schizophrenia in later life. Several animal models attempt to reproduce these complications to study the yet unknown steps between an insult in early life and outbreak of the disease decades later. However, it is very challenging to find the right type and severity of insult leading to a disease-like phenotype in the animal, but not causing necrosis and focal neurological deficits. By contrast, too mild, repetitive insults may even be protective via conditioning effects. Thus, it is not surprising that animal models of hypoxia lead to mixed results. To achieve clinically translatable findings, better protocols are urgently needed. Therefore, we compare widely used models of hypoxia and HI and propose future directions for the field.
Collapse
Affiliation(s)
- Dimitri Hefter
- RG Animal Models in Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Heidelberg, Germany.,RG Neuro- and Sensory Physiology, Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Hugo H Marti
- RG Neurovascular Research, Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Peter Gass
- RG Animal Models in Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Heidelberg, Germany
| | - Dragos Inta
- RG Animal Models in Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Heidelberg, Germany.,Department of Psychiatry, University of Basel, Basel, Switzerland
| |
Collapse
|
55
|
Murray RM, Bhavsar V, Tripoli G, Howes O. 30 Years on: How the Neurodevelopmental Hypothesis of Schizophrenia Morphed Into the Developmental Risk Factor Model of Psychosis. Schizophr Bull 2017; 43:1190-1196. [PMID: 28981842 PMCID: PMC5737804 DOI: 10.1093/schbul/sbx121] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
At its re-birth 30 years ago, the neurodevelopment hypothesis of schizophrenia focussed on aberrant genes and early neural hazards, but then it grew to include ideas concerning aberrant synaptic pruning in adolescence. The hypothesis had its own stormy development and it endured some difficult teenage years when a resurgence of interest in neurodegeneration threatened its survival. In early adult life, it over-reached itself with some reductionists claiming that schizophrenia was simply a neurodevelopmental disease. However, by age 30, the hypothesis has matured sufficiently to incorporated childhood and adult adversity, urban living and migration, as well as heavy cannabis use, as important risk factors. Thus, it morphed into the developmental risk factor model of psychosis and integrated new evidence concerning dysregulated striatal dopamine as the final step on the pathway linking risk factors to psychotic symptoms.
Collapse
Affiliation(s)
- Robin M Murray
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King’s College, London, UK,National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King’s College, London, UK,To whom correspondence should be addressed;
| | - Vishal Bhavsar
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King’s College, London, UK
| | - Giada Tripoli
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King’s College, London, UK
| | - Oliver Howes
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King’s College, London, UK,Psychiatric Imaging Group, Clinical Science Centre, Imperial College, London, UK
| |
Collapse
|
56
|
Nagano R, Nagano M, Nakai A, Takeshita T, Suzuki H. Differential effects of neonatal SSRI treatments on hypoxia-induced behavioral changes in male and female offspring. Neuroscience 2017; 360:95-105. [PMID: 28778701 DOI: 10.1016/j.neuroscience.2017.07.051] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 07/19/2017] [Accepted: 07/19/2017] [Indexed: 02/07/2023]
Abstract
Prenatal hypoxia induced by transient intrauterine ischemia is a serious clinical problem, and at present, effective treatments are lacking. Currently, it is unknown how prenatal hypoxia affects behaviors in adulthood. Therefore, we developed a mouse model that mimics prenatal hypoxia in humans using uterine artery occlusion in late gestation. We examined whether prenatal hypoxia induces behavioral changes in adult male and female offspring by conducting a series of behavioral tests. In adulthood, longer immobility was observed in the forced swim test in males, whereas females showed decreased inhibition in the prepulse inhibition test. We then investigated the effects of two different selective serotonin reuptake inhibitors (SSRIs), fluoxetine (FLX) and escitalopram (ESC), on these behavioral changes. These drugs affect the neurodevelopmental process and have long-term neurological consequences. FLX treatment from postnatal day 3 (P3) to P21 ameliorated the behavioral changes in both male and female mice. In comparison, ESC treatment ameliorated the behavioral changes only in female mice. Neurochemical analysis revealed that dopamine was increased in the female hippocampus, but not in males. Thus, neonatal SSRI treatment decreases dopamine levels in the hippocampus in females selectively. Our findings suggest that prenatal hypoxia is a risk factor for behavioral abnormalities in adulthood, and that neonatal SSRI treatment might have clinical potential for alleviating these long-term behavioral deficits.
Collapse
Affiliation(s)
- Reiko Nagano
- Department of Obstetrics and Gynecology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan.
| | - Masatoshi Nagano
- Department of Pharmacology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan.
| | - Akihito Nakai
- Department of Obstetrics and Gynecology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan.
| | - Toshiyuki Takeshita
- Department of Obstetrics and Gynecology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan.
| | - Hidenori Suzuki
- Department of Pharmacology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan.
| |
Collapse
|
57
|
Katsel P, Roussos P, Pletnikov M, Haroutunian V. Microvascular anomaly conditions in psychiatric disease. Schizophrenia - angiogenesis connection. Neurosci Biobehav Rev 2017; 77:327-339. [PMID: 28396239 PMCID: PMC5497758 DOI: 10.1016/j.neubiorev.2017.04.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 12/31/2022]
Abstract
Schizophrenia (SZ) is a severe mental disorder with unknown etiology and elusive neuropathological and neurobiological features have been a focus of many theoretical hypotheses and empirical studies. Current genetic and neurobiology information relevant to SZ implicates neuronal developmental and synaptic plasticity abnormalities, and neurotransmitter, microglial and oligodendrocytes dysfunction. Several recent theories have highlighted the neurovascular unit as a potential contributor to the pathophysiology of SZ. We explored the biological plausibility of a link between SZ and the neurovascular system by examining insights gained from genetic, neuroimaging and postmortem studies, which include gene expression and neuropathology analyses. We also reviewed information from animal models of cerebral angiogenesis in order to understand better the complex interplay between angiogenic and neurotrophic factors in development, vascular endothelium/blood brain barrier remodeling and maintenance, all of which contribute to sustaining adequate regional blood flow and safeguarding normal brain function. Microvascular and hemodynamic alterations in SZ highlight the importance of further research and reveal the neurovascular unit as a potential therapeutic target in SZ.
Collapse
Affiliation(s)
- Pavel Katsel
- Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Panos Roussos
- Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, USA; Mental Illness Research, Education and Clinical Center (MIRECC), James J Peters VA Medical Center, Bronx, NY, USA
| | - Mikhail Pletnikov
- Departments of Psychiatry, Neuroscience, Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vahram Haroutunian
- Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Neuroscience, The Icahn School of Medicine at Mount Sinai, New York, NY, USA; Mental Illness Research, Education and Clinical Center (MIRECC), James J Peters VA Medical Center, Bronx, NY, USA
| |
Collapse
|
58
|
Lizano PL, Yao JK, Tandon N, Mothi SS, Montrose DM, Keshavan MS. Association of sFlt-1 and worsening psychopathology in relatives at high risk for psychosis: A longitudinal study. Schizophr Res 2017; 183:75-81. [PMID: 27863935 PMCID: PMC5432401 DOI: 10.1016/j.schres.2016.11.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/07/2016] [Accepted: 11/11/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND Angiogenic dysfunction and abnormalities in psychopathology and brain structure have been reported in schizophrenia, but their relationships are mostly unknown. We recently demonstrated that sFlt-1, anti-angiogenic factor, was significantly elevated in patients at familial high-risk for psychosis (FHR). We hypothesized that elevated sFlt-1 correlates with baseline and longitudinal changes in psychopathology, cognition, and brain structure. METHODS Plasma sFlt-1 in FHR (n=35) and HC (n=39) was obtained at baseline. Schizotypal, cognitive, soft neurologic signs, and structural brain imaging (1.5T T1-weighted MRI, FreeSurfer software) measures were obtained in both groups. Longitudinal clinical and brain structural measures were obtained in a subgroup of FHR patients. Baseline data analysis used correlations between sFlt-1 and clinical/imaging measures and adjusted for multiple corrections. Linear mixed-effects models described differences in trajectories between high sFlt-1 and low sFlt-1. RESULTS Baseline sFlt-1 was significantly correlated with soft neurologic signs (r=0.27, p=0.02) and right entorhinal volume (r=0.50, p=0.02), but not other baseline clinical/brain structural measures. Longitudinal examination of the FHR group (sFlt-1 high, n=14; sFlt-1 low, n=14) demonstrated that high sFlt-1 was significantly associated with worsening schizotypal symptoms (t=2.4, p=0.018). Reduced right hippocampal/parahippocampal volume/thickness trajectories were observed in high versus low sFlt-1 groups. CONCLUSIONS The findings from this FHR study demonstrate that peripheral markers of angiogenic dysfunction can predict longitudinal clinical and brain structural changes. Also, these findings further support the hypothesis of altered microvascular circulation in schizophrenia and those at risk.
Collapse
Affiliation(s)
- Paulo L Lizano
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, United States; Division of Public Psychiatry, Massachusetts Mental Health Center, Boston, MA, United States
| | - Jeffrey K Yao
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; VA Pittsburgh Healthcare System, Medical Research Service, Pittsburgh, PA, United States; Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, United States.
| | - Neeraj Tandon
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, United States; Baylor College of Medicine, Houston, TX, United States
| | - Suraj Sarvode Mothi
- Division of Public Psychiatry, Massachusetts Mental Health Center, Boston, MA, United States
| | - Debra M Montrose
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Matcheri S Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, United States; Division of Public Psychiatry, Massachusetts Mental Health Center, Boston, MA, United States; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Psychiatry, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
59
|
Potentially important periods of change in the development of social and role functioning in youth at clinical high risk for psychosis. Dev Psychopathol 2017; 30:39-47. [PMID: 28420458 DOI: 10.1017/s0954579417000451] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The developmental course of daily functioning prior to first psychosis-onset remains poorly understood. This study explored age-related periods of change in social and role functioning. The longitudinal study included youth (aged 12-23, mean follow-up years = 1.19) at clinical high risk (CHR) for psychosis (converters [CHR-C], n = 83; nonconverters [CHR-NC], n = 275) and a healthy control group (n = 164). Mixed-model analyses were performed to determine age-related differences in social and role functioning. We limited our analyses to functioning before psychosis conversion; thus, data of CHR-C participants gathered after psychosis onset were excluded. In controls, social and role functioning improved over time. From at least age 12, functioning in CHR was poorer than in controls, and this lag persisted over time. Between ages 15 and 18, social functioning in CHR-C stagnated and diverged from that of CHR-NC, who continued to improve (p = .001). Subsequently, CHR-C lagged behind in improvement between ages 21 and 23, further distinguishing them from CHR-NC (p < .001). A similar period of stagnation was apparent for role functioning, but to a lesser extent (p = .007). The results remained consistent when we accounted for the time to conversion. Our findings suggest that CHR-C start lagging behind CHR-NC in social and role functioning in adolescence, followed by a period of further stagnation in adulthood.
Collapse
|
60
|
Van der Auwera S, Wittfeld K, Shumskaya E, Bralten J, Zwiers MP, Onnink AMH, Usberti N, Hertel J, Völzke H, Völker U, Hosten N, Franke B, Grabe HJ. Predicting brain structure in population-based samples with biologically informed genetic scores for schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2017; 174:324-332. [PMID: 28304149 DOI: 10.1002/ajmg.b.32519] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 12/01/2016] [Indexed: 01/08/2023]
Abstract
Schizophrenia is associated with brain structural abnormalities including gray and white matter volume reductions. Whether these alterations are caused by genetic risk variants for schizophrenia is unclear. Previous attempts to detect associations between polygenic factors for schizophrenia and structural brain phenotypes in healthy subjects have been negative or remain non-replicated. In this study, we used genetic risk scores that were based on the accumulated effect of selected risk variants for schizophrenia belonging to specific biological systems like synaptic function, neurodevelopment, calcium signaling, and glutamatergic neurotransmission. We hypothesized that this "biologically informed" approach would provide the missing link between genetic risk for schizophrenia and brain structural phenotypes. We applied whole-brain voxel-based morphometry (VBM) analyses in two population-based target samples and subsequent regions of interest (ROIs) analyses in an independent replication sample (total N = 2725). No consistent association between the genetic scores and brain volumes were observed in the investigated samples. These results suggest that in healthy subjects with a higher genetic risk for schizophrenia additional factors apart from common genetic variants (e.g., infection, trauma, rare genetic variants, or gene-gene interactions) are required to induce structural abnormalities of the brain. Further studies are recommended to test for possible gene-gene or gene-environment effects. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sandra Van der Auwera
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany.,German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Germany
| | - Katharina Wittfeld
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Germany
| | - Elena Shumskaya
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands.,Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Janita Bralten
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcel P Zwiers
- Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - A Marten H Onnink
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Niccolo Usberti
- Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Johannes Hertel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany.,German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany.,DZHK-German Centre for Cardiovascular Research, Partner Site Greifswald, Greifswald, Germany.,DZD-German Centre for Diabetes Research, Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and, Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Norbert Hosten
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Barbara Franke
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany.,German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Germany
| |
Collapse
|
61
|
Dieset I, Andreassen OA, Haukvik UK. Somatic Comorbidity in Schizophrenia: Some Possible Biological Mechanisms Across the Life Span. Schizophr Bull 2016; 42:1316-1319. [PMID: 27033328 PMCID: PMC5049521 DOI: 10.1093/schbul/sbw028] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Schizophrenia is associated with decreased life expectancy (15-25 y) compared to the general population, with comorbid somatic diseases and in particular cardiovascular diseases being a major cause. Life style and medication probably account for much of the increased mortality risk due to somatic diseases in schizophrenia, but the evidence implicating biological pathways potentially affecting both body and brain is increasing. This includes overlapping genes between schizophrenia and somatic diseases, prenatal risk factors such as hypoxia and infections, and increased cardiovascular disease risk in drug-naïve patients at illness onset. Although environmental bias increases throughout the disease course, there are also some studies on chronic schizophrenia and postmortem brain samples that warrant further attention. In the following, we will attempt to move beyond environmental impact and explore some of the shared pathophysiological mechanisms potentially underlying both schizophrenia and somatic diseases.
Collapse
Affiliation(s)
- Ingrid Dieset
- NORMENT K.G. Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; NORMENT and K.G. Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway;
| | - Ole A Andreassen
- NORMENT K.G. Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; NORMENT and K.G. Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Unn K Haukvik
- Department of Adult Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatry, Ostfold Hospital Trust, Fredrikstad, Norway
| |
Collapse
|
62
|
Faa G, Manchia M, Pintus R, Gerosa C, Marcialis MA, Fanos V. Fetal programming of neuropsychiatric disorders. ACTA ACUST UNITED AC 2016; 108:207-223. [DOI: 10.1002/bdrc.21139] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 09/28/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Gavino Faa
- Division of Pathology, Department of Surgery; University Hospital San Giovanni di Dio; Cagliari Italy
| | - Mirko Manchia
- Section of Psychiatry, Department of Public Health, Clinical and Molecular Medicine; University of Cagliari; Cagliari Italy
- Department of Pharmacology; Dalhousie University; Halifax Nova Scotia Canada
| | - Roberta Pintus
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section; AOU Cagliari and University of Cagliari; Cagliari Italy
| | - Clara Gerosa
- Division of Pathology, Department of Surgery; University Hospital San Giovanni di Dio; Cagliari Italy
| | - Maria Antonietta Marcialis
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section; AOU Cagliari and University of Cagliari; Cagliari Italy
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section; AOU Cagliari and University of Cagliari; Cagliari Italy
| |
Collapse
|
63
|
Can Psychological, Social and Demographical Factors Predict Clinical Characteristics Symptomatology of Bipolar Affective Disorder and Schizophrenia? Psychiatr Q 2016; 87:501-13. [PMID: 26646576 PMCID: PMC4945684 DOI: 10.1007/s11126-015-9405-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Schizophrenia (SCH) is a complex, psychiatric disorder affecting 1 % of population. Its clinical phenotype is heterogeneous with delusions, hallucinations, depression, disorganized behaviour and negative symptoms. Bipolar affective disorder (BD) refers to periodic changes in mood and activity from depression to mania. It affects 0.5-1.5 % of population. Two types of disorder (type I and type II) are distinguished by severity of mania episodes. In our analysis, we aimed to check if clinical and demographical characteristics of the sample are predictors of symptom dimensions occurrence in BD and SCH cases. We included total sample of 443 bipolar and 439 schizophrenia patients. Diagnosis was based on DSM-IV criteria using Structured Clinical Interview for DSM-IV. We applied regression models to analyse associations between clinical and demographical traits from OPCRIT and symptom dimensions. We used previously computed dimensions of schizophrenia and bipolar affective disorder as quantitative traits for regression models. Male gender seemed protective factor for depression dimension in schizophrenia and bipolar disorder sample. Presence of definite psychosocial stressor prior disease seemed risk factor for depressive and suicidal domain in BD and SCH. OPCRIT items describing premorbid functioning seemed related with depression, positive and disorganised dimensions in schizophrenia and psychotic in BD. We proved clinical and demographical characteristics of the sample are predictors of symptom dimensions of schizophrenia and bipolar disorder. We also saw relation between clinical dimensions and course of disorder and impairment during disorder.
Collapse
|
64
|
Crider A, Pillai A. The Neurobiological Basis for Social Affiliation in Autism Spectrum Disorder and Schizophrenia. Curr Behav Neurosci Rep 2016; 3:154-164. [DOI: 10.1007/s40473-016-0079-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
65
|
Cassoli JS, Iwata K, Steiner J, Guest PC, Turck CW, Nascimento JM, Martins-de-Souza D. Effect of MK-801 and Clozapine on the Proteome of Cultured Human Oligodendrocytes. Front Cell Neurosci 2016; 10:52. [PMID: 26973466 PMCID: PMC4776125 DOI: 10.3389/fncel.2016.00052] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 02/15/2016] [Indexed: 01/06/2023] Open
Abstract
Separate lines of evidence have demonstrated the involvement of N-methyl-D-aspartate (NMDA) receptor and oligodendrocyte dysfunctions in schizophrenia. Here, we have carried out shotgun mass spectrometry proteome analysis of oligodendrocytes treated with the NMDA receptor antagonist MK-801 to gain potential insights into these effects at the molecular level. The MK-801 treatment led to alterations in the levels of 68 proteins, which are associated with seven distinct biological processes. Most of these proteins are involved in energy metabolism and many have been found to be dysregulated in previous proteomic studies of post-mortem brain tissues from schizophrenia patients. Finally, addition of the antipsychotic clozapine to MK-801-treated oligodendrocyte cultures resulted in changes in the levels of 45 proteins and treatment with clozapine alone altered 122 proteins and many of these showed opposite changes to the MK-801 effects. Therefore, these proteins and the associated energy metabolism pathways should be explored as potential biomarkers of antipsychotic efficacy. In conclusion, MK-801 treatment of oligodendrocytes may provide a useful model for testing the efficacy of novel treatment approaches.
Collapse
Affiliation(s)
- Juliana S Cassoli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas Campinas, Brazil
| | - Keiko Iwata
- United Graduate School of Child Development, Department of Development of Functional Brain Activities, Research Center for Child Mental Development, Hamamatsu University School of Medicine, Osaka University and Kanazawa University and Chiba University and University of Fukui Fukui, Japan
| | - Johann Steiner
- Department of Psychiatry, University of Magdeburg Magdeburg, Germany
| | - Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas Campinas, Brazil
| | - Christoph W Turck
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry Munich, Germany
| | - Juliana M Nascimento
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of CampinasCampinas, Brazil; D'Or Institute for Research and Education Rio de Janeiro, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of CampinasCampinas, Brazil; UNICAMP Neurobiology CenterCampinas, Brazil
| |
Collapse
|
66
|
Pre- and Perinatal Ischemia-Hypoxia, the Ischemia-Hypoxia Response Pathway, and ADHD Risk. Behav Genet 2016; 46:467-77. [DOI: 10.1007/s10519-016-9784-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 02/10/2016] [Indexed: 02/06/2023]
|
67
|
Ursini G, Cavalleri T, Fazio L, Angrisano T, Iacovelli L, Porcelli A, Maddalena G, Punzi G, Mancini M, Gelao B, Romano R, Masellis R, Calabrese F, Rampino A, Taurisano P, Di Giorgio A, Keller S, Tarantini L, Sinibaldi L, Quarto T, Popolizio T, Caforio G, Blasi G, Riva MA, De Blasi A, Chiariotti L, Bollati V, Bertolino A. BDNF rs6265 methylation and genotype interact on risk for schizophrenia. Epigenetics 2016; 11:11-23. [PMID: 26889735 DOI: 10.1080/15592294.2015.1117736] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Epigenetic mechanisms can mediate gene-environment interactions relevant for complex disorders. The BDNF gene is crucial for development and brain plasticity, is sensitive to environmental stressors, such as hypoxia, and harbors the functional SNP rs6265 (Val(66)Met), which creates or abolishes a CpG dinucleotide for DNA methylation. We found that methylation at the BDNF rs6265 Val allele in peripheral blood of healthy subjects is associated with hypoxia-related early life events (hOCs) and intermediate phenotypes for schizophrenia in a distinctive manner, depending on rs6265 genotype: in ValVal individuals increased methylation is associated with exposure to hOCs and impaired working memory (WM) accuracy, while the opposite is true for ValMet subjects. Also, rs6265 methylation and hOCs interact in modulating WM-related prefrontal activity, another intermediate phenotype for schizophrenia, with an analogous opposite direction in the 2 genotypes. Consistently, rs6265 methylation has a different association with schizophrenia risk in ValVals and ValMets. The relationships of methylation with BDNF levels and of genotype with BHLHB2 binding likely contribute to these opposite effects of methylation. We conclude that BDNF rs6265 methylation interacts with genotype to bridge early environmental exposures to adult phenotypes, relevant for schizophrenia. The study of epigenetic changes in regions containing genetic variation relevant for human diseases may have beneficial implications for the understanding of how genes are actually translated into phenotypes.
Collapse
Affiliation(s)
- Gianluca Ursini
- a Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro' , Bari , Italy.,b Lieber Institute for Brain Development, Johns Hopkins University Medical Campus , Baltimore , MD , US
| | - Tommaso Cavalleri
- c Department of Clinical and Community Sciences, Università degli Studi di Milano , Milan , Italy.,d Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Maggiore Hospital, Mangiagalli and Regina Elena Foundation , Milan , Italy
| | - Leonardo Fazio
- a Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro' , Bari , Italy
| | - Tiziana Angrisano
- e Istituto di Endocrinologia e Oncologia Sperimentale, IEOS, CNR, and Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II' , Naples
| | - Luisa Iacovelli
- f Department of Physiology and Pharmacology 'V. Erspamer', University of Rome 'Sapienza' , Rome , Italy
| | - Annamaria Porcelli
- a Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro' , Bari , Italy
| | - Giancarlo Maddalena
- a Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro' , Bari , Italy
| | - Giovanna Punzi
- a Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro' , Bari , Italy.,b Lieber Institute for Brain Development, Johns Hopkins University Medical Campus , Baltimore , MD , US
| | - Marina Mancini
- a Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro' , Bari , Italy
| | - Barbara Gelao
- a Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro' , Bari , Italy
| | - Raffaella Romano
- a Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro' , Bari , Italy
| | - Rita Masellis
- a Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro' , Bari , Italy
| | - Francesca Calabrese
- g Center of Neuropharmacology; Dipartimento di Scienze Farmacologiche e Biomolecolari; Università degli Studi di Milano , Milan , Italy
| | - Antonio Rampino
- a Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro' , Bari , Italy
| | - Paolo Taurisano
- a Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro' , Bari , Italy
| | - Annabella Di Giorgio
- h Department of Neuroradiology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) 'Casa Sollievo della Sofferenza' , San Giovanni Rotondo , Italy
| | - Simona Keller
- e Istituto di Endocrinologia e Oncologia Sperimentale, IEOS, CNR, and Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II' , Naples
| | - Letizia Tarantini
- c Department of Clinical and Community Sciences, Università degli Studi di Milano , Milan , Italy.,d Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Maggiore Hospital, Mangiagalli and Regina Elena Foundation , Milan , Italy
| | - Lorenzo Sinibaldi
- i Mendel Laboratory, Istituto di Ricoveroe Cura a Carattere Scientifico (IRCCS) 'Casa Sollievo della Sofferenza' , San Giovanni Rotondo , Italy
| | - Tiziana Quarto
- a Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro' , Bari , Italy.,j Cognitive Brain Research Unit, Institute of Behavioral Sciences, University of Helsinki , Helsinki , Finland
| | - Teresa Popolizio
- h Department of Neuroradiology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) 'Casa Sollievo della Sofferenza' , San Giovanni Rotondo , Italy
| | - Grazia Caforio
- a Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro' , Bari , Italy
| | - Giuseppe Blasi
- a Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro' , Bari , Italy
| | - Marco A Riva
- g Center of Neuropharmacology; Dipartimento di Scienze Farmacologiche e Biomolecolari; Università degli Studi di Milano , Milan , Italy
| | - Antonio De Blasi
- k Department of Molecular Medicine, University of Rome 'Sapienza' , Rome , Italy
| | - Lorenzo Chiariotti
- e Istituto di Endocrinologia e Oncologia Sperimentale, IEOS, CNR, and Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II' , Naples
| | - Valentina Bollati
- c Department of Clinical and Community Sciences, Università degli Studi di Milano , Milan , Italy.,d Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Maggiore Hospital, Mangiagalli and Regina Elena Foundation , Milan , Italy
| | - Alessandro Bertolino
- a Psychiatric Neuroscience Group, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari 'Aldo Moro' , Bari , Italy.,h Department of Neuroradiology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) 'Casa Sollievo della Sofferenza' , San Giovanni Rotondo , Italy
| |
Collapse
|
68
|
Ibi D, Yamada K. Therapeutic Targets for Neurodevelopmental Disorders Emerging from Animal Models with Perinatal Immune Activation. Int J Mol Sci 2015; 16:28218-29. [PMID: 26633355 PMCID: PMC4691039 DOI: 10.3390/ijms161226092] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 11/17/2015] [Accepted: 11/20/2015] [Indexed: 01/02/2023] Open
Abstract
Increasing epidemiological evidence indicates that perinatal infection with various viral pathogens enhances the risk for several psychiatric disorders. The pathophysiological significance of astrocyte interactions with neurons and/or gut microbiomes has been reported in neurodevelopmental disorders triggered by pre- and postnatal immune insults. Recent studies with the maternal immune activation or neonatal polyriboinosinic polyribocytidylic acid models of neurodevelopmental disorders have identified various candidate molecules that could be responsible for brain dysfunction. Here, we review the functions of several candidate molecules in neurodevelopment and brain function and discuss their potential as therapeutic targets for psychiatric disorders.
Collapse
Affiliation(s)
- Daisuke Ibi
- Department of Chemical Pharmacology, Faculty of Pharmaceutical Sciences, Meijo University, 150 Yagotoyama, Tenpaku-ku, Nagoya 468-8503, Japan.
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan.
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan.
| |
Collapse
|
69
|
Schmidt-Kastner R. Genomic approach to selective vulnerability of the hippocampus in brain ischemia–hypoxia. Neuroscience 2015; 309:259-79. [DOI: 10.1016/j.neuroscience.2015.08.034] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 08/12/2015] [Accepted: 08/17/2015] [Indexed: 01/06/2023]
|
70
|
Ayhan Y, McFarland R, Pletnikov MV. Animal models of gene-environment interaction in schizophrenia: A dimensional perspective. Prog Neurobiol 2015; 136:1-27. [PMID: 26510407 DOI: 10.1016/j.pneurobio.2015.10.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 09/07/2015] [Accepted: 10/22/2015] [Indexed: 12/12/2022]
Abstract
Schizophrenia has long been considered as a disorder with multifactorial origins. Recent discoveries have advanced our understanding of the genetic architecture of the disease. However, even with the increase of identified risk variants, heritability estimates suggest an important contribution of non-genetic factors. Various environmental risk factors have been proposed to play a role in the etiopathogenesis of schizophrenia. These include season of birth, maternal infections, obstetric complications, adverse events at early childhood, and drug abuse. Despite the progress in identification of genetic and environmental risk factors, we still have a limited understanding of the mechanisms whereby gene-environment interactions (G × E) operate in schizophrenia and psychoses at large. In this review we provide a critical analysis of current animal models of G × E relevant to psychotic disorders and propose that dimensional perspective will advance our understanding of the complex mechanisms of these disorders.
Collapse
Affiliation(s)
- Yavuz Ayhan
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA; Hacettepe University Faculty of Medicine, Department of Psychiatry, Turkey
| | - Ross McFarland
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, USA
| | - Mikhail V Pletnikov
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA; Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, USA; Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, USA.
| |
Collapse
|
71
|
Cassoli JS, Guest PC, Malchow B, Schmitt A, Falkai P, Martins-de-Souza D. Disturbed macro-connectivity in schizophrenia linked to oligodendrocyte dysfunction: from structural findings to molecules. NPJ SCHIZOPHRENIA 2015; 1:15034. [PMID: 27336040 PMCID: PMC4849457 DOI: 10.1038/npjschz.2015.34] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/10/2015] [Accepted: 08/19/2015] [Indexed: 01/20/2023]
Abstract
Schizophrenia is a severe psychiatric disorder with multi-factorial characteristics. A number of findings have shown disrupted synaptic connectivity in schizophrenia patients and emerging evidence suggests that this results from dysfunctional oligodendrocytes, the cells responsible for myelinating axons in white matter to promote neuronal conduction. The exact cause of this is not known, although recent imaging and molecular profiling studies of schizophrenia patients have identified changes in white matter tracts connecting multiple brain regions with effects on protein signaling networks involved in the myelination process. Further understanding of oligodendrocyte dysfunction in schizophrenia could lead to identification of novel drug targets for this devastating disease.
Collapse
Affiliation(s)
- Juliana Silva Cassoli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP) , Campinas, Brazil
| | - Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP) , Campinas, Brazil
| | - Berend Malchow
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University (LMU) , Munich, Germany
| | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University (LMU), Munich, Germany; Laboratory of Neurosciences (LIM-27), Institute of Psychiatry, University of São Paulo (USP), São Paulo, Brazil
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University (LMU) , Munich, Germany
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil; Laboratory of Neurosciences (LIM-27), Institute of Psychiatry, University of São Paulo (USP), São Paulo, Brazil; UNICAMP's Neurobiology Center, Campinas, Brazil
| |
Collapse
|
72
|
Moises HW, Wollschläger D, Binder H. Functional genomics indicate that schizophrenia may be an adult vascular-ischemic disorder. Transl Psychiatry 2015; 5:e616. [PMID: 26261884 PMCID: PMC4564558 DOI: 10.1038/tp.2015.103] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 05/26/2015] [Accepted: 06/14/2015] [Indexed: 01/17/2023] Open
Abstract
In search for the elusive schizophrenia pathway, candidate genes for the disorder from a discovery sample were localized within the energy-delivering and ischemia protection pathway. To test the adult vascular-ischemic (AVIH) and the competing neurodevelopmental hypothesis (NDH), functional genomic analyses of practically all available schizophrenia-associated genes from candidate gene, genome-wide association and postmortem expression studies were performed. Our results indicate a significant overrepresentation of genes involved in vascular function (P < 0.001), vasoregulation (that is, perivascular (P < 0.001) and shear stress (P < 0.01), cerebral ischemia (P < 0.001), neurodevelopment (P < 0.001) and postischemic repair (P < 0.001) among schizophrenia-associated genes from genetic association studies. These findings support both the NDH and the AVIH. The genes from postmortem studies showed an upregulation of vascular-ischemic genes (P = 0.020) combined with downregulated synaptic (P = 0.005) genes, and ND/repair (P = 0.003) genes. Evidence for the AVIH and the NDH is critically discussed. We conclude that schizophrenia is probably a mild adult vascular-ischemic and postischemic repair disorder. Adult postischemic repair involves ND genes for adult neurogenesis, synaptic plasticity, glutamate and increased long-term potentiation of excitatory neurotransmission (i-LTP). Schizophrenia might be caused by the cerebral analog of microvascular angina.
Collapse
Affiliation(s)
- H W Moises
- Molecular Genetics Laboratory, Department of Psychiatry (Retired), Kiel University Hospital, Kiel, Germany,Computational Genomics Lab, Frankfurt, Germany,Computational Genomics Lab, Beethovenstrasse 5, 60325 Frankfurt, Germany. E-mail:
| | - D Wollschläger
- Division Biostatistics/Bioinformatics, Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - H Binder
- Division Biostatistics/Bioinformatics, Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
73
|
Docherty AR, Bigdeli TB, Edwards AC, Bacanu S, Lee D, Neale MC, Wormley BK, Walsh D, O'Neill FA, Riley BP, Kendler KS, Fanous AH. Genome-wide gene pathway analysis of psychotic illness symptom dimensions based on a new schizophrenia-specific model of the OPCRIT. Schizophr Res 2015; 164:181-6. [PMID: 25778617 PMCID: PMC4409533 DOI: 10.1016/j.schres.2015.02.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 02/16/2015] [Accepted: 02/22/2015] [Indexed: 12/30/2022]
Abstract
Empirically derived phenotypic measurements have the potential to enhance gene-finding efforts in schizophrenia. Previous research based on factor analyses of symptoms has typically included schizoaffective cases. Deriving factor loadings from analysis of only narrowly defined schizophrenia cases could yield more sensitive factor scores for gene pathway and gene ontology analyses. Using an Irish family sample, this study 1) factor analyzed clinician-rated Operational Criteria Checklist items in cases with schizophrenia only, 2) scored the full sample based on these factor loadings, and 3) implemented genome-wide association, gene-based, and gene-pathway analysis of these SCZ-based symptom factors (final N=507). Three factors emerged from the analysis of the schizophrenia cases: a manic, a depressive, and a positive symptom factor. In gene-based analyses of these factors, multiple genes had q<0.01. Of particular interest are findings for PTPRG and WBP1L, both of which were previously implicated by the Psychiatric Genomics Consortium study of SCZ; results from this study suggest that variants in these genes might also act as modifiers of SCZ symptoms. Gene pathway analyses of the first factor indicated over-representation of glutamatergic transmission, GABA-A receptor, and cyclic GMP pathways. Results suggest that these pathways may have differential influence on affective symptom presentation in schizophrenia.
Collapse
Affiliation(s)
- Anna R Docherty
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth, University School of Medicine, VA, USA.
| | - T Bernard Bigdeli
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth, University School of Medicine, VA, USA
| | - Alexis C Edwards
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth, University School of Medicine, VA, USA
| | - Silviu Bacanu
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth, University School of Medicine, VA, USA
| | - Donghyung Lee
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth, University School of Medicine, VA, USA
| | - Michael C Neale
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth, University School of Medicine, VA, USA
| | - Brandon K Wormley
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth, University School of Medicine, VA, USA
| | | | | | - Brien P Riley
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth, University School of Medicine, VA, USA
| | - Kenneth S Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth, University School of Medicine, VA, USA
| | - Ayman H Fanous
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth, University School of Medicine, VA, USA; Washington Veterans Affairs Healthcare System, Washington D.C. USA; Georgetown University School of Medicine, Washington D.C. USA
| |
Collapse
|
74
|
Meier MH, Gillespie NA, Hansell NK, Hewitt AW, Hickie IB, Lu Y, McGrath J, MacGregor S, Medland SE, Sun C, Wong TY, Wright MJ, Zhu G, Martin NG, Mackey DA. Retinal microvessels reflect familial vulnerability to psychotic symptoms: A comparison of twins discordant for psychotic symptoms and controls. Schizophr Res 2015; 164:47-52. [PMID: 25694186 PMCID: PMC4409503 DOI: 10.1016/j.schres.2015.01.045] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 01/26/2015] [Accepted: 01/29/2015] [Indexed: 11/29/2022]
Abstract
Mounting evidence suggests that individuals with schizophrenia have an underlying vulnerability to cardiovascular disease, and a recent study suggested that this vulnerability might be reflected in the retinal microvasculature. The purpose of this study was to test the hypothesis that the retinal microvessels reflect familial vulnerability to psychotic symptoms. Participants were 531 adolescent and young adult twins who took part in the Brisbane Longitudinal Twin Study and the Twins Eye Study in Tasmania. The twins had photographs taken of their retina when they were adolescents or young adults (M age=20.6 years), and retinal vessel diameter was assessed using computer software. The twins completed an assessment of psychosis symptoms approximately six years later. We compared retinal venular diameters of individuals with one or more symptoms of psychosis (n=45), their unaffected co-twins (n=24), and controls (n=462). Individuals with one or more symptoms of psychosis had wider venules (standardized mean=0.29) than controls (standardized mean=-0.04; p=.03), and unaffected co-twins had venular diameters that were intermediate (standardized mean=0.13) between the two groups, suggesting that wide venules may represent a proxy marker of familial vulnerability to psychosis symptoms. Consistent with previous work, there were no differences in arteriolar diameter between individuals with and without symptoms of psychosis. Findings suggest that wide retinal venules may serve as a proxy marker of familial liability to psychosis symptoms. The pathophysiological mechanisms linking psychosis and cardiovascular disease may be operative from early in life, possibly at the level of the microvasculature.
Collapse
Affiliation(s)
| | - Nathan A. Gillespie
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond,VA, USA,QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Alex W. Hewitt
- Centre for Eye Research Australia (CERA), University of Melbourne, Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia,Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Ian B. Hickie
- Brain and Mind Research Institute, University of Sydney, Sydney, Australia
| | - Yi Lu
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - John McGrath
- Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, Australia,Queensland Brain Institute, University of Queensland, St Lucia, Australia
| | | | | | - Cong Sun
- Environmental and Genetic Epidemiology Research, Murdoch Children's Research Institute, Parkville, Victoria, Australia,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Tien Y. Wong
- Singapore Eye Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,Office of Clinical Sciences, Duke-NUS Graduate Medical School, Singapore
| | | | - Gu Zhu
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - David A. Mackey
- Centre for Eye Research Australia (CERA), University of Melbourne, Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia,Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
75
|
Caudle WM. Vulnerability of synapses in the frontal cortex of mice developmentally exposed to an insecticide: Potential contribution to neuropsychiatric disease. ACTA ACUST UNITED AC 2015; 2. [PMID: 26052547 DOI: 10.14800/nt.514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Increasingly, exposure to various chemicals found in our environment has been found to be a significant contributor to the risk of developing neurological disease, such as Parkinson disease, autism spectrum disorder, as well as other deficits in thought and function. Exposure to these compounds during critical periods of neurodevelopment, encompassing exposures that occur in utero, during infancy, childhood, and adolescence, represents a time period of nervous system growth that is uniquely vulnerable to disruption by environmental chemicals. Indeed, a contemporary hypothesis suggests that the pathological cascade associated with many common neurological disorders has its origin in disturbances of normal neurodevelopment. Moreover, alterations to the ontogeny of the synapse and neurotransmitter signaling during neurodevelopment may be a premier pathological event that underlies neuropsychiatric and neurodegenerative disease. To interrogate the impact of exposure to a ubiquitous environmental chemical, the pesticide, endosulfan, on development of neurotransmitter circuits, we coupled in vitro and in vivo platforms to evaluate its effect on the formation of GABAergic, glutamatergic, and dopaminergic pathways in the frontal cortex. With this approach we found exposure of cortical neurons, in vitro, exhibited a marked reduction in the length of their neurite process as well as the number of synaptic connections. Further investigation using an in vivo model of developmental exposure identified significant alterations to pre and postsynaptic proteins involved in neurotransmitter handling and signaling in each of the neurotransmitter systems investigated. These findings suggest that exposure to endosulfan during vulnerable periods of neurodevelopment can alter the normal development and potential function of neurotransmission in the frontal cortex. Interestingly, the alterations identified in our study closely mimic the pathological markers associated with schizophrenia, which shows disturbances in synaptic proteins important for GABAergic, glutamatergic, and dopaminergic signaling in the frontal cortex. These findings provide important support for the impact of exposure to environmental chemicals during neurodevelopment and risk for neurological disease.
Collapse
Affiliation(s)
- W Michael Caudle
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia 30322-3090 ; Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, Georgia 30322-3090
| |
Collapse
|
76
|
Debnath M, Venkatasubramanian G, Berk M. Fetal programming of schizophrenia: select mechanisms. Neurosci Biobehav Rev 2015; 49:90-104. [PMID: 25496904 PMCID: PMC7112550 DOI: 10.1016/j.neubiorev.2014.12.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 11/24/2014] [Accepted: 12/01/2014] [Indexed: 12/16/2022]
Abstract
Mounting evidence indicates that schizophrenia is associated with adverse intrauterine experiences. An adverse or suboptimal fetal environment can cause irreversible changes in brain that can subsequently exert long-lasting effects through resetting a diverse array of biological systems including endocrine, immune and nervous. It is evident from animal and imaging studies that subtle variations in the intrauterine environment can cause recognizable differences in brain structure and cognitive functions in the offspring. A wide variety of environmental factors may play a role in precipitating the emergent developmental dysregulation and the consequent evolution of psychiatric traits in early adulthood by inducing inflammatory, oxidative and nitrosative stress (IO&NS) pathways, mitochondrial dysfunction, apoptosis, and epigenetic dysregulation. However, the precise mechanisms behind such relationships and the specificity of the risk factors for schizophrenia remain exploratory. Considering the paucity of knowledge on fetal programming of schizophrenia, it is timely to consolidate the recent advances in the field and put forward an integrated overview of the mechanisms associated with fetal origin of schizophrenia.
Collapse
Affiliation(s)
- Monojit Debnath
- Department of Human Genetics, National Institute of Mental Health & Neurosciences, Bangalore 560029, India.
| | - Ganesan Venkatasubramanian
- Translational Psychiatry Laboratory, Neurobiology Research Centre and Department of Psychiatry, National Institute of Mental Health & Neurosciences, Hosur Road, Bangalore 560029, India
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Barwon Health, Geelong, Victoria, Australia; Department of Psychiatry, The Florey Institute of Neuroscience and Mental Health, and Orygen, The National Centre of Excellence in Youth Mental Health, University of Melbourne, Parkville, Australia
| |
Collapse
|
77
|
Schmidt MJ, Mirnics K. Neurodevelopment, GABA system dysfunction, and schizophrenia. Neuropsychopharmacology 2015; 40:190-206. [PMID: 24759129 PMCID: PMC4262918 DOI: 10.1038/npp.2014.95] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/03/2014] [Accepted: 04/11/2014] [Indexed: 02/07/2023]
Abstract
The origins of schizophrenia have eluded clinicians and researchers since Kraepelin and Bleuler began documenting their findings. However, large clinical research efforts in recent decades have identified numerous genetic and environmental risk factors for schizophrenia. The combined data strongly support the neurodevelopmental hypothesis of schizophrenia and underscore the importance of the common converging effects of diverse insults. In this review, we discuss the evidence that genetic and environmental risk factors that predispose to schizophrenia disrupt the development and normal functioning of the GABAergic system.
Collapse
Affiliation(s)
- Martin J Schmidt
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA
| | - Karoly Mirnics
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, USA
- Department of Psychiatry, University of Szeged, Szeged, Hungary
| |
Collapse
|
78
|
Smith TF, Anastopoulos AD, Garrett ME, Arias-Vasquez A, Franke B, Oades RD, Sonuga-Barke E, Asherson P, Gill M, Buitelaar JK, Sergeant JA, Kollins SH, Faraone SV, Ashley-Koch A. Angiogenic, neurotrophic, and inflammatory system SNPs moderate the association between birth weight and ADHD symptom severity. Am J Med Genet B Neuropsychiatr Genet 2014; 165B:691-704. [PMID: 25346392 DOI: 10.1002/ajmg.b.32275] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 09/25/2014] [Indexed: 12/31/2022]
Abstract
Low birth weight is associated with increased risk for Attention-Deficit/Hyperactivity Disorder (ADHD); however, the etiological underpinnings of this relationship remain unclear. This study investigated if genetic variants in angiogenic, dopaminergic, neurotrophic, kynurenine, and cytokine-related biological pathways moderate the relationship between birth weight and ADHD symptom severity. A total of 398 youth from two multi-site, family-based studies of ADHD were included in the analysis. The sample consisted of 360 ADHD probands, 21 affected siblings, and 17 unaffected siblings. A set of 164 SNPs from 31 candidate genes, representing five biological pathways, were included in our analyses. Birth weight and gestational age data were collected from a state birth registry, medical records, and parent report. Generalized Estimating Equations tested for main effects and interactions between individual SNPs and birth weight centile in predicting ADHD symptom severity. SNPs within neurotrophic (NTRK3) and cytokine genes (CNTFR) were associated with ADHD inattentive symptom severity. There was no main effect of birth weight centile on ADHD symptom severity. SNPs within angiogenic (NRP1 & NRP2), neurotrophic (NTRK1 & NTRK3), cytokine (IL16 & S100B), and kynurenine (CCBL1 & CCBL2) genes moderate the association between birth weight centile and ADHD symptom severity. The SNP main effects and SNP × birth weight centile interactions remained significant after adjusting for multiple testing. Genetic variability in angiogenic, neurotrophic, and inflammatory systems may moderate the association between restricted prenatal growth, a proxy for an adverse prenatal environment, and risk to develop ADHD.
Collapse
Affiliation(s)
- Taylor F Smith
- Department of Psychology and Child Development, California Polytechnic State University, San Luis Obispo, California; Department of Psychology, University of North Carolina at Greensboro, Greensboro, North Carolina
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Differential distribution of hypoxia-inducible factor 1-beta (ARNT or ARNT2) in mouse substantia nigra and ventral tegmental area. J Chem Neuroanat 2014; 61-62:64-71. [DOI: 10.1016/j.jchemneu.2014.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 06/03/2014] [Accepted: 07/02/2014] [Indexed: 01/27/2023]
|
80
|
Maternal immune activation and abnormal brain development across CNS disorders. Nat Rev Neurol 2014; 10:643-60. [PMID: 25311587 DOI: 10.1038/nrneurol.2014.187] [Citation(s) in RCA: 641] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Epidemiological studies have shown a clear association between maternal infection and schizophrenia or autism in the progeny. Animal models have revealed maternal immune activation (mIA) to be a profound risk factor for neurochemical and behavioural abnormalities in the offspring. Microglial priming has been proposed as a major consequence of mIA, and represents a critical link in a causal chain that leads to the wide spectrum of neuronal dysfunctions and behavioural phenotypes observed in the juvenile, adult or aged offspring. Such diversity of phenotypic outcomes in the mIA model are mirrored by recent clinical evidence suggesting that infectious exposure during pregnancy is also associated with epilepsy and, to a lesser extent, cerebral palsy in children. Preclinical research also suggests that mIA might precipitate the development of Alzheimer and Parkinson diseases. Here, we summarize and critically review the emerging evidence that mIA is a shared environmental risk factor across CNS disorders that varies as a function of interactions between genetic and additional environmental factors. We also review ongoing clinical trials targeting immune pathways affected by mIA that may play a part in disease manifestation. In addition, future directions and outstanding questions are discussed, including potential symptomatic, disease-modifying and preventive treatment strategies.
Collapse
|
81
|
Sohn H, Kim B, Kim KH, Kim MK, Choi TK, Lee SH. Effects of VRK2 (rs2312147) on white matter connectivity in patients with schizophrenia. PLoS One 2014; 9:e103519. [PMID: 25079070 PMCID: PMC4117506 DOI: 10.1371/journal.pone.0103519] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 07/02/2014] [Indexed: 12/04/2022] Open
Abstract
Recent genome-wide association studies of schizophrenia reported a novel risk variant, rs2312147 at vaccinia-related kinase 2 gene (VRK2), in multiple Asian and European samples. However, its effect on the brain structure in schizophrenia is little known. We analyzed the brain structure of 36 schizophrenia patients and 18 healthy subjects with regard to rs2312147 genotype groups. Brain magnetic resonance scans for gray matter (GM) and white matter (WM) analysis, and genotype analysis for VRK2 rs2312147, were conducted. The Positive and Negative Syndrome Scale and the Digit Symbol Test were assessed for schizophrenia patients. There was no significant difference in either GM volume or WM connectivity with regard to rs2312147 genotype in healthy subjects. In contrast, we found significant differences in the WM connectivity between rs2312147 CC and CT/TT genotype groups of schizophrenia patients. The related brain areas included the splenium of corpus callosum, the left occipital lobe WM, the internal capsule (left anterior limb and right retrolenticular part), the bilateral temporal lobe WM, the left fornix/stria terminalis, the left cingulate gyrus WM, and the left parietal lobe WM. Voxelwise correlation analysis revealed that the Digit Symbol Test scores (age corrected) correlated with the fractional anisotropy in WM tracts that previously showed significant group differences between the CT/TT and CC genotypes in the rs2312147 CT/TT genotype group, while no significant correlation was found in the CC genotype group. Our data may provide evidence for the effect of VRK2 on WM connectivity in patients with schizophrenia.
Collapse
Affiliation(s)
- Hoyoung Sohn
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Borah Kim
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Keun Hyang Kim
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Min-Kyoung Kim
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Tai Kiu Choi
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Sang-Hyuk Lee
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
- * E-mail:
| |
Collapse
|
82
|
Yamada S, Nagai T, Nakai T, Ibi D, Nakajima A, Yamada K. Matrix metalloproteinase-3 is a possible mediator of neurodevelopmental impairment due to polyI:C-induced innate immune activation of astrocytes. Brain Behav Immun 2014; 38:272-82. [PMID: 24594387 DOI: 10.1016/j.bbi.2014.02.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/13/2014] [Accepted: 02/24/2014] [Indexed: 01/01/2023] Open
Abstract
Increasing epidemiological evidence indicates that prenatal infection and childhood central nervous system infection with various viral pathogens enhance the risk for several neuropsychiatric disorders. Polyriboinosinic-polyribocytidilic acid (polyI:C) is known to induce strong innate immune responses that mimic immune activation by viral infections. Our previous findings suggested that activation of the innate immune system in astrocytes results in impairments of neurite outgrowth and spine formation, which lead to behavioral abnormalities in adulthood. To identify candidates of astrocyte-derived humoral factors that affect neuronal development, we analyzed astrocyte-conditioned medium (ACM) from murine astrocyte cultures treated with polyI:C (polyI:C-ACM) by two-dimensional fluorescence difference gel electrophoresis (2D-DIGE). Through a quantitative proteomic screen, we found that 13 protein spots were differentially expressed compared with ACM from vehicle-treated astrocytes (control-ACM), and characterized one of the candidates, matrix metalloproteinase-3 (Mmp3). PolyI:C treatment significantly increased the expression levels of Mmp3 mRNA and protein in astrocytes, but not microglia. PolyI:C-ACM was associated with significantly higher Mmp3 protein level and enzyme activity than control-ACM. The addition of recombinant Mmp3 into control-ACM impaired dendritic elongation of primary cultured hippocampal neurons, while the deleterious effect of polyI:C-ACM on neurite elongation was attenuated by knockdown of Mmp3 in astrocytes. These results suggest that Mmp3 is a possible mediator of polyI:C-ACM-induced neurodevelopmental impairment.
Collapse
Affiliation(s)
- Shinnosuke Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan
| | - Tsuyoshi Nakai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan
| | - Daisuke Ibi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan
| | - Akira Nakajima
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan.
| |
Collapse
|
83
|
Cheng J, Wang Y, Zhou K, Wang L, Li J, Zhuang Q, Xu X, Xu L, Zhang K, Dai D, Zheng R, Li G, Zhang A, Gao S, Duan S. Male-specific association between dopamine receptor D4 gene methylation and schizophrenia. PLoS One 2014; 9:e89128. [PMID: 24586542 PMCID: PMC3929639 DOI: 10.1371/journal.pone.0089128] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 01/20/2014] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE The goal of our study was to investigate whether DRD4 gene DNA methylation played an important role in the susceptibility of Han Chinese SCZ. METHODS Using the bisulphite pyrosequencing technology, DNA methylation levels of 6 CpG dinucleotides in DRD4 CpG island were measured among 30 paranoid SCZ patients, 30 undifferentiated SCZ patients, and 30 age- and gender-matched healthy controls. RESULTS Strong correlation was observed among the six CpG sites (r>0.5, P<0.01), thus average methylation levels were applied thereafter. Our results indicated that there was a significant association between DRD4 methylation and the risk of SCZ (P = 0.003), although there was no significant difference in DRD4 methylation between the two SCZ subtypes (P = 0.670). A breakdown analysis by gender showed that the significant association of DRD4 methylation and SCZ was driven by males (P<0.001) but not by females (P = 0.835). DRD4 methylation was significantly associated with p300 in male SCZ patients (r = -0.543, P = 0.005) but not in female SCZ patients (r = 0.110, P = 0.599). Moreover, receiver operating characteristic (ROC) curves showed DRD4 methylation was able to predict the status of SCZ in males [area under curve (AUC) = 0.832, P = 0.002] but not in females (AUC = 0.483, P = 0.876). Finally, a further expression experiment showed that DRD4 methylation in the gene body was positively associated with gene expression, although the exact mechanism of gene regulation remained unknown for this interesting DRD4 methylation. CONCLUSION The gender disparity in the DRD4 DNA methylation provides novel insights into the pathogenesis of SCZ.
Collapse
Affiliation(s)
- Jia Cheng
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang, China
| | - Yunliang Wang
- Neurology Department of the 148th Hospital of PLA, Zibo, Shandong, China
| | - Kena Zhou
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Lingyan Wang
- Bank of Blood Products, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Jinfeng Li
- Neurology Department of the 148th Hospital of PLA, Zibo, Shandong, China
| | - Qidong Zhuang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Xuting Xu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Leiting Xu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Kai Zhang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Dongjun Dai
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Rongjiong Zheng
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Guangxue Li
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang, China
| | - Aiping Zhang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Shugui Gao
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang, China
| | - Shiwei Duan
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
84
|
Schmitt A, Malchow B, Hasan A, Falkai P. The impact of environmental factors in severe psychiatric disorders. Front Neurosci 2014; 8:19. [PMID: 24574956 PMCID: PMC3920481 DOI: 10.3389/fnins.2014.00019] [Citation(s) in RCA: 203] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 01/23/2014] [Indexed: 01/08/2023] Open
Abstract
During the last decades, schizophrenia has been regarded as a developmental disorder. The neurodevelopmental hypothesis proposes schizophrenia to be related to genetic and environmental factors leading to abnormal brain development during the pre- or postnatal period. First disease symptoms appear in early adulthood during the synaptic pruning and myelination process. Meta-analyses of structural MRI studies revealing hippocampal volume deficits in first-episode patients and in the longitudinal disease course confirm this hypothesis. Apart from the influence of risk genes in severe psychiatric disorders, environmental factors may also impact brain development during the perinatal period. Several environmental factors such as antenatal maternal virus infections, obstetric complications entailing hypoxia as common factor or stress during neurodevelopment have been identified to play a role in schizophrenia and bipolar disorder, possibly contributing to smaller hippocampal volumes. In major depression, psychosocial stress during the perinatal period or in adulthood is an important trigger. In animal studies, chronic stress or repeated administration of glucocorticoids have been shown to induce degeneration of glucocorticoid-sensitive hippocampal neurons and may contribute to the pathophysiology of affective disorders. Epigenetic mechanisms altering the chromatin structure such as histone acetylation and DNA methylation may mediate effects of environmental factors to transcriptional regulation of specific genes and be a prominent factor in gene-environmental interaction. In animal models, gene-environmental interaction should be investigated more intensely to unravel pathophysiological mechanisms. These findings may lead to new therapeutic strategies influencing epigenetic targets in severe psychiatric disorders.
Collapse
Affiliation(s)
- Andrea Schmitt
- Department of Psychiatry and Psychotherapy, LMU Munich Munich, Germany ; Laboratory of Neuroscience (LIM27), Institute of Psychiatry, University of Sao Paulo São Paulo, Brazil
| | - Berend Malchow
- Department of Psychiatry and Psychotherapy, LMU Munich Munich, Germany
| | - Alkomiet Hasan
- Department of Psychiatry and Psychotherapy, LMU Munich Munich, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, LMU Munich Munich, Germany
| |
Collapse
|
85
|
Anti-brain autoantibodies in the serum of schizophrenic patients: a case-control study. Psychiatry Res 2013; 210:800-5. [PMID: 24103908 DOI: 10.1016/j.psychres.2013.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Revised: 06/04/2013] [Accepted: 09/08/2013] [Indexed: 11/23/2022]
Abstract
Schizophrenia is considered a neurodevelopmental disorder with a multifactorial pathogenesis where autoimmune factors may play a significant role. The aim of this study was to verify the presence of anti-brain autoantibodies in the serum of schizophrenic patients compared to healthy controls. Autoantibodies against brain were detected by the immunofluorescence method, utilizing sections of rat hippocampus and hypothalamus and of monkey cerebellum. Three different fluorescence patterns were observed, staining the nucleus-cytoplasm of neurons, the neuroendothelial of blood vessel and the neurofilaments. Search for other organ-specific and non organ-specific autoantibodies was performed in all sera by indirect immunofluorescence method, enzyme linked immunosorbent assay and chemiluminescence immunoassay. Results showed a significant association between schizophrenia and anti-brain autoantibodies against the neuroendothelium of blood vessel in hypothalamus, hippocampus and cerebellum; a significant nuclear and cytoplasmic staining of neurons was assessed only for the hippocampus. No other significant association was found, except between schizophrenia and anti-nuclear autoantibodies on HEp-2 cells. In conclusion, these results support the hypothesis of a significant association between schizophrenia and circulating anti-brain autoantibodies, suggesting a diffuse reactivity against the neuroendothelium of blood vessel and highlighting a nuclear and cytoplasmic staining of the neurons of hippocampus.
Collapse
|
86
|
Meier MH, Shalev I, Moffitt TE, Kapur S, Keefe RSE, Wong TY, Belsky DW, Harrington H, Hogan S, Houts R, Caspi A, Poulton R. Microvascular abnormality in schizophrenia as shown by retinal imaging. Am J Psychiatry 2013; 170:1451-9. [PMID: 24030514 PMCID: PMC3857729 DOI: 10.1176/appi.ajp.2013.13020234] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Retinal and cerebral microvessels are structurally and functionally homologous, but unlike cerebral microvessels, retinal microvessels can be noninvasively measured in vivo by retinal imaging. The authors tested the hypothesis that individuals with schizophrenia exhibit microvascular abnormality and evaluated the utility of retinal imaging as a tool for schizophrenia research. METHOD Participants were members of the Dunedin Study, a population-representative cohort followed from birth with 95% retention. Study members underwent retinal imaging at age 38. The authors assessed retinal arteriolar and venular caliber for all members of the cohort, including individuals who developed schizophrenia. RESULTS Study members who developed schizophrenia were distinguished by wider retinal venules, suggesting microvascular abnormality reflective of insufficient brain oxygen supply. Analyses that controlled for confounding health conditions suggested that wider retinal venules are not simply an artifact of co-occurring health problems in schizophrenia patients. Wider venules were also associated with a dimensional measure of adult psychosis symptoms and with psychosis symptoms reported in childhood. CONCLUSIONS The findings provide initial support for the hypothesis that individuals with schizophrenia show microvascular abnormality. Moreover, the results suggest that the same vascular mechanisms underlie subthreshold symptoms and clinical disorder and that these associations may begin early in life. These findings highlight the promise of retinal imaging as a tool for understanding the pathogenesis of schizophrenia.
Collapse
|
87
|
O-GlcNAc transferase (OGT) as a placental biomarker of maternal stress and reprogramming of CNS gene transcription in development. Proc Natl Acad Sci U S A 2013; 110:5169-74. [PMID: 23487789 DOI: 10.1073/pnas.1300065110] [Citation(s) in RCA: 189] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Maternal stress is a key risk factor for neurodevelopmental disorders, including schizophrenia and autism, which often exhibit a sex bias in rates of presentation, age of onset, and symptom severity. The placenta is an endocrine tissue that functions as an important mediator in responding to perturbations in the intrauterine environment and is accessible for diagnostic purposes, potentially providing biomarkers predictive of disease. Therefore, we have used a genome-wide array approach to screen placental expression across pregnancy for gene candidates that are sex-biased and stress-responsive in mice and translate to human tissue. We identifed O-linked-N-acetylglucosamine (O-GlcNAc) transferase (OGT), an X-linked gene important in regulating proteins involved in chromatin remodeling, as fitting these criteria. Levels of both OGT and its biochemical mark, O-GlcNAcylation, were significantly lower in males and further reduced by prenatal stress. Examination of human placental tissue found similar patterns related to X chromosome dosage. As a demonstration of the importance of placental OGT in neurodevelopment, we found that hypothalamic gene expression and the broad epigenetic microRNA environment in the neonatal brain of placental-specific hemizygous OGT mice was substantially altered. These studies identified OGT as a promising placental biomarker of maternal stress exposure that may relate to sex-biased outcomes in neurodevelopment.
Collapse
|
88
|
Patel CJ, Chen R, Butte AJ. Data-driven integration of epidemiological and toxicological data to select candidate interacting genes and environmental factors in association with disease. Bioinformatics 2013; 28:i121-6. [PMID: 22689751 PMCID: PMC3371861 DOI: 10.1093/bioinformatics/bts229] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Motivation: Complex diseases, such as Type 2 Diabetes Mellitus (T2D), result from the interplay of both environmental and genetic factors. However, most studies investigate either the genetics or the environment and there are a few that study their possible interaction in context of disease. One key challenge in documenting interactions between genes and environment includes choosing which of each to test jointly. Here, we attempt to address this challenge through a data-driven integration of epidemiological and toxicological studies. Specifically, we derive lists of candidate interacting genetic and environmental factors by integrating findings from genome-wide and environment-wide association studies. Next, we search for evidence of toxicological relationships between these genetic and environmental factors that may have an etiological role in the disease. We illustrate our method by selecting candidate interacting factors for T2D. Contact:abutte@stanford.edu
Collapse
Affiliation(s)
- Chirag J Patel
- Division of Systems Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | |
Collapse
|
89
|
Obstetric complications in early psychosis: relation with family history of psychosis. Psychiatry Res 2012; 200:708-14. [PMID: 22868179 DOI: 10.1016/j.psychres.2012.07.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2011] [Revised: 07/12/2012] [Accepted: 07/15/2012] [Indexed: 10/28/2022]
Abstract
The people classified as being at ultra-high risk (UHR) of developing psychosis are expected to share many risk factors for psychosis with the patients diagnosed with schizophrenia, including an enhanced incidence of obstetric complications (OCs). This study set out to investigate the incidence and correlates of OCs in a sample of patients accessing an early intervention center. Patients' mothers were asked whether they had suffered from any somatic complication during pregnancy from a list of OCs with potential direct relevance to the physical wellbeing of the offspring. Out of 86 patients diagnosed with first-episode psychosis, 20 (23%) cases were positive for the occurrence of severe OCs, as reported by their mothers during an interview; out of 83 UHR patients, 21 (25%) cases were positive for OCs. OCs were more common in individuals with a family history of psychosis than in those without such a history. OCs might interact with genetic vulnerability to increase the risk of psychosis. Lack of comparison to healthy controls is a limitation that decreases the value of these findings.
Collapse
|
90
|
Hypoxic induction of the regulator of G-protein signalling 4 gene is mediated by the hypoxia-inducible factor pathway. PLoS One 2012; 7:e44564. [PMID: 22970249 PMCID: PMC3436875 DOI: 10.1371/journal.pone.0044564] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 08/08/2012] [Indexed: 11/19/2022] Open
Abstract
The transcriptional response to hypoxia is largely dependent on the Hypoxia Inducible Factors (HIF-1 and HIF-2) in mammalian cells. Many target genes have been characterised for these heterodimeric transcription factors, yet there is evidence that the full range of HIF-regulated genes has not yet been described. We constructed a TetON overexpression system in the rat pheochromocytoma PC-12 cell line to search for novel HIF and hypoxia responsive genes. The Rgs4 gene encodes the Regulator of G-Protein Signalling 4 (RGS4) protein, an inhibitor of signalling from G-protein coupled receptors, and dysregulation of Rgs4 is linked to disease states such as schizophrenia and cardiomyopathy. Rgs4 was found to be responsive to HIF-2α overexpression, hypoxic treatment, and hypoxia mimetic drugs in PC-12 cells. Similar responses were observed in human neuroblastoma cell lines SK-N-SH and SK-N-BE(2)C, but not in endothelial cells, where Rgs4 transcript is readily detected but does not respond to hypoxia. Furthermore, this regulation was found to be dependent on transcription, and occurs in a manner consistent with direct HIF transactivation of Rgs4 transcription. However, no HIF binding site was detectable within 32 kb of the human Rgs4 gene locus, leading to the possibility of regulation by long-distance genomic interactions. Further research into Rgs4 regulation by hypoxia and HIF may result in better understanding of disease states such as schizophrenia, and also shed light on the other roles of HIF yet to be discovered.
Collapse
|
91
|
Stolp H, Neuhaus A, Sundramoorthi R, Molnár Z. The Long and the Short of it: Gene and Environment Interactions During Early Cortical Development and Consequences for Long-Term Neurological Disease. Front Psychiatry 2012; 3:50. [PMID: 22701439 PMCID: PMC3372875 DOI: 10.3389/fpsyt.2012.00050] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 05/01/2012] [Indexed: 01/21/2023] Open
Abstract
Cortical development is a complex amalgamation of proliferation, migration, differentiation, and circuit formation. These processes follow defined timescales and are controlled by a combination of intrinsic and extrinsic factors. It is currently unclear how robust and flexible these processes are and whether the developing brain has the capacity to recover from disruptions. What is clear is that there are a number of cognitive disorders or conditions that are elicited as a result of disrupted cortical development, although it may take a long time for the full pathophysiology of the conditions to be realized clinically. The critical window for the manifestation of a neurodevelopmental disorder is prolonged, and there is the potential for a complex interplay between genes and environment. While there have been extended investigations into the genetic basis of a number of neurological and mental disorders, limited definitive associations have been discovered. Many environmental factors, including inflammation and stress, have been linked to neurodevelopmental disorders, and it may be that a better understanding of the interplay between genes and environment will speed progress in this field. In particular, the development of the brain needs to be considered in the context of the whole materno-fetal unit as the degree of the metabolic, endocrine, or inflammatory responses, for example, will greatly influence the environment in which the brain develops. This review will emphasize the importance of extending neurodevelopmental studies to the contribution of the placenta, vasculature, cerebrospinal fluid, and to maternal and fetal immune response. These combined investigations are more likely to reveal genetic and environmental factors that influence the different stages of neuronal development and potentially lead to the better understanding of the etiology of neurological and mental disorders such as autism, epilepsy, cerebral palsy, and schizophrenia.
Collapse
Affiliation(s)
- Helen Stolp
- Department of Physiology, Anatomy and Genetics, University of Oxford Oxford, UK
| | | | | | | |
Collapse
|