401
|
Itahashi T, Noda Y, Iwata Y, Tarumi R, Tsugawa S, Plitman E, Honda S, Caravaggio F, Kim J, Matsushita K, Gerretsen P, Uchida H, Remington G, Mimura M, Aoki YY, Graff-Guerrero A, Nakajima S. Dimensional distribution of cortical abnormality across antipsychotics treatment-resistant and responsive schizophrenia. NEUROIMAGE-CLINICAL 2021; 32:102852. [PMID: 34638035 PMCID: PMC8527893 DOI: 10.1016/j.nicl.2021.102852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/13/2021] [Accepted: 10/04/2021] [Indexed: 11/30/2022]
Abstract
Different etiology is assumed in treatment-resistant
and responsive schizophrenia. Patients with treatment-resistant schizophrenia were
classified from controls. Patients with non-treatment-resistant schizophrenia
were classified from controls. Two classifications reached area under the curve as
high as 0.69 and 0.85. Area under the curve remained as high as 0.69 when
two classifiers were swapped.
Background One-third of patients with schizophrenia are
treatment-resistant to non-clozapine antipsychotics (TRS), while the rest
respond (NTRS). Examining whether TRS and NTRS represent different
pathophysiologies is an important step toward precision
medicine. Methods Focusing on cortical thickness (CT), we analyzed
international multi-site cross-sectional datasets of magnetic resonance imaging
comprising 110 patients with schizophrenia (NTRS = 46, TRS = 64) and 52 healthy
controls (HCs). We utilized a logistic regression with L1-norm regularization to
find brain regions related to either NTRS or TRS. We conducted nested 10-fold
cross-validation and computed the accuracy and area under the curve (AUC). Then,
we applied the NTRS classifier to patients with TRS, and vice
versa. Results Patients with NTRS and TRS were classified from HCs with
65% and 78% accuracies and with the AUC of 0.69 and 0.85
(p = 0.014 and < 0.001, corrected), respectively.
The left planum temporale (PT) and left anterior insula/inferior frontal gyrus
(IFG) contributed to both NTRS and TRS classifiers. The left supramarginal gyrus
only contributed to NTRS and right superior temporal sulcus and right lateral
orbitofrontal cortex only to the TRS. The NTRS classifiers successfully
distinguished those with TRS from HCs with the AUC of 0.78
(p < 0.001), while the TRS classifiers classified
those with NTRS from HCs with the AUC of 0.69
(p = 0.015). Conclusion Both NTRS and TRS could be distinguished from HCs on the
basis of CT. The CT pathological basis of NTRS and TRS has commonalities, and
TRS presents unique CT features.
Collapse
Affiliation(s)
- Takashi Itahashi
- Medical Institute of Developmental Disabilities Research, Showa University, Tokyo, Japan
| | - Yoshihiro Noda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Yusuke Iwata
- Department of Neuropsychiatry, University of Yamanashi Faculty of Medicine, Yamanashi, Japan
| | - Ryosuke Tarumi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Sakiko Tsugawa
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Eric Plitman
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Shiori Honda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Fernando Caravaggio
- Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Julia Kim
- Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Karin Matsushita
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Philip Gerretsen
- Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Hiroyuki Uchida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan; Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - Gary Remington
- Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Masaru Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Yuta Y Aoki
- Medical Institute of Developmental Disabilities Research, Showa University, Tokyo, Japan.
| | - Ariel Graff-Guerrero
- Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| | - Shinichiro Nakajima
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan; Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada.
| |
Collapse
|
402
|
Cortical surface abnormalities are different depending on the stage of schizophrenia: A cross-sectional vertexwise mega-analysis of thickness, area and gyrification. Schizophr Res 2021; 236:104-114. [PMID: 34481405 DOI: 10.1016/j.schres.2021.08.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 05/28/2021] [Accepted: 08/09/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Brain magnetic resonance imaging studies have not investigated the cortical surface comprehensively in schizophrenia subjects by assessing thickness, surface area and gyrification separately during the first-episode of psychosis (FEP) or chronic schizophrenia (ChSch). METHODS We investigated cortical surface abnormalities in 137 FEP patients and 240 ChSch subjects compared to 297 Healthy Controls (HC) contributed by five cohorts. Maps showing results of vertexwise between-group comparisons of cortical thickness, area, and gyrification were produced using T1-weighted datasets processed using FreeSurfer 5.3, followed by validated quality control protocols. RESULTS FEP subjects showed large clusters of increased area and gyrification relative to HC in prefrontal and insuli cortices (Cohen's d: 0.049 to 0.28). These between-group differences occurred partially beyond the effect of sample. ChSch subjects displayed reduced cortical thickness relative to HC in smaller fronto-temporal foci (d: -0.73 to -0.35), but not beyond the effect of sample. Differences between FEP and HC subjects were associated with male gender, younger age, and earlier illness onset, while differences between ChSch and HC were associated with treatment-resistance and first-generation antipsychotic (FGA) intake independently of sample effect. CONCLUSIONS Separate assessments of FEP and ChSch revealed abnormalities that differed in regional distribution, phenotypes affected and effect size. In FEP, associations of greater cortical area and gyrification abnormalities with earlier age of onset suggest an origin on anomalous neurodevelopment, while thickness reductions in ChSch are at least partially explained by treatment-resistance and FGA intake. Associations of between-group differences with clinical variables retained statistical significance beyond the effect of sample.
Collapse
|
403
|
Implications for Early Diagnosis and Treatment in Schizophrenia Due to Correlation between Auditory Perceptual Deficits and Cognitive Impairment. J Clin Med 2021; 10:jcm10194557. [PMID: 34640571 PMCID: PMC8509531 DOI: 10.3390/jcm10194557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/27/2021] [Indexed: 11/17/2022] Open
Abstract
It is indicated that auditory perception deficits are present in schizophrenia and related to formal thought disorder. The purpose of the present study was to investigate the association of auditory deficits with cognitive impairment in schizophrenia. An experimental group of 50 schizophrenia patients completed a battery of auditory processing evaluation and a neuropsychological battery of tests. Correlations between neuropsychological battery scores and auditory processing scores were examined. Cognitive impairment was correlated with auditory processing deficits in schizophrenia patients. All neuropsychological test scores were significantly correlated with at least one auditory processing test score. Our findings support the coexistence of auditory processing disorder, severe cognitive impairment, and formal thought disorder in a subgroup of schizophrenia patients. This may have important implications in schizophrenia research, as well as in early diagnosis and nonpharmacological treatment of the disorder.
Collapse
|
404
|
Mikolas P, Bröckel K, Vogelbacher C, Müller DK, Marxen M, Berndt C, Sauer C, Jung S, Fröhner JH, Fallgatter AJ, Ethofer T, Rau A, Kircher T, Falkenberg I, Lambert M, Kraft V, Leopold K, Bechdolf A, Reif A, Matura S, Stamm T, Bermpohl F, Fiebig J, Juckel G, Flasbeck V, Correll CU, Ritter P, Bauer M, Jansen A, Pfennig A. Individuals at increased risk for development of bipolar disorder display structural alterations similar to people with manifest disease. Transl Psychiatry 2021; 11:485. [PMID: 34545071 PMCID: PMC8452775 DOI: 10.1038/s41398-021-01598-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/06/2021] [Accepted: 08/25/2021] [Indexed: 02/08/2023] Open
Abstract
In psychiatry, there has been a growing focus on identifying at-risk populations. For schizophrenia, these efforts have led to the development of early recognition and intervention measures. Despite a similar disease burden, the populations at risk of bipolar disorder have not been sufficiently characterized. Within the BipoLife consortium, we used magnetic resonance imaging (MRI) data from a multicenter study to assess structural gray matter alterations in N = 263 help-seeking individuals from seven study sites. We defined the risk using the EPIbipolar assessment tool as no-risk, low-risk, and high-risk and used a region-of-interest approach (ROI) based on the results of two large-scale multicenter studies of bipolar disorder by the ENIGMA working group. We detected significant differences in the thickness of the left pars opercularis (Cohen's d = 0.47, p = 0.024) between groups. The cortex was significantly thinner in high-risk individuals compared to those in the no-risk group (p = 0.011). We detected no differences in the hippocampal volume. Exploratory analyses revealed no significant differences in other cortical or subcortical regions. The thinner cortex in help-seeking individuals at risk of bipolar disorder is in line with previous findings in patients with the established disorder and corresponds to the region of the highest effect size in the ENIGMA study of cortical alterations. Structural alterations in prefrontal cortex might be a trait marker of bipolar risk. This is the largest structural MRI study of help-seeking individuals at increased risk of bipolar disorder.
Collapse
Affiliation(s)
- Pavol Mikolas
- Department of Psychiatry and Psychotherapy, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany.
| | - Kyra Bröckel
- grid.412282.f0000 0001 1091 2917Department of Psychiatry and Psychotherapy, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Christoph Vogelbacher
- grid.10253.350000 0004 1936 9756Core-Facility Brainimaging, Faculty of Medicine, University of Marburg, Marburg, Germany ,grid.10253.350000 0004 1936 9756Department of Psychiatry, University of Marburg, Marburg, Germany ,grid.8664.c0000 0001 2165 8627Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| | - Dirk K. Müller
- grid.412282.f0000 0001 1091 2917Department of Psychiatry and Psychotherapy, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany ,grid.4488.00000 0001 2111 7257Neuroimaging Center, Technische Universität Dresden, Dresden, Germany ,grid.4488.00000 0001 2111 7257Institute for Medical Informatics and Biometry, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Michael Marxen
- grid.412282.f0000 0001 1091 2917Department of Psychiatry and Psychotherapy, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany ,grid.4488.00000 0001 2111 7257Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Christina Berndt
- grid.412282.f0000 0001 1091 2917Department of Psychiatry and Psychotherapy, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Cathrin Sauer
- grid.412282.f0000 0001 1091 2917Department of Psychiatry and Psychotherapy, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Stine Jung
- grid.412282.f0000 0001 1091 2917Department of Psychiatry and Psychotherapy, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Juliane Hilde Fröhner
- grid.412282.f0000 0001 1091 2917Department of Psychiatry and Psychotherapy, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany ,grid.4488.00000 0001 2111 7257Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Andreas J. Fallgatter
- grid.10392.390000 0001 2190 1447Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany
| | - Thomas Ethofer
- grid.10392.390000 0001 2190 1447Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany ,grid.10392.390000 0001 2190 1447Department for Biomedical Resonance, University of Tübingen, Tübingen, Germany
| | - Anne Rau
- grid.10392.390000 0001 2190 1447Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany
| | - Tilo Kircher
- grid.10253.350000 0004 1936 9756Core-Facility Brainimaging, Faculty of Medicine, University of Marburg, Marburg, Germany ,grid.10253.350000 0004 1936 9756Department of Psychiatry, University of Marburg, Marburg, Germany ,grid.8664.c0000 0001 2165 8627Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| | - Irina Falkenberg
- grid.10253.350000 0004 1936 9756Core-Facility Brainimaging, Faculty of Medicine, University of Marburg, Marburg, Germany ,grid.10253.350000 0004 1936 9756Department of Psychiatry, University of Marburg, Marburg, Germany ,grid.8664.c0000 0001 2165 8627Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| | - Martin Lambert
- grid.13648.380000 0001 2180 3484Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vivien Kraft
- grid.13648.380000 0001 2180 3484Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karolina Leopold
- grid.6363.00000 0001 2218 4662Department of Psychiatry, Psychotherapy and Psychosomatic Medicine, Vivantes Hospital Am Urban and Vivantes Hospital Im Friedrichshain, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Bechdolf
- grid.6363.00000 0001 2218 4662Department of Psychiatry, Psychotherapy and Psychosomatic Medicine, Vivantes Hospital Am Urban and Vivantes Hospital Im Friedrichshain, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Silke Matura
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Thomas Stamm
- grid.6363.00000 0001 2218 4662Department of Psychiatry and Neurosciences, Charité Campus Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany ,grid.473452.3Department of Clinical Psychiatry and Psychotherapy, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Felix Bermpohl
- grid.6363.00000 0001 2218 4662Department of Psychiatry and Neurosciences, Charité Campus Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jana Fiebig
- grid.6363.00000 0001 2218 4662Department of Psychiatry and Neurosciences, Charité Campus Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Georg Juckel
- grid.5570.70000 0004 0490 981XDepartment of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University, Bochum, Germany
| | - Vera Flasbeck
- grid.5570.70000 0004 0490 981XDepartment of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University, Bochum, Germany
| | - Christoph U. Correll
- grid.6363.00000 0001 2218 4662Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin Berlin, Berlin, Germany ,grid.440243.50000 0004 0453 5950Department of Psychiatry, Northwell Health, The Zucker Hillside Hospital, Glen Oaks, NY USA ,grid.512756.20000 0004 0370 4759Department of Psychiatry and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY USA
| | - Philipp Ritter
- grid.412282.f0000 0001 1091 2917Department of Psychiatry and Psychotherapy, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Michael Bauer
- grid.412282.f0000 0001 1091 2917Department of Psychiatry and Psychotherapy, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Andreas Jansen
- grid.10253.350000 0004 1936 9756Core-Facility Brainimaging, Faculty of Medicine, University of Marburg, Marburg, Germany ,grid.10253.350000 0004 1936 9756Department of Psychiatry, University of Marburg, Marburg, Germany ,grid.8664.c0000 0001 2165 8627Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| | - Andrea Pfennig
- grid.412282.f0000 0001 1091 2917Department of Psychiatry and Psychotherapy, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
405
|
Nuño L, Guilera G, Rojo E, Gómez-Benito J, Barrios M. An Integrated Account of Expert Perspectives on Functioning in Schizophrenia. J Clin Med 2021; 10:jcm10184223. [PMID: 34575332 PMCID: PMC8465628 DOI: 10.3390/jcm10184223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 09/09/2021] [Indexed: 11/28/2022] Open
Abstract
An integrated and interdisciplinary care system for individuals with schizophrenia is essential, which implies the need for a tool that assesses the difficulties and contextual factors of relevance to their functioning, and facilitates coordinated working across the different professions involved in their care. The International Classification of Functioning, Disability and Health Core Sets (ICF-CS) cover these requirements. This study aimed to evaluate the content validity of the ICF-CSs for schizophrenia from the perspective of experts. Six three-round Delphi studies were conducted with expert panels from different professional backgrounds which have played a significant role in the treatment of individuals with schizophrenia (psychiatry, psychology, nursing, occupational therapy, social work and physiotherapy). In total, 790 experts from 85 different countries participated in the first round. In total, 90 ICF categories and 28 Personal factors reached expert consensus (reached consensus from four or more professional perspectives). All the categories in the brief version of the ICF-CS for schizophrenia reached consensus from all the professional perspectives considered. As for the comprehensive version, 89.7% of its categories reached expert consensus. The results support the worldwide content validity of the ICF-CSs for schizophrenia from an expert perspective and underline the importance of assessing functioning by considering all the components implied.
Collapse
Affiliation(s)
- Laura Nuño
- Clinical Institute of Neuroscience (ICN), Hospital Clinic, 08036 Barcelona, Spain
- Department of Social Psychology and Quantitative Psychology, University of Barcelona, 08007 Barcelona, Spain; (G.G.); (J.G.-B.); (M.B.)
- Correspondence:
| | - Georgina Guilera
- Department of Social Psychology and Quantitative Psychology, University of Barcelona, 08007 Barcelona, Spain; (G.G.); (J.G.-B.); (M.B.)
- Group on Measurement Invariance and Analysis of Change (GEIMAC), Institute of Neurosciences, University of Barcelona, 08007 Barcelona, Spain
| | - Emilio Rojo
- Hospital Benito Menni CASM, Sisters Hospitallers, 08830 Sant Boi de Llobregat, Spain;
- Department of Psychiatry, International University of Catalonia, 08007 Barcelona, Spain
| | - Juana Gómez-Benito
- Department of Social Psychology and Quantitative Psychology, University of Barcelona, 08007 Barcelona, Spain; (G.G.); (J.G.-B.); (M.B.)
- Group on Measurement Invariance and Analysis of Change (GEIMAC), Institute of Neurosciences, University of Barcelona, 08007 Barcelona, Spain
| | - Maite Barrios
- Department of Social Psychology and Quantitative Psychology, University of Barcelona, 08007 Barcelona, Spain; (G.G.); (J.G.-B.); (M.B.)
- Group on Measurement Invariance and Analysis of Change (GEIMAC), Institute of Neurosciences, University of Barcelona, 08007 Barcelona, Spain
| |
Collapse
|
406
|
Alizadeh M, Delborde Y, Ahmadpanah M, Seifrabiee MA, Jahangard L, Bazzazi N, Brand S. Non-linear associations between retinal nerve fibre layer (RNFL) and positive and negative symptoms among men with acute and chronic schizophrenia spectrum disorder. J Psychiatr Res 2021; 141:81-91. [PMID: 34182380 DOI: 10.1016/j.jpsychires.2021.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/21/2021] [Accepted: 06/04/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND Schizophrenia Spectrum Disorder (SSD) is a chronic psychiatric disorder with modest treatment outcomes. Changes in neuronal morphology may be associated with the symptomatology of SSD. In the present study, we compared the retinal nerve fibre layer thickness (RNFLT) of typically developed adults with that of individuals with SSD in both acute and chronic stages. METHODS Fifteen healthy adult males (mean age: 36.40 years) and 30 individuals with SSD (mean age: 37.9 years) took part in the study. Among the latter, 15 had a chronic mean SSD for 15.33 years, while 15 were in an acute psychotic phase with a mean illness duration of 12.20 years. Experts rated positive and negative symptoms of SSD. Retinal nerve fibre layer thickness (RNFLT) of all participants was measured with optical coherence tomography (OCT). RESULTS Compared to healthy controls, individuals with acute SSD had the lowest macula thickness in the right eye. For nerve fiber layer atrophy, participants with acute SSD showed the largest atrophy (right eye, inferior quadrant). For retinal thickness and macular volume cube, compared to healthy controls, participants with acute SSD had the lowest thickness in the subfield of the right eye. Non-linear associations were observed between RNFL and positive and negative symptoms: e.g., for macula central and subfoveal thickness (left and right eye) and for participants with both acute and chronic SSD, exclusively positive and exclusively negative symptoms (as opposed to prevalently negative with some positive symptoms or prevalently positive with some negative symptoms) were associated with lower volumes. In participants with acute SSD, a longer disease duration was associated with thicker RNFL, while in participants with a chronic SSD a longer disease duration was associated with a thinner RNFL. CONCLUSION The present results confirm previous findings that specific neuronal morphological abnormalities can be observed among individuals with SSD. The non-linear associations between neuronal alterations and positive and negative symptomatology suggested that higher pronounced SSD severity appears to be particularly related to morphological changes. Disease duration and RNFL thickness were linearly associated, though, in opposite directions depending on the chronic or acute state.
Collapse
Affiliation(s)
- Mehdi Alizadeh
- Hamadan University of Medical Sciences. Department of Ophthalmology, Hamadan, Iran
| | - Yegane Delborde
- Hamadan University of Medical Sciences. Research Center for Behavioral Disorders and Substances Abuse. Hamadan, Iran
| | - Mohammad Ahmadpanah
- Hamadan University of Medical Sciences. Research Center for Behavioral Disorders and Substances Abuse. Hamadan, Iran
| | | | - Leila Jahangard
- Hamadan University of Medical Sciences. Research Center for Behavioral Disorders and Substances Abuse. Hamadan, Iran
| | - Nooshin Bazzazi
- Hamadan University of Medical Sciences. Department of Ophthalmology, Hamadan, Iran.
| | - Serge Brand
- University of Basel Psychiatric Clinics (UPK), Center for Affective, Stress and Sleep Disorders, Basel, Switzerland; University of Basel, Department of Sport, Exercise and Health, Division of Sport Science and Psychosocial Health, Basel, Switzerland; Kermanshah University of Medical Sciences (KUMS), Substance Abuse Prevention Research Center, Health Institute, Kermanshah, Iran; Kermanshah University of Medical Sciences (KUMS), Sleep Disorders Research Center, Health Institute, Kermanshah, Iran; Tehran University of Medical Sciences (TUMS), School of Medicine, Tehran, Iran.
| |
Collapse
|
407
|
Han LKM, Dinga R, Hahn T, Ching CRK, Eyler LT, Aftanas L, Aghajani M, Aleman A, Baune BT, Berger K, Brak I, Filho GB, Carballedo A, Connolly CG, Couvy-Duchesne B, Cullen KR, Dannlowski U, Davey CG, Dima D, Duran FLS, Enneking V, Filimonova E, Frenzel S, Frodl T, Fu CHY, Godlewska BR, Gotlib IH, Grabe HJ, Groenewold NA, Grotegerd D, Gruber O, Hall GB, Harrison BJ, Hatton SN, Hermesdorf M, Hickie IB, Ho TC, Hosten N, Jansen A, Kähler C, Kircher T, Klimes-Dougan B, Krämer B, Krug A, Lagopoulos J, Leenings R, MacMaster FP, MacQueen G, McIntosh A, McLellan Q, McMahon KL, Medland SE, Mueller BA, Mwangi B, Osipov E, Portella MJ, Pozzi E, Reneman L, Repple J, Rosa PGP, Sacchet MD, Sämann PG, Schnell K, Schrantee A, Simulionyte E, Soares JC, Sommer J, Stein DJ, Steinsträter O, Strike LT, Thomopoulos SI, van Tol MJ, Veer IM, Vermeiren RRJM, Walter H, van der Wee NJA, van der Werff SJA, Whalley H, Winter NR, Wittfeld K, Wright MJ, Wu MJ, Völzke H, Yang TT, Zannias V, de Zubicaray GI, Zunta-Soares GB, Abé C, Alda M, Andreassen OA, Bøen E, Bonnin CM, Canales-Rodriguez EJ, Cannon D, Caseras X, Chaim-Avancini TM, Elvsåshagen T, Favre P, Foley SF, Fullerton JM, et alHan LKM, Dinga R, Hahn T, Ching CRK, Eyler LT, Aftanas L, Aghajani M, Aleman A, Baune BT, Berger K, Brak I, Filho GB, Carballedo A, Connolly CG, Couvy-Duchesne B, Cullen KR, Dannlowski U, Davey CG, Dima D, Duran FLS, Enneking V, Filimonova E, Frenzel S, Frodl T, Fu CHY, Godlewska BR, Gotlib IH, Grabe HJ, Groenewold NA, Grotegerd D, Gruber O, Hall GB, Harrison BJ, Hatton SN, Hermesdorf M, Hickie IB, Ho TC, Hosten N, Jansen A, Kähler C, Kircher T, Klimes-Dougan B, Krämer B, Krug A, Lagopoulos J, Leenings R, MacMaster FP, MacQueen G, McIntosh A, McLellan Q, McMahon KL, Medland SE, Mueller BA, Mwangi B, Osipov E, Portella MJ, Pozzi E, Reneman L, Repple J, Rosa PGP, Sacchet MD, Sämann PG, Schnell K, Schrantee A, Simulionyte E, Soares JC, Sommer J, Stein DJ, Steinsträter O, Strike LT, Thomopoulos SI, van Tol MJ, Veer IM, Vermeiren RRJM, Walter H, van der Wee NJA, van der Werff SJA, Whalley H, Winter NR, Wittfeld K, Wright MJ, Wu MJ, Völzke H, Yang TT, Zannias V, de Zubicaray GI, Zunta-Soares GB, Abé C, Alda M, Andreassen OA, Bøen E, Bonnin CM, Canales-Rodriguez EJ, Cannon D, Caseras X, Chaim-Avancini TM, Elvsåshagen T, Favre P, Foley SF, Fullerton JM, Goikolea JM, Haarman BCM, Hajek T, Henry C, Houenou J, Howells FM, Ingvar M, Kuplicki R, Lafer B, Landén M, Machado-Vieira R, Malt UF, McDonald C, Mitchell PB, Nabulsi L, Otaduy MCG, Overs BJ, Polosan M, Pomarol-Clotet E, Radua J, Rive MM, Roberts G, Ruhe HG, Salvador R, Sarró S, Satterthwaite TD, Savitz J, Schene AH, Schofield PR, Serpa MH, Sim K, Soeiro-de-Souza MG, Sutherland AN, Temmingh HS, Timmons GM, Uhlmann A, Vieta E, Wolf DH, Zanetti MV, Jahanshad N, Thompson PM, Veltman DJ, Penninx BWJH, Marquand AF, Cole JH, Schmaal L. Brain aging in major depressive disorder: results from the ENIGMA major depressive disorder working group. Mol Psychiatry 2021; 26:5124-5139. [PMID: 32424236 PMCID: PMC8589647 DOI: 10.1038/s41380-020-0754-0] [Show More Authors] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 04/01/2020] [Accepted: 04/23/2020] [Indexed: 01/15/2023]
Abstract
Major depressive disorder (MDD) is associated with an increased risk of brain atrophy, aging-related diseases, and mortality. We examined potential advanced brain aging in adult MDD patients, and whether this process is associated with clinical characteristics in a large multicenter international dataset. We performed a mega-analysis by pooling brain measures derived from T1-weighted MRI scans from 19 samples worldwide. Healthy brain aging was estimated by predicting chronological age (18-75 years) from 7 subcortical volumes, 34 cortical thickness and 34 surface area, lateral ventricles and total intracranial volume measures separately in 952 male and 1236 female controls from the ENIGMA MDD working group. The learned model coefficients were applied to 927 male controls and 986 depressed males, and 1199 female controls and 1689 depressed females to obtain independent unbiased brain-based age predictions. The difference between predicted "brain age" and chronological age was calculated to indicate brain-predicted age difference (brain-PAD). On average, MDD patients showed a higher brain-PAD of +1.08 (SE 0.22) years (Cohen's d = 0.14, 95% CI: 0.08-0.20) compared with controls. However, this difference did not seem to be driven by specific clinical characteristics (recurrent status, remission status, antidepressant medication use, age of onset, or symptom severity). This highly powered collaborative effort showed subtle patterns of age-related structural brain abnormalities in MDD. Substantial within-group variance and overlap between groups were observed. Longitudinal studies of MDD and somatic health outcomes are needed to further assess the clinical value of these brain-PAD estimates.
Collapse
Grants
- RF1 AG041915 NIA NIH HHS
- G0802594 Medical Research Council
- R01 MH083968 NIMH NIH HHS
- MR/L010305/1 Medical Research Council
- R01 MH116147 NIMH NIH HHS
- T32 AG058507 NIA NIH HHS
- R01 HD050735 NICHD NIH HHS
- R21 MH113871 NIMH NIH HHS
- T35 AG026757 NIA NIH HHS
- R56 AG058854 NIA NIH HHS
- K23 MH090421 NIMH NIH HHS
- Wellcome Trust
- R61 AT009864 NCCIH NIH HHS
- P41 EB015922 NIBIB NIH HHS
- P20 GM121312 NIGMS NIH HHS
- R37 MH101495 NIMH NIH HHS
- P41 RR008079 NCRR NIH HHS
- T32 MH073526 NIMH NIH HHS
- 104036/Z/14/Z Wellcome Trust
- UL1 TR001872 NCATS NIH HHS
- Department of Health
- U54 EB020403 NIBIB NIH HHS
- R01 MH117601 NIMH NIH HHS
- MR/R024790/2 Medical Research Council
- K01 MH117442 NIMH NIH HHS
- R01 MH085734 NIMH NIH HHS
- R21 AT009173 NCCIH NIH HHS
- RF1 AG051710 NIA NIH HHS
- R01 AG059874 NIA NIH HHS
- CC was supported by NIH grants U54 EB020403, RF1 AG041915, RF1AG051710, P41EB015922, R01MH116147, and R56AG058854
- Russian Science Foundation (RSF)
- The study was supported by a grant from the German Federal Ministry of Education and Research (BMBF; grant FKZ-01ER0816 and FKZ-01ER1506)
- Dr. Busatto was supported by the funding agencies FAPESP and CNPq, Brazil
- Department of Health | National Health and Medical Research Council (NHMRC)
- Deutsche Forschungsgemeinschaft (German Research Foundation)
- This study was funded by National Health and Medical Research Council of Australia (NHMRC) Project Grants 1064643 (Principal Investigator BJH) and 1024570 (Principal Investigator CGD).
- Science Foundation Ireland (SFI)
- This work was supported by NIH grant R37 MH101495
- The Study of Health in Pomerania (SHIP) is part of the Community Medicine Research net (CMR) (http://www.medizin.uni-greifswald.de/icm) of the University Medicine Greifswald, which is supported by the German Federal State of Mecklenburg- West Pomerania. MRI scans in SHIP and SHIP-TREND have been supported by a joint grant from Siemens Healthineers, Erlangen, Germany and the Federal State of Mecklenburg-West Pomerania. This study was further supported by the EU-JPND Funding for BRIDGET (FKZ:01ED1615).
- Gratama Foundation, the Netherlands (2012/35 to NG)
- This work was partially supported by the Deutsche Forschungsgemeinschaft (DFG) via grants to OG (GR1950/5-1 and GR1950/10-1).
- This study was supported by the following National Health and Medical Research Council funding sources: Programme Grant (no. 566529), Centres of Clinical Research Excellence Grant (no. 264611), Australia Fellowship (no. 511921) and Clinical Research Fellowship (no. 402864).
- This study was funded by the National Institute of Mental health grant K23MH090421 (D. Cullen) and Biotechnology Research Center grant P41RR008079 (Center for Magnetic Resonance Research), the National Alliance for Research on Schizophrenia and Depression, the University of Minnesota Graduate School, and the Minnesota Medical Foundation. This work was carried out in part using computing resources at the University of Minnesota Supercomputing Institute.
- This work was funded by the German Research Foundation (DFG, grant FOR2107 KR 3822/7-2 to AK; FOR2107 KI 588/14-2 to TK and FOR2107 JA 1890/7-2 to AJ)
- The research leading to these results was supported by IMAGEMEND, which received funding from the European Community's Seventh Framework Programme (FP7/2007-2013) under grant agreement no. 602450. This paper reflects only the author’s views and the European Union is not liable for any use that may be made of the information contained therein. This work was also supported by a Wellcome Trust Strategic Award 104036/Z/14/Z
- The QTIM dataset was supported by the Australian National Health and Medical Research Council (Project Grants No. 496682 and 1009064) and US National Institute of Child Health and Human Development(RO1HD050735)
- MJP was funded by Ministerio de Ciencia e Innovación of Spanish Government (ISCIII) through a "Miguel Servet II" (CP16/00020)
- Jair C. Soares supported by the Pat Rutherford Chair in Psychiatry, UTHealth. Jair Soares has received research support from Allergan, Pfizer, Johnson & Johnson, Alquermes and COMPASS. He is a member of the speakers’ bureaus for Sunovion and Sanofi and he is a consultant for Johnson & Johnson.
- The QTIM dataset was supported by the Australian National Health and Medical Research Council (Project Grants No. 496682 and 1009064) and US National Institute of Child Health and Human Development (RO1HD050735)
- SIT was supported in part by NIH grants U54 EB020403, RF1 AG041915, RF1AG051710, P41EB015922, R01MH116147, and R56AG058854
- The CODE cohort was collected from studies funded by Lundbeck and the German Research Foundation (WA 1539/4-1, SCHN 1205/3-1, SCHR443/11-1)
- Canadian Institutes of Health Research (142255)
- Fundet by Research Council of Norway (223273, 248778, 273291), NIH (ENIGMA grants)
- Funded by the South-Eastern Norway Regional Health Authority and a research grant from Mrs. Throne-Holst.
- This work was supported by the Health Research Board, Ireland and the Irish Research Council
- The Cardiff dataset was supported through a 2010 NARSAD Young Investigator Award (ref: 17319) to Dr. Xavier Caseras
- This work was supported by the FRM (Fondation pour la recherche Biomédicale) "Bio-informatique pour la biologie" 2014 grant
- Canadian Institutes of Health Research (103703, 106469), Nova Scotia Health Research Foundation, Dalhousie Clinical Research Scholarship to T. Hajek, Brain & Behavior Research Foundation (formerly NARSAD) 2007 Young Investigator and 2015 Independent Investigator Awards to T. Hajek
- This work was supported by the University Research Council of the University of Cape Town and the National Research Foundation of South Africa.
- Australian NHMRC Program Grant 1037196 and Project Grants 1063960 and 1066177.
- This work was supported by research grants from Grenoble University Hospital
- This work was supported by the Generalitat de Catalunya (2014 SGR 1573) and Instituto de Salud Carlos III (CPII16/00018) and (PI14/01151 and PI14/01148).
- The DIADE dataset was suported by a ZonMW OOG 2007 grant (100-002-034). HG Ruhe was supported by a ZonMW VENI grant (016.126.059)
- JS is supported by the National Institute of General Medical Sciences (P20GM121312) and the National Insitute of Mental Health (R21MH113871)
- Dr. Mauricio was supported by the funding agencies CAPES, Brazil
- This study was supported by R01MH083968, Desert-Pacific Mental Illness Research Education and Clinical Center, and the US National Science Foundation (Science Gateways Community Institutes; XSEDE).
- GT's work was supported by the National Institutes of Health, Grant T35 AG026757/AG/NIA and the University of California San Diego, Stein Institute for Research on Aging
- "EV thanks the support of the Spanish Ministry of Science, Innovation and Universities (PI15/00283) integrated into the Plan Nacional de I+D+I y cofinanciado por el ISCIII-Subdirección General de Evaluación y el Fondo Europeo de Desarrollo Regional (FEDER); CIBERSAM; and the Comissionat per a Universitats i Recerca del DIUE de la Generalitat de Catalunya to the Bipolar Disorders Group (2017 SGR 1365) and the project SLT006/17/00357, from PERIS 2016-2020 (Departament de Salut). CERCA Programme/Generalitat de Catalunya. "
- Dr. Zanetti was supported by FAPESP, Brazil (grant no. 2013/03905-4).
- NIH grants R01 MH117601, R01 AG059874, U54 EB020403, RF1 AG041915, RF1AG051710, P41EB015922, R01MH116147, and R56AG058854
- PT was supported in part by NIH grants U54 EB020403, RF1 AG041915, RF1AG051710, P41EB015922, R01MH116147, and R56AG058854
- Dr Cole is funded by a UKRI Innovation Fellowship
- This work was supported by NIH grants U54 EB020403 and R01 MH116147. LS is supported by a NHMRC Career Development Fellowship (1140764).
Collapse
Affiliation(s)
- Laura K M Han
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit & GGZinGeest, Amsterdam, The Netherlands.
| | - Richard Dinga
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit & GGZinGeest, Amsterdam, The Netherlands
- Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, The Netherlands
| | - Tim Hahn
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Christopher R K Ching
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lisa T Eyler
- Desert-Pacific Mental Illness Research Education and Clinical Center, VA San Diego Healthcare, San Diego, CA, USA
- Department of Psychiatry, University of California San Diego, Los Angeles, CA, USA
| | - Lyubomir Aftanas
- FSSBI "Scientific Research Institute of Physiology & Basic Medicine", Laboratory of Affective, Cognitive & Translational Neuroscience, Novosibirsk, Russia
- Department of Neuroscience, Novosibirsk State University, Novosibirsk, Russia
| | - Moji Aghajani
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit & GGZinGeest, Amsterdam, The Netherlands
| | - André Aleman
- Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Clinical and Developmental Neuropsychology, University of Groningen, Groningen, The Netherlands
| | - Bernhard T Baune
- Department of Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Ivan Brak
- FSSBI "Scientific Research Institute of Physiology & Basic Medicine", Laboratory of Affective, Cognitive & Translational Neuroscience, Novosibirsk, Russia
- Laboratory of Experimental & Translational Neuroscience, Novosibirsk State University, Novosibirsk, Russia
| | - Geraldo Busatto Filho
- Laboratory of Psychiatric Neuroimaging (LIM-21), Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Angela Carballedo
- Department for Psychiatry, Trinity College Dublin, Dublin, Ireland
- North Dublin Mental Health Services, Dublin, Ireland
| | - Colm G Connolly
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA
| | | | - Kathryn R Cullen
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Udo Dannlowski
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Christopher G Davey
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Danai Dima
- Department of Psychology, School of Arts and Social Sciences, City, University of London, London, UK
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College, London, UK
| | - Fabio L S Duran
- Laboratory of Psychiatric Neuroimaging (LIM-21), Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Verena Enneking
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Elena Filimonova
- FSSBI "Scientific Research Institute of Physiology & Basic Medicine", Laboratory of Affective, Cognitive & Translational Neuroscience, Novosibirsk, Russia
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Frodl
- Department for Psychiatry, Trinity College Dublin, Dublin, Ireland
- Department of Psychiatry and Psychotherapy, Otto von Guericke University (OVGU), Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Cynthia H Y Fu
- Centre for Affective Disorders, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- School of Psychology, University of East London, London, UK
| | | | - Ian H Gotlib
- Department of Psychology, Stanford University, Stanford, CA, USA
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center of Neurodegenerative Diseases (DZNE) Site Rostock/Greifswald, Greifswald, Germany
| | - Nynke A Groenewold
- Interdisciplinary Center Psychopathology and Emotion regulation (ICPE), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | | | - Oliver Gruber
- Section for Experimental Psychopathology and Neuroimaging, Department of Psychiatry, University of Heidelberg, Heidelberg, Germany
| | - Geoffrey B Hall
- Department of Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, ON, Canada
| | - Ben J Harrison
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Melbourne, VIC, Australia
| | - Sean N Hatton
- Youth Mental Health Team, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Department of Neuroscience, University of California San Diego, San Diego, CA, USA
| | - Marco Hermesdorf
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Ian B Hickie
- Youth Mental Health Team, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - Tiffany C Ho
- Department of Psychology, Stanford University, Stanford, CA, USA
- Department of Psychiatry & Behavioral Sciences, Standord University, Stanford, CA, USA
| | - Norbert Hosten
- Department of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Andreas Jansen
- Department of Psychiatry, Philipps-University Marburg, Marburg, Germany
| | - Claas Kähler
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Tilo Kircher
- Department of Psychiatry, Philipps-University Marburg, Marburg, Germany
| | | | - Bernd Krämer
- Section for Experimental Psychopathology and Neuroimaging, Department of Psychiatry, University of Heidelberg, Heidelberg, Germany
| | - Axel Krug
- Department of Psychiatry, Philipps-University Marburg, Marburg, Germany
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Jim Lagopoulos
- Youth Mental Health Team, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Sunshine Coast Mind and Neuroscience Institute, University of the Sunshine Coast QLD, Sippy Downs, QLD, Australia
| | - Ramona Leenings
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Frank P MacMaster
- Departments of Psychiatry and Pediatrics, University of Calgary, Calgary, AB, Canada
- Addictions and Mental Health Strategic Clinical Network, Calgary, AB, Canada
| | - Glenda MacQueen
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada
| | - Andrew McIntosh
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Quinn McLellan
- Departments of Psychiatry and Pediatrics, University of Calgary, Calgary, AB, Canada
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Katie L McMahon
- School of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Sarah E Medland
- QIMR Berghofer Medical Research Instititute, Brisbane, QLD, Australia
| | - Bryon A Mueller
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Benson Mwangi
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Evgeny Osipov
- Laboratory of Experimental & Translational Neuroscience, Novosibirsk State University, Novosibirsk, Russia
| | - Maria J Portella
- Institut d'Investigació Biomèdica Sant Pau, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Cibersam, Spain
| | - Elena Pozzi
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, Minnesota, USA
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Melbourne, VIC, Australia
| | - Liesbeth Reneman
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, AMC, Amsterdam, The Netherlands
| | - Jonathan Repple
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Pedro G P Rosa
- Laboratory of Psychiatric Neuroimaging (LIM-21), Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Matthew D Sacchet
- Center for Depression, Anxiety, and Stress Research, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | | | - Knut Schnell
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Department of Psychiatry and Psychotherapy, Asklepios Fachklinikum Göttingen, Göttingen, Germany
| | - Anouk Schrantee
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, AMC, Amsterdam, The Netherlands
| | - Egle Simulionyte
- Department of Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, ON, Canada
| | - Jair C Soares
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jens Sommer
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Dan J Stein
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- SA MRC Unit on Risk and Resilience, University of Cape Town, Cape Town, South Africa
| | - Olaf Steinsträter
- Department of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Lachlan T Strike
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| | - Sophia I Thomopoulos
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Marie-José van Tol
- Cognitive Neuroscience Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ilya M Veer
- Division of Mind and Brain Research, Department of Psychiatry and Psychotherapy CCM, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Robert R J M Vermeiren
- Department of Child Psychiatry, University Medical Center, Leiden, The Netherlands
- Leiden Institute for Brain and Cognition, Leiden University, Leiden, The Netherlands
| | - Henrik Walter
- Division of Mind and Brain Research, Department of Psychiatry and Psychotherapy CCM, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nic J A van der Wee
- Leiden Institute for Brain and Cognition, Leiden University, Leiden, The Netherlands
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
| | - Steven J A van der Werff
- Leiden Institute for Brain and Cognition, Leiden University, Leiden, The Netherlands
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
| | - Heather Whalley
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Nils R Winter
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center of Neurodegenerative Diseases (DZNE) Site Rostock/Greifswald, Greifswald, Germany
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
- Centre for Advanced Imaging, University of Queensland, Brisbane, QLD, Australia
| | - Mon-Ju Wu
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Tony T Yang
- Department of Psychiatry, Division of Child and Adolescent Psychiatry, UCSF School of Medicine, UCSF, San Francisco, CA, USA
| | | | - Greig I de Zubicaray
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
- Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Giovana B Zunta-Soares
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Christoph Abé
- Department of Clinical Neuroscience, Osher Center, Karolinska Institutet, Stockholm, Sweden
| | - Martin Alda
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Ole A Andreassen
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Erlend Bøen
- Clinic for Mental Health and Dependency, C-L psychiatry and Psychosomatic Unit, Oslo University Hospital, Oslo, Norway
| | - Caterina M Bonnin
- Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | | | - Dara Cannon
- Centre for Neuroimaging & Cognitive Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, H91 TK33, Galway, Ireland
| | - Xavier Caseras
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Tiffany M Chaim-Avancini
- Laboratory of Psychiatric Neuroimaging (LIM-21), Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Torbjørn Elvsåshagen
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Pauline Favre
- UNIACT, Psychiatry Team, Neurospin, Atomic Energy Commission, Gif-Sur-Yvette, France
- Translational Psychiatry Team, Pôle de psychiatrie, Faculté de Médecine, APHP, Hôpitaux Universitaires Mondor, INSERM, U955, Créteil, France
| | - Sonya F Foley
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, UK
| | - Janice M Fullerton
- Neuroscience Research Australia, Randwick, Sydney, NSW, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Jose M Goikolea
- Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - Bartholomeus C M Haarman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tomas Hajek
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Chantal Henry
- Université de Paris, Service Hospitalo-Universitaire, GHU Paris Psychiatrie & Neuroscience, F-75014, Paris, France
| | - Josselin Houenou
- UNIACT, Psychiatry Team, Neurospin, Atomic Energy Commission, Gif-Sur-Yvette, France
- Translational Psychiatry Team, Pôle de psychiatrie, Faculté de Médecine, APHP, Hôpitaux Universitaires Mondor, INSERM, U955, Créteil, France
| | - Fleur M Howells
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Martin Ingvar
- Department of Clinical Neuroscience, Osher Center, Karolinska Institutet, Stockholm, Sweden
| | | | - Beny Lafer
- Department of Psychiatry, School of Medicine, University of Sao Paulo (FMUSP), Sao Paulo, Brazil
| | - Mikael Landén
- Department of Clinical Neuroscience, Osher Center, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Rodrigo Machado-Vieira
- Department of Psychiatry, School of Medicine, University of Sao Paulo (FMUSP), Sao Paulo, Brazil
| | - Ulrik F Malt
- Department of Clinical Neuroscience, University of Oslo, Oslo, Norway
- Clinic for Psychiatry and Dependency, C-L psychiatry and Psychosomatic Unit, Oslo University Hospital, Oslo, Norway
| | - Colm McDonald
- Centre for Neuroimaging & Cognitive Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, H91 TK33, Galway, Ireland
| | - Philip B Mitchell
- School of Psychiatry, University of New South Wales, Kingsford, Sydney, NSW, Australia
- Black Dog Institute, Prince of Wales Hospital, Randwick, Sydney, NSW, Australia
| | - Leila Nabulsi
- Centre for Neuroimaging & Cognitive Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, H91 TK33, Galway, Ireland
| | - Maria Concepcion Garcia Otaduy
- Instituto de Radiologia, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Bronwyn J Overs
- Neuroscience Research Australia, Randwick, Sydney, NSW, Australia
| | - Mircea Polosan
- Department of Psychiatry and Neurology, CHU Grenoble Alpes, Université Grenoble Alpes, F-38000, Grenoble, France
- Inserm 1216, Grenoble Institut des Neurosciences, GIN, F-38000, Grenoble, France
| | - Edith Pomarol-Clotet
- FIDMAG Germanes Hospitalàries Research Foundation, CIBERSAM, Barcelona, Catalonia, Spain
| | - Joaquim Radua
- Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - Maria M Rive
- Department of Psychiatry, Amsterdam University Medical Centers, AMC, Amsterdam, The Netherlands
| | - Gloria Roberts
- School of Psychiatry, University of New South Wales, Kingsford, Sydney, NSW, Australia
- Black Dog Institute, Prince of Wales Hospital, Randwick, Sydney, NSW, Australia
| | - Henricus G Ruhe
- Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, The Netherlands
- Department of Psychiatry, Amsterdam University Medical Centers, AMC, Amsterdam, The Netherlands
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Raymond Salvador
- FIDMAG Germanes Hospitalàries Research Foundation, CIBERSAM, Barcelona, Catalonia, Spain
| | - Salvador Sarró
- FIDMAG Germanes Hospitalàries Research Foundation, CIBERSAM, Barcelona, Catalonia, Spain
| | - Theodore D Satterthwaite
- Department of Psychiatry, University of Pennsylvannia Perelman School of Medicine, Philadelphia, PA, USA
| | - Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Oxley College of Health Sciences, The University of Tulsa, Tulsa, OK, USA
| | - Aart H Schene
- Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, The Netherlands
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter R Schofield
- Neuroscience Research Australia, Randwick, Sydney, NSW, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Mauricio H Serpa
- Laboratory of Psychiatric Neuroimaging (LIM-21), Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Kang Sim
- West Region and Research Division, Institute of Mental Health, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Ashley N Sutherland
- Department of Psychiatry, University of California San Diego, Los Angeles, CA, USA
| | - Henk S Temmingh
- Section for Experimental Psychopathology and Neuroimaging, Department of Psychiatry, University of Heidelberg, Heidelberg, Germany
- Valkenberg Psychiatric Hospital, Cape Town, South Africa
| | - Garrett M Timmons
- Department of Psychiatry, University of California San Diego, Los Angeles, CA, USA
| | - Anne Uhlmann
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Eduard Vieta
- Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - Daniel H Wolf
- Department of Psychiatry, University of Pennsylvannia Perelman School of Medicine, Philadelphia, PA, USA
| | - Marcus V Zanetti
- Laboratory of Psychiatric Neuroimaging (LIM-21), Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
- Instituto de Ensino e Pesquisa, Hospital Sírio-Libanês, Sao Paulo, SP, Brazil
| | - Neda Jahanshad
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Dick J Veltman
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit & GGZinGeest, Amsterdam, The Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit & GGZinGeest, Amsterdam, The Netherlands
| | - Andre F Marquand
- Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - James H Cole
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College, London, UK
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- Dementia Research Centre, Institute of Neurology, University College London, London, UK
| | - Lianne Schmaal
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
408
|
Cheng W, Frei O, van der Meer D, Wang Y, O’Connell KS, Chu Y, Bahrami S, Shadrin AA, Alnæs D, Hindley GFL, Lin A, Karadag N, Fan CC, Westlye LT, Kaufmann T, Molden E, Dale AM, Djurovic S, Smeland OB, Andreassen OA. Genetic Association Between Schizophrenia and Cortical Brain Surface Area and Thickness. JAMA Psychiatry 2021; 78:1020-1030. [PMID: 34160554 PMCID: PMC8223140 DOI: 10.1001/jamapsychiatry.2021.1435] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/28/2021] [Indexed: 01/03/2023]
Abstract
Importance Schizophrenia is a complex heritable disorder associated with many genetic variants, each with a small effect. While cortical differences between patients with schizophrenia and healthy controls are consistently reported, the underlying molecular mechanisms remain elusive. Objective To investigate the extent of shared genetic architecture between schizophrenia and brain cortical surface area (SA) and thickness (TH) and to identify shared genomic loci. Design, Setting, and Participants Independent genome-wide association study data on schizophrenia (Psychiatric Genomics Consortium and CLOZUK: n = 105 318) and SA and TH (UK Biobank: n = 33 735) were obtained. The extent of polygenic overlap was investigated using MiXeR. The specific shared genomic loci were identified by conditional/conjunctional false discovery rate analysis and were further examined in 3 independent cohorts. Data were collected from December 2019 to February 2021, and data analysis was performed from May 2020 to February 2021. Main Outcomes and Measures The primary outcomes were estimated fractions of polygenic overlap between schizophrenia, total SA, and average TH and a list of functionally characterized shared genomic loci. Results Based on genome-wide association study data from 139 053 participants, MiXeR estimated schizophrenia to be more polygenic (9703 single-nucleotide variants [SNVs]) than total SA (2101 SNVs) and average TH (1363 SNVs). Most SNVs associated with total SA (1966 of 2101 [93.6%]) and average TH (1322 of 1363 [97.0%]) may be associated with the development of schizophrenia. Subsequent conjunctional false discovery rate analysis identified 44 and 23 schizophrenia risk loci shared with total SA and average TH, respectively. The SNV associations of shared loci between schizophrenia and total SA revealed en masse concordant association between the discovery and independent cohorts. After removing high linkage disequilibrium regions, such as the major histocompatibility complex region, the shared loci were enriched in immunologic signature gene sets. Polygenic overlap and shared loci between schizophrenia and schizophrenia-associated regions of interest for SA (superior frontal and middle temporal gyri) and for TH (superior temporal, inferior temporal, and superior frontal gyri) were also identified. Conclusions and Relevance This study demonstrated shared genetic loci between cortical morphometry and schizophrenia, among which a subset are associated with immunity. These findings provide an insight into the complex genetic architecture and associated with schizophrenia.
Collapse
Affiliation(s)
- Weiqiu Cheng
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Oleksandr Frei
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dennis van der Meer
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Yunpeng Wang
- Centre for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
| | - Kevin S. O’Connell
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Yunhan Chu
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Shahram Bahrami
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alexey A. Shadrin
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dag Alnæs
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Guy F. L. Hindley
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Psychosis Studies, Institute of Psychiatry, Psychology and Neurosciences, King’s College London, London, United Kingdom
| | - Aihua Lin
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Naz Karadag
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Chun-Chieh Fan
- Population Neuroscience and Genetics Lab, University of California, San Diego, La Jolla
- Center for Human Development, University of California, San Diego, La Jolla
| | - Lars T. Westlye
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Tobias Kaufmann
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Espen Molden
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway
- Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Anders M. Dale
- Department of Radiology, University of California, San Diego, La Jolla
- Department of Psychiatry, University of California, San Diego, La Jolla
- Department of Neurosciences, University of California San Diego, La Jolla
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Olav B. Smeland
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole A. Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
409
|
Interindividual variability of functional connectome in schizophrenia. Schizophr Res 2021; 235:65-73. [PMID: 34329851 DOI: 10.1016/j.schres.2021.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 07/08/2021] [Accepted: 07/11/2021] [Indexed: 11/21/2022]
Abstract
Schizophrenia is a complex psychiatric disorder that displays an outstanding interindividual variability in clinical manifestation and neurobiological substrates. A better characterization and quantification of this heterogeneity could guide the search for both common abnormalities (linked to lower intersubject variability) and the presence of biological subtypes (leading to a greater heterogeneity across subjects). In the current study, we address interindividual variability in functional connectome by means of resting-state fMRI in a large sample of patients with schizophrenia and healthy controls. Among the different metrics of distance/dissimilarity used to assess variability, geodesic distance showed robust results to head motion. The main findings of the current study point to (i) a higher between subject heterogeneity in the functional connectome of patients, (ii) variable levels of heterogeneity throughout the cortex, with greater variability in frontoparietal and default mode networks, and lower variability in the salience network, and (iii) an association of whole-brain variability with levels of clinical symptom severity and with topological properties of brain networks, suggesting that the average functional connectome overrepresents those patients with lower functional integration and with more severe clinical symptoms. Moreover, after performing a graph theoretical analysis of brain networks, we found that patients with more severe clinical symptoms had decreased connectivity at both whole-brain level and within the salience network, and that patients with higher negative symptoms had large-scale functional integration deficits.
Collapse
|
410
|
Lam MTY, Duttke SH, Odish MF, Le HD, Hansen EA, Nguyen CT, Trescott S, Kim R, Deota S, Chang MW, Patel A, Hepokoski M, Alotaibi M, Rolfsen M, Perofsky K, Warden AS, Foley J, Ramirez SI, Dan JM, Abbott RK, Crotty S, Crotty Alexander LE, Malhotra A, Panda S, Benner CW, Coufal NG. Profiling Transcription Initiation in Peripheral Leukocytes Reveals Severity-Associated Cis-Regulatory Elements in Critical COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.08.24.457187. [PMID: 34462742 DOI: 10.1101/2021.10.28.466336] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The contribution of transcription factors (TFs) and gene regulatory programs in the immune response to COVID-19 and their relationship to disease outcome is not fully understood. Analysis of genome-wide changes in transcription at both promoter-proximal and distal cis-regulatory DNA elements, collectively termed the 'active cistrome,' offers an unbiased assessment of TF activity identifying key pathways regulated in homeostasis or disease. Here, we profiled the active cistrome from peripheral leukocytes of critically ill COVID-19 patients to identify major regulatory programs and their dynamics during SARS-CoV-2 associated acute respiratory distress syndrome (ARDS). We identified TF motifs that track the severity of COVID- 19 lung injury, disease resolution, and outcome. We used unbiased clustering to reveal distinct cistrome subsets delineating the regulation of pathways, cell types, and the combinatorial activity of TFs. We found critical roles for regulatory networks driven by stimulus and lineage determining TFs, showing that STAT and E2F/MYB regulatory programs targeting myeloid cells are activated in patients with poor disease outcomes and associated with single nucleotide genetic variants implicated in COVID-19 susceptibility. Integration with single-cell RNA-seq found that STAT and E2F/MYB activation converged in specific neutrophils subset found in patients with severe disease. Collectively we demonstrate that cistrome analysis facilitates insight into disease mechanisms and provides an unbiased approach to evaluate global changes in transcription factor activity and stratify patient disease severity.
Collapse
Affiliation(s)
- Michael Tun Yin Lam
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, CA USA
- Laboratory of Regulatory Biology, Salk Institute of Biological Studies, La Jolla, CA, USA
| | - Sascha H Duttke
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, CA, USA
| | - Mazen F Odish
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, CA USA
| | - Hiep D Le
- Laboratory of Regulatory Biology, Salk Institute of Biological Studies, La Jolla, CA, USA
| | - Emily A Hansen
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
- Department of Pediatrics, University of California, San Diego, CA, USA
| | - Celina T Nguyen
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Samantha Trescott
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
- Department of Pediatrics, University of California, San Diego, CA, USA
| | - Roy Kim
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
- Department of Pediatrics, University of California, San Diego, CA, USA
| | - Shaunak Deota
- Laboratory of Regulatory Biology, Salk Institute of Biological Studies, La Jolla, CA, USA
| | - Max W Chang
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, CA, USA
| | - Arjun Patel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, CA USA
| | - Mark Hepokoski
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, CA USA
| | - Mona Alotaibi
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, CA USA
| | - Mark Rolfsen
- Internal Medicine Residency Program, Department of Medicine, UC San Diego, CA, USA
| | - Katherine Perofsky
- Department of Pediatrics, University of California, San Diego, CA, USA
- Rady Children's Hospital, San Diego, CA
| | - Anna S Warden
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, CA, USA
| | | | - Sydney I Ramirez
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego
- Center for Infectious Diseases and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA
| | - Jennifer M Dan
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego
- Center for Infectious Diseases and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA
| | - Robert K Abbott
- Center for Infectious Diseases and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA
- Consortium for HIV/AIDS Vaccine Development (CHVAD), The Scripps Research Institute, La Jolla, CA, USA
| | - Shane Crotty
- Center for Infectious Diseases and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA
| | - Laura E Crotty Alexander
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, CA USA
| | - Atul Malhotra
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, CA USA
| | - Satchidananda Panda
- Laboratory of Regulatory Biology, Salk Institute of Biological Studies, La Jolla, CA, USA
| | - Christopher W Benner
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, CA, USA
| | - Nicole G Coufal
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
- Department of Pediatrics, University of California, San Diego, CA, USA
- Rady Children's Hospital, San Diego, CA
| |
Collapse
|
411
|
Koike S, Uematsu A, Sasabayashi D, Maikusa N, Takahashi T, Ohi K, Nakajima S, Noda Y, Hirano Y. Recent Advances and Future Directions in Brain MR Imaging Studies in Schizophrenia: Toward Elucidating Brain Pathology and Developing Clinical Tools. Magn Reson Med Sci 2021; 21:539-552. [PMID: 34408115 DOI: 10.2463/mrms.rev.2021-0050] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Schizophrenia is a common severe psychiatric disorder that affects approximately 1% of general population through the life course. Historically, in Kraepelin's time, schizophrenia was a disease unit conceptualized as dementia praecox; however, since then, the disease concept has changed. Recent MRI studies had shown that the neuropathology of the brain in this disorder was characterized by mild progression before and after the onset of the disease, and that the brain alterations were relatively smaller than assumed. Although genetic factors contribute to the brain alterations in schizophrenia, which are thought to be trait differences, other changes include factors that are common in psychiatric diseases. Furthermore, it has been shown that the brain differences specific to schizophrenia were relatively small compared to other changes, such as those caused by brain development, aging, and gender. In addition, compared to the disease and participant factors, machine and imaging protocol differences could affect MRI signals, which should be addressed in multi-site studies. Recent advances in MRI modalities, such as multi-shell diffusion-weighted imaging, magnetic resonance spectroscopy, and multimodal brain imaging analysis, may be candidates to sharpen the characterization of schizophrenia-specific factors and provide new insights. The Brain/MINDS Beyond Human Brain MRI (BMB-HBM) project has been launched considering the differences and noises irrespective of the disease pathologies and includes the future perspectives of MRI studies for various psychiatric and neurological disorders. The sites use restricted MRI machines and harmonized multi-modal protocols, standardized image preprocessing, and traveling subject harmonization. Data sharing to the public will be planned in FY 2024. In the future, we believe that combining a high-quality human MRI dataset with genetic data, randomized controlled trials, and MRI for non-human primates and animal models will enable us to understand schizophrenia, elucidate its neural bases and therapeutic targets, and provide tools for clinical application at bedside.
Collapse
Affiliation(s)
- Shinsuke Koike
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and Sciences, The University of Tokyo.,University of Tokyo Institute for Diversity & Adaptation of Human Mind (UTIDAHM).,University of Tokyo Center for Integrative Science of Human Behavior (CiSHuB).,The International Research Center for Neurointelligence (WPI-IRCN), Institutes for Advanced Study (UTIAS), The University of Tokyo
| | - Akiko Uematsu
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and Sciences, The University of Tokyo
| | - Daiki Sasabayashi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences.,Research Center for Idling Brain Science (RCIBS), University of Toyama
| | - Norihide Maikusa
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and Sciences, The University of Tokyo
| | - Tsutomu Takahashi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences.,Research Center for Idling Brain Science (RCIBS), University of Toyama
| | - Kazutaka Ohi
- Department of Psychiatry and Psychotherapy, Gifu University Graduate School of Medicine
| | | | - Yoshihiro Noda
- Department of Neuropsychiatry, Keio University School of Medicine
| | - Yoji Hirano
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University.,Institute of Industrial Science, The University of Tokyo
| |
Collapse
|
412
|
Maikusa N, Zhu Y, Uematsu A, Yamashita A, Saotome K, Okada N, Kasai K, Okanoya K, Yamashita O, Tanaka SC, Koike S. Comparison of traveling-subject and ComBat harmonization methods for assessing structural brain characteristics. Hum Brain Mapp 2021; 42:5278-5287. [PMID: 34402132 PMCID: PMC8519865 DOI: 10.1002/hbm.25615] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/25/2022] Open
Abstract
Multisite magnetic resonance imaging (MRI) is increasingly used in clinical research and development. Measurement biases—caused by site differences in scanner/image‐acquisition protocols—negatively influence the reliability and reproducibility of image‐analysis methods. Harmonization can reduce bias and improve the reproducibility of multisite datasets. Herein, a traveling‐subject (TS) dataset including 56 T1‐weighted MRI scans of 20 healthy participants in three different MRI procedures—20, 19, and 17 subjects in Procedures 1, 2, and 3, respectively—was considered to compare the reproducibility of TS‐GLM, ComBat, and TS‐ComBat harmonization methods. The minimum participant count required for harmonization was determined, and the Cohen's d between different MRI procedures was evaluated as a measurement‐bias indicator. The measurement‐bias reduction realized with different methods was evaluated by comparing test–retest scans for 20 healthy participants. Moreover, the minimum subject count for harmonization was determined by comparing test–retest datasets. The results revealed that TS‐GLM and TS‐ComBat reduced measurement bias by up to 85 and 81.3%, respectively. Meanwhile, ComBat showed a reduction of only 59.0%. At least 6 TSs were required to harmonize data obtained from different MRI scanners, complying with the imaging protocol predetermined for multisite investigations and operated with similar scan parameters. The results indicate that TS‐based harmonization outperforms ComBat for measurement‐bias reduction and is optimal for MRI data in well‐prepared multisite investigations. One drawback is the small sample size used, potentially limiting the applicability of ComBat. Investigation on the number of subjects needed for a large‐scale study is an interesting future problem.
Collapse
Affiliation(s)
- Norihide Maikusa
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and Sciences, The University of Tokyo, Tokyo, Japan.,Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yinghan Zhu
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and Sciences, The University of Tokyo, Tokyo, Japan
| | - Akiko Uematsu
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and Sciences, The University of Tokyo, Tokyo, Japan
| | - Ayumu Yamashita
- Brain Information Communication Research Laboratory Group, Advanced Telecommunications Research Institutes International, Kyoto, Japan
| | - Kousaku Saotome
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and Sciences, The University of Tokyo, Tokyo, Japan
| | - Naohiro Okada
- The International Research Center for Neurointelligence (WPI-IRCN), Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan.,Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kiyoto Kasai
- The International Research Center for Neurointelligence (WPI-IRCN), Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan.,Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,The University of Tokyo Center for Integrative Science of Human Behavior (CiSHuB), Tokyo, Japan.,The University of Tokyo Institute for Diversity Adaptation of Human Mind (UTIDAHM), Tokyo, Japan
| | - Kazuo Okanoya
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and Sciences, The University of Tokyo, Tokyo, Japan.,The International Research Center for Neurointelligence (WPI-IRCN), Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan.,The University of Tokyo Center for Integrative Science of Human Behavior (CiSHuB), Tokyo, Japan.,The University of Tokyo Institute for Diversity Adaptation of Human Mind (UTIDAHM), Tokyo, Japan
| | - Okito Yamashita
- Brain Information Communication Research Laboratory Group, Advanced Telecommunications Research Institutes International, Kyoto, Japan.,Center for Advanced Intelligence Project (AIP), RIKEN, Tokyo, Japan
| | - Saori C Tanaka
- Brain Information Communication Research Laboratory Group, Advanced Telecommunications Research Institutes International, Kyoto, Japan
| | - Shinsuke Koike
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and Sciences, The University of Tokyo, Tokyo, Japan.,The International Research Center for Neurointelligence (WPI-IRCN), Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan.,The University of Tokyo Center for Integrative Science of Human Behavior (CiSHuB), Tokyo, Japan.,The University of Tokyo Institute for Diversity Adaptation of Human Mind (UTIDAHM), Tokyo, Japan
| |
Collapse
|
413
|
Moreau CA, Raznahan A, Bellec P, Chakravarty M, Thompson PM, Jacquemont S. Dissecting autism and schizophrenia through neuroimaging genomics. Brain 2021; 144:1943-1957. [PMID: 33704401 PMCID: PMC8370419 DOI: 10.1093/brain/awab096] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/24/2020] [Accepted: 01/08/2021] [Indexed: 12/23/2022] Open
Abstract
Neuroimaging genomic studies of autism spectrum disorder and schizophrenia have mainly adopted a 'top-down' approach, beginning with the behavioural diagnosis, and moving down to intermediate brain phenotypes and underlying genetic factors. Advances in imaging and genomics have been successfully applied to increasingly large case-control studies. As opposed to diagnostic-first approaches, the bottom-up strategy begins at the level of molecular factors enabling the study of mechanisms related to biological risk, irrespective of diagnoses or clinical manifestations. The latter strategy has emerged from questions raised by top-down studies: why are mutations and brain phenotypes over-represented in individuals with a psychiatric diagnosis? Are they related to core symptoms of the disease or to comorbidities? Why are mutations and brain phenotypes associated with several psychiatric diagnoses? Do they impact a single dimension contributing to all diagnoses? In this review, we aimed at summarizing imaging genomic findings in autism and schizophrenia as well as neuropsychiatric variants associated with these conditions. Top-down studies of autism and schizophrenia identified patterns of neuroimaging alterations with small effect-sizes and an extreme polygenic architecture. Genomic variants and neuroimaging patterns are shared across diagnostic categories suggesting pleiotropic mechanisms at the molecular and brain network levels. Although the field is gaining traction; characterizing increasingly reproducible results, it is unlikely that top-down approaches alone will be able to disentangle mechanisms involved in autism or schizophrenia. In stark contrast with top-down approaches, bottom-up studies showed that the effect-sizes of high-risk neuropsychiatric mutations are equally large for neuroimaging and behavioural traits. Low specificity has been perplexing with studies showing that broad classes of genomic variants affect a similar range of behavioural and cognitive dimensions, which may be consistent with the highly polygenic architecture of psychiatric conditions. The surprisingly discordant effect sizes observed between genetic and diagnostic first approaches underscore the necessity to decompose the heterogeneity hindering case-control studies in idiopathic conditions. We propose a systematic investigation across a broad spectrum of neuropsychiatric variants to identify putative latent dimensions underlying idiopathic conditions. Gene expression data on temporal, spatial and cell type organization in the brain have also considerable potential for parsing the mechanisms contributing to these dimensions' phenotypes. While large neuroimaging genomic datasets are now available in unselected populations, there is an urgent need for data on individuals with a range of psychiatric symptoms and high-risk genomic variants. Such efforts together with more standardized methods will improve mechanistically informed predictive modelling for diagnosis and clinical outcomes.
Collapse
Affiliation(s)
- Clara A Moreau
- Sainte Justine Research Center, University of Montréal, Montréal, Québec H3T 1C5, Canada
- Centre de Recherche de l’Institut Universitaire de Gériatrie de Montréal, Montreal, Québec H3W 1W5, Canada
- Human Genetics and Cognitive Functions, CNRS UMR 3571, Université de Paris, Institut Pasteur, Paris, France
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health Intramural Research Program, Bethesda, MD 20892, USA
| | - Pierre Bellec
- Centre de Recherche de l’Institut Universitaire de Gériatrie de Montréal, Montreal, Québec H3W 1W5, Canada
| | - Mallar Chakravarty
- Cerebral Imaging Centre, Douglas Hospital Mental Health University Institute, Verdun, Québec H4H 1R3, Canada
| | - Paul M Thompson
- Imaging Genetics Center, Stevens Institute for Neuroimaging and Informatics, USC Keck School of Medicine, Marina del Rey, CA 90033, USA
| | - Sebastien Jacquemont
- Sainte Justine Research Center, University of Montréal, Montréal, Québec H3T 1C5, Canada
| |
Collapse
|
414
|
Campos AI, Thompson PM, Veltman DJ, Pozzi E, van Veltzen LS, Jahanshad N, Adams MJ, Baune BT, Berger K, Brosch K, Bülow R, Connolly CG, Dannlowski U, Davey CG, de Zubicaray GI, Dima D, Erwin-Grabner T, Evans JW, Fu CHY, Gotlib IH, Goya-Maldonado R, Grabe HJ, Grotegerd D, Harris MA, Harrison BJ, Hatton SN, Hermesdorf M, Hickie IB, Ho TC, Kircher T, Krug A, Lagopoulos J, Lemke H, McMahon K, MacMaster FP, Martin NG, McIntosh AM, Medland SE, Meinert S, Meller T, Nenadic I, Opel N, Redlich R, Reneman L, Repple J, Sacchet MD, Schmitt S, Schrantee A, Sim K, Singh A, Stein F, Strike LT, van der Wee NJA, van der Werff SJA, Völzke H, Waltemate L, Whalley HC, Wittfeld K, Wright MJ, Yang TT, Zarate CA, Schmaal L, Rentería ME. Brain Correlates of Suicide Attempt in 18,925 Participants Across 18 International Cohorts. Biol Psychiatry 2021; 90:243-252. [PMID: 34172278 PMCID: PMC8324512 DOI: 10.1016/j.biopsych.2021.03.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 03/12/2021] [Accepted: 03/13/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Neuroimaging studies of suicidal behavior have so far been conducted in small samples, prone to biases and false-positive associations, yielding inconsistent results. The ENIGMA-MDD Working Group aims to address the issues of poor replicability and comparability by coordinating harmonized analyses across neuroimaging studies of major depressive disorder and related phenotypes, including suicidal behavior. METHODS Here, we pooled data from 18 international cohorts with neuroimaging and clinical measurements in 18,925 participants (12,477 healthy control subjects and 6448 people with depression, of whom 694 had attempted suicide). We compared regional cortical thickness and surface area and measures of subcortical, lateral ventricular, and intracranial volumes between suicide attempters, clinical control subjects (nonattempters with depression), and healthy control subjects. RESULTS We identified 25 regions of interest with statistically significant (false discovery rate < .05) differences between groups. Post hoc examinations identified neuroimaging markers associated with suicide attempt including smaller volumes of the left and right thalamus and the right pallidum and lower surface area of the left inferior parietal lobe. CONCLUSIONS This study addresses the lack of replicability and consistency in several previously published neuroimaging studies of suicide attempt and further demonstrates the need for well-powered samples and collaborative efforts. Our results highlight the potential involvement of the thalamus, a structure viewed historically as a passive gateway in the brain, and the pallidum, a region linked to reward response and positive affect. Future functional and connectivity studies of suicidal behaviors may focus on understanding how these regions relate to the neurobiological mechanisms of suicide attempt risk.
Collapse
Affiliation(s)
- Adrian I Campos
- Genetic Epidemiology Lab, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Paul M Thompson
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, California
| | - Dick J Veltman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Psychiatry, Department of Anatomy & Neurosciences, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Elena Pozzi
- Department of Psychiatry, The University of Melbourne & Melbourne Health, Melbourne, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, Australia
| | - Laura S van Veltzen
- Department of Psychiatry, The University of Melbourne & Melbourne Health, Melbourne, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, Australia
| | - Neda Jahanshad
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, California
| | - Mark J Adams
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Bernhard T Baune
- Department of Psychiatry, The University of Melbourne, Melbourne, Victoria, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia; Department of Psychiatry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Katharina Brosch
- Department of Psychiatry, Philipps-University Marburg, Marburg, Hesse, Germany
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Mecklenburg-Vorpommern, Germany
| | - Colm G Connolly
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida
| | - Udo Dannlowski
- Department of Psychiatry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Christopher G Davey
- Department of Psychiatry, The University of Melbourne & Melbourne Health, Melbourne, Victoria, Australia
| | - Greig I de Zubicaray
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Danai Dima
- Department of Psychology, School of Arts and Social Sciences, City, University of London, London, United Kingdom; Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Tracy Erwin-Grabner
- Laboratory of Systems Neuroscience and Imaging in Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Lower Saxony, Germany
| | - Jennifer W Evans
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Cynthia H Y Fu
- Centre for Affective Disorders, Institute of Psychology, Psychiatry and Neuroscience, King's College London, London, United Kingdom; School of Psychology, University of East London, London, United Kingdom
| | - Ian H Gotlib
- Department of Psychology, Stanford University, Stanford, California
| | - Roberto Goya-Maldonado
- Laboratory of Systems Neuroscience and Imaging in Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Lower Saxony, Germany
| | - Hans J Grabe
- German Center for Neurodegenerative Disease, Greifswald, Mecklenburg-Vorpommern, Germany
| | - Dominik Grotegerd
- Department of Psychiatry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Matthew A Harris
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Ben J Harrison
- Department of Psychiatry, The University of Melbourne & Melbourne Health, Melbourne, Victoria, Australia
| | - Sean N Hatton
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Marco Hermesdorf
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Ian B Hickie
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Tiffany C Ho
- Department of Psychiatry & Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California
| | - Tilo Kircher
- Department of Psychiatry, Philipps-University Marburg, Marburg, Hesse, Germany
| | - Axel Krug
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, North Rhine-Westphalia, Germany; Department of Psychiatry, Philipps-University Marburg, Marburg, Hesse, Germany
| | - Jim Lagopoulos
- Thompson Institute, University of the Sunshine Coast, Sunshine Coast, Queensland, Australia; Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Hannah Lemke
- Department of Psychiatry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Katie McMahon
- Herston Imaging Research Facility & School of Clinical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Frank P MacMaster
- Department of Pediatrics and Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Strategic Clinical Network for Addictions and Mental Health, Alberta Health Services, Calgary, Alberta, Canada
| | - Nicholas G Martin
- Genetic Epidemiology Lab, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Andrew M McIntosh
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Sarah E Medland
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia; Psychiatric Genetics Lab, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Austalia; School of Psychology, The University of Queensland, Brisbane, Queensland, Australia
| | - Susanne Meinert
- Department of Psychiatry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Tina Meller
- Department of Psychiatry, Philipps-University Marburg, Marburg, Hesse, Germany
| | - Igor Nenadic
- Department of Psychiatry, Philipps-University Marburg, Marburg, Hesse, Germany
| | - Nils Opel
- Department of Psychiatry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Ronny Redlich
- Department of Psychiatry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Liesbeth Reneman
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Jonathan Repple
- Department of Psychiatry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Matthew D Sacchet
- Center for Depression, Anxiety, and Stress Research, McLean Hospital, Harvard Medical School, Belmont, Massachusetts
| | - Simon Schmitt
- Department of Psychiatry, Philipps-University Marburg, Marburg, Hesse, Germany
| | - Anouk Schrantee
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Kang Sim
- West Region, Institute of Mental Health, Buangkok View, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Aditya Singh
- Laboratory of Systems Neuroscience and Imaging in Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Lower Saxony, Germany
| | - Frederike Stein
- Department of Psychiatry, Philipps-University Marburg, Marburg, Hesse, Germany
| | - Lachlan T Strike
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Nic J A van der Wee
- Department of Psychiatry, Leiden University Medical Center, Leiden, the Netherlands; Leiden Institute for Brain and Cognition, Leiden, the Netherlands
| | - Steven J A van der Werff
- Department of Psychiatry, Leiden University Medical Center, Leiden, the Netherlands; Leiden Institute for Brain and Cognition, Leiden, the Netherlands
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Mecklenburg-Vorpommern, Germany
| | - Lena Waltemate
- Department of Psychiatry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Heather C Whalley
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Katharina Wittfeld
- German Center for Neurodegenerative Disease, Greifswald, Mecklenburg-Vorpommern, Germany
| | - Margaret J Wright
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia; Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland, Australia
| | - Tony T Yang
- Department of Psychiatry & Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Lianne Schmaal
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, Australia.
| | - Miguel E Rentería
- Genetic Epidemiology Lab, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
415
|
Qiu X, Xu W, Zhang R, Yan W, Ma W, Xie S, Zhou M. Regional Homogeneity Brain Alterations in Schizophrenia: An Activation Likelihood Estimation Meta-Analysis. Psychiatry Investig 2021; 18:709-717. [PMID: 34333896 PMCID: PMC8390947 DOI: 10.30773/pi.2021.0062] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/24/2021] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Resting state functional magnetic resonance imaging (rsfMRI) provides a lot of evidence for local abnormal brain activity in schizophrenia, but the results are not consistent. Our aim is to find out the consistent abnormal brain regions of the patients with schizophrenia by using regional homogeneity (ReHo), and indirectly understand the degree of brain damage of the patients with drug-naive first episode schizophrenia (Dn-FES) and chronic schizophrenia. METHODS We performed the experiment by activation likelihood estimation (ALE) software to analysis the differences between people with schizophrenia group (all schizophrenia group and chronic schizophrenia group) and healthy controls. RESULTS Thirteen functional imaging studies were included in quantitative meta-analysis. All schizophrenia group showed decreased ReHo in bilateral precentral gyrus (PreCG) and left middle occipital gyrus (MOG), and increased ReHo in bilateral superior frontal gyrus (SFG) and right insula. Chronic schizophrenia group showed decreased ReHo in bilateral MOG, right fusiform gyrus, left PreCG, left cerebellum, right precuneus, left medial frontal gyrus and left anterior cingulate cortex (ACC). No significant increased brain areas were found in patients with chronic schizophrenia. CONCLUSION Our findings suggest that patients with chronic schizophrenia have more extensive brain damage than FES, which may contribute to our understanding of the progressive pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Xiaolei Qiu
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Wenwen Xu
- Department of Neurology, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Rongrong Zhang
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Yan
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Wenying Ma
- Department of Neurology, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Shiping Xie
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Min Zhou
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
416
|
Wang X, Xie H, Chen T, Cotton AS, Salminen LE, Logue MW, Clarke-Rubright EK, Wall J, Dennis EL, O'Leary BM, Abdallah CG, Andrew E, Baugh LA, Bomyea J, Bruce SE, Bryant R, Choi K, Daniels JK, Davenport ND, Davidson RJ, DeBellis M, deRoon-Cassini T, Disner SG, Fani N, Fercho KA, Fitzgerald J, Forster GL, Frijling JL, Geuze E, Gomaa H, Gordon EM, Grupe D, Harpaz-Rotem I, Haswell CC, Herzog JI, Hofmann D, Hollifield M, Hosseini B, Hudson AR, Ipser J, Jahanshad N, Jovanovic T, Kaufman ML, King AP, Koch SBJ, Koerte IK, Korgaonkar MS, Krystal JH, Larson C, Lebois LAM, Levy I, Li G, Magnotta VA, Manthey A, May G, McLaughlin KA, Mueller SC, Nawijn L, Nelson SM, Neria Y, Nitschke JB, Olff M, Olson EA, Peverill M, Phan KL, Rashid FM, Ressler K, Rosso IM, Sambrook K, Schmahl C, Shenton ME, Sierk A, Simons JS, Simons RM, Sponheim SR, Stein MB, Stein DJ, Stevens JS, Straube T, Suarez-Jimenez B, Tamburrino M, Thomopoulos SI, van der Wee NJA, van der Werff SJA, van Erp TGM, van Rooij SJH, van Zuiden M, Varkevisser T, Veltman DJ, Vermeiren RRJM, Walter H, Wang L, Zhu Y, Zhu X, Thompson PM, Morey RA, Liberzon I. Cortical volume abnormalities in posttraumatic stress disorder: an ENIGMA-psychiatric genomics consortium PTSD workgroup mega-analysis. Mol Psychiatry 2021; 26:4331-4343. [PMID: 33288872 PMCID: PMC8180531 DOI: 10.1038/s41380-020-00967-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 10/29/2020] [Accepted: 11/18/2020] [Indexed: 01/31/2023]
Abstract
Studies of posttraumatic stress disorder (PTSD) report volume abnormalities in multiple regions of the cerebral cortex. However, findings for many regions, particularly regions outside commonly studied emotion-related prefrontal, insular, and limbic regions, are inconsistent and tentative. Also, few studies address the possibility that PTSD abnormalities may be confounded by comorbid depression. A mega-analysis investigating all cortical regions in a large sample of PTSD and control subjects can potentially provide new insight into these issues. Given this perspective, our group aggregated regional volumes data of 68 cortical regions across both hemispheres from 1379 PTSD patients to 2192 controls without PTSD after data were processed by 32 international laboratories using ENIGMA standardized procedures. We examined whether regional cortical volumes were different in PTSD vs. controls, were associated with posttraumatic stress symptom (PTSS) severity, or were affected by comorbid depression. Volumes of left and right lateral orbitofrontal gyri (LOFG), left superior temporal gyrus, and right insular, lingual and superior parietal gyri were significantly smaller, on average, in PTSD patients than controls (standardized coefficients = -0.111 to -0.068, FDR corrected P values < 0.039) and were significantly negatively correlated with PTSS severity. After adjusting for depression symptoms, the PTSD findings in left and right LOFG remained significant. These findings indicate that cortical volumes in PTSD patients are smaller in prefrontal regulatory regions, as well as in broader emotion and sensory processing cortical regions.
Collapse
Affiliation(s)
- Xin Wang
- Department of Psychiatry, University of Toledo, Toledo, OH, USA.
| | - Hong Xie
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Tian Chen
- Department of Mathematics and Statistics, University of Toledo, Toledo, OH, USA
| | - Andrew S Cotton
- Department of Psychiatry, University of Toledo, Toledo, OH, USA
| | - Lauren E Salminen
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA, USA
| | - Mark W Logue
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Emily K Clarke-Rubright
- Brain Imaging and Analysis Center, Duke University, Durham, NC, USA
- VISN 6 MIRECC, Durham VA Health Care System, Durham, NC, USA
| | - John Wall
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Emily L Dennis
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA, USA
- Department of Neurology, University of Utah, Salt Lake City, UT, USA
| | - Brian M O'Leary
- Department of Psychiatry, University of Toledo, Toledo, OH, USA
| | - Chadi G Abdallah
- Clinical Neuroscience Division, National Center for PTSD, VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | | | - Lee A Baugh
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
- Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
- Sioux Falls VA Health Care System, Sioux Falls, SD, USA
| | - Jessica Bomyea
- Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Steven E Bruce
- Center for Trauma Recovery, Department of Psychological Sciences, University of Missouri-St. Louis, St. Louis, MO, USA
| | - Richard Bryant
- School of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - Kyle Choi
- Health Services Research Center, University of California, San Diego, La Jolla, CA, USA
| | - Judith K Daniels
- Department of Clinical Psychology, University of Groningen, Groningen, The Netherlands
| | - Nicholas D Davenport
- Minneapolis VA Health Care System, Minneapolis, MN, USA
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Richard J Davidson
- Center for Healthy Minds, University of Wisconsin-Madison, Madison, WI, USA
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, USA
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael DeBellis
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Terri deRoon-Cassini
- Department of Surgery, Division of Trauma & Acute Care Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Seth G Disner
- Minneapolis VA Health Care System, Minneapolis, MN, USA
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Negar Fani
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Kelene A Fercho
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
- Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
- Sioux Falls VA Health Care System, Sioux Falls, SD, USA
- Civil Aerospace Medical Institute, US Federal Aviation Administration, Oklahoma City, OK, USA
| | | | - Gina L Forster
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
- Brain Health Research Centre, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Jessie L Frijling
- Department of Psychiatry, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Elbert Geuze
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
- Brain Research and Innovation Centre, Ministry of Defence, Utrecht, The Netherlands
| | - Hassaan Gomaa
- Department of Psychiatry and Behavioral Health, Penn State College of Medicine, Hershey, PA, USA
| | - Evan M Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Dan Grupe
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, USA
| | - Ilan Harpaz-Rotem
- Clinical Neuroscience Division, National Center for PTSD, VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Courtney C Haswell
- Brain Imaging and Analysis Center, Duke University, Durham, NC, USA
- VISN 6 MIRECC, Durham VA Health Care System, Durham, NC, USA
| | - Julia I Herzog
- Department of Psychosomatic Medicine and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - David Hofmann
- Institute of Medical Psychology and Systems Neuroscience, University of Münster, Münster, Germany
| | - Michael Hollifield
- Program for Traumatic Stress, Tibor Rubin VA Medical Center, Long Beach, CA, USA
| | - Bobak Hosseini
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Anna R Hudson
- Department of Experimental Clinical and Health Psychology, Ghent University, Ghent, Belgium
| | - Jonathan Ipser
- Department of Psychiatry, University of Cape Town, Cape Town, South Africa
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA, USA
| | - Tanja Jovanovic
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University, Detroit, MI, USA
| | - Milissa L Kaufman
- Division of Women's Mental Health, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Anthony P King
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Saskia B J Koch
- Department of Psychiatry, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
- Donders Institute for Brain, Cognition and Behavior, Centre for Cognitive Neuroimaging, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Inga K Koerte
- Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
| | - Mayuresh S Korgaonkar
- Brain Dynamics Centre, Westmead Institute of Medical Research, University of Sydney, Westmead, NSW, Australia
| | - John H Krystal
- Clinical Neuroscience Division, National Center for PTSD, VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Christine Larson
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Lauren A M Lebois
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, MA, USA
| | - Ifat Levy
- Clinical Neuroscience Division, National Center for PTSD, VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Gen Li
- Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Vincent A Magnotta
- Departments of Radiology, Psychiatry, and Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Antje Manthey
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Geoffrey May
- VISN 17 Center of Excellence for Research on Returning War Veterans, Doris Miller VA Medical Center, Waco, TX, USA
- Center for Vital Longevity, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, USA
- Department of Psychiatry and Behavioral Science, Texas A&M University College of Medicine, College Station, TX, USA
| | | | - Sven C Mueller
- Department of Experimental Clinical and Health Psychology, Ghent University, Ghent, Belgium
- Department of Personality, Psychological Assessment and Treatment, University of Deusto, Bilbao, Spain
| | - Laura Nawijn
- Department of Psychiatry, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
- Department of Psychiatry, Amsterdam University Medical Centers, Location VU University Medical Center, VU University, Amsterdam, The Netherlands
| | - Steven M Nelson
- VISN 17 Center of Excellence for Research on Returning War Veterans, Doris Miller VA Medical Center, Waco, TX, USA
- Center for Vital Longevity, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, USA
| | - Yuval Neria
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Jack B Nitschke
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Miranda Olff
- Department of Psychiatry, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
- ARQ National Psychotrauma Centrum, Diemen, The Netherlands
| | - Elizabeth A Olson
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Depression, Anxiety, and Stress Research, McLean Hospital, Belmont, MA, USA
| | - Matthew Peverill
- Department of Psychology, University of Washington, Seattle, WA, USA
| | - K Luan Phan
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
- The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Mental Health Service Line, Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Faisal M Rashid
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA, USA
| | - Kerry Ressler
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, MA, USA
| | - Isabelle M Rosso
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Depression, Anxiety, and Stress Research, McLean Hospital, Belmont, MA, USA
| | - Kelly Sambrook
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Christian Schmahl
- Department of Psychosomatic Medicine and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Department of Psychiatry, University of Western Ontario, London, ON, Canada
| | - Martha E Shenton
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- Department of Psychiatry, VA Boston Healthcare System, Brockton, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Anika Sierk
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Institute of Cognitive Neuroscience, University College London, London, United Kingdom
| | - Jeffrey S Simons
- Sioux Falls VA Health Care System, Sioux Falls, SD, USA
- Department of Psychology, University of South Dakota, Vermillion, SD, USA
| | - Raluca M Simons
- Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
- Department of Psychology, University of South Dakota, Vermillion, SD, USA
| | - Scott R Sponheim
- Minneapolis VA Health Care System, Minneapolis, MN, USA
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Murray B Stein
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Department of Family Medicine and Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Dan J Stein
- SAMRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Jennifer S Stevens
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Thomas Straube
- Institute of Medical Psychology and Systems Neuroscience, University of Münster, Münster, Germany
| | - Benjamin Suarez-Jimenez
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | | | - Sophia I Thomopoulos
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA, USA
| | - Nic J A van der Wee
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Institute for Brain and Cognition, Leiden, The Netherlands
| | - Steven J A van der Werff
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Institute for Brain and Cognition, Leiden, The Netherlands
| | - Theo G M van Erp
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, USA
| | - Sanne J H van Rooij
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Mirjam van Zuiden
- Department of Psychiatry, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Tim Varkevisser
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
- Brain Research and Innovation Centre, Ministry of Defence, Utrecht, The Netherlands
| | - Dick J Veltman
- Department of Psychiatry, Amsterdam University Medical Centers, Location VU University Medical Center, VU University, Amsterdam, The Netherlands
| | - Robert R J M Vermeiren
- Child and Adolescent Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
- Youz-Parnassia Group, Leiden, The Netherlands
| | - Henrik Walter
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Li Wang
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
- Laboratory for Traumatic Stress Studies, CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Ye Zhu
- Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Xi Zhu
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of the University of Southern California, Marina del Rey, CA, USA
| | - Rajendra A Morey
- Brain Imaging and Analysis Center, Duke University, Durham, NC, USA
- VISN 6 MIRECC, Durham VA Health Care System, Durham, NC, USA
| | - Israel Liberzon
- Department of Psychiatry and Behavioral Science, Texas A&M University College of Medicine, College Station, TX, USA
| |
Collapse
|
417
|
Jiang Y, Wang Y, Huang H, He H, Tang Y, Su W, Xu L, Wei Y, Zhang T, Hu H, Wang J, Yao D, Wang J, Luo C. Antipsychotics Effects on Network-Level Reconfiguration of Cortical Morphometry in First-Episode Schizophrenia. Schizophr Bull 2021; 48:231-240. [PMID: 34313782 PMCID: PMC8781340 DOI: 10.1093/schbul/sbab082] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Cortical thickness reductions are evident in schizophrenia (SZ). Associations between antipsychotic medications (APMs) and cortical morphometry have been explored in SZ patients. This raises the question of whether the reconfiguration of morphological architecture by APM plays potential compensatory roles for abnormalities in the cerebral cortex. Structural magnetic resonance imaging was obtained from 127 medication-naive first-episode SZ patients and 133 matched healthy controls. Patients received 12 weeks of APM and were categorized as responders (n = 75) or nonresponders (NRs, n = 52) at follow-up. Using surface-based morphometry and structural covariance (SC) analysis, this study investigated the short-term effects of antipsychotics on cortical thickness and cortico-cortical covariance. Global efficiency was computed to characterize network integration of the large-scale structural connectome. The relationship between covariance and cortical thinning was examined by SC analysis among the top-n regions with thickness reduction. Widespread cortical thickness reductions were observed in pre-APM patients. Post-APM patients showed more reductions in cortical thickness, even in the frontotemporal regions without baseline reductions. Covariance analysis revealed strong cortico-cortical covariance and higher network integration in responders than in NRs. For the NRs, some of the prefrontal and temporal nodes were not covariant between the top-n regions with cortical thickness reduction. Antipsychotic effects are not restricted to a single brain region but rather exhibit a network-level covariance pattern. Neuroimaging connectomics highlights the positive effects of antipsychotics on the reconfiguration of brain architecture, suggesting that abnormalities in regional morphology may be compensated by increasing interregional covariance when symptoms are controlled by antipsychotics.
Collapse
Affiliation(s)
- Yuchao Jiang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, PR China,High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Yingchan Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Huan Huang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, PR China,High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Hui He
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Yingying Tang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Wenjun Su
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Lihua Xu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yanyan Wei
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Tianhong Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hao Hu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jinhong Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Dezhong Yao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, PR China,Research Unit of NeuroInformation (2019RU035), Chinese Academy of Medical Sciences, Chengdu, PR China
| | - Jijun Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China,CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Science, Shanghai, PR China,Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, PR China
| | - Cheng Luo
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, PR China,High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China,Research Unit of NeuroInformation (2019RU035), Chinese Academy of Medical Sciences, Chengdu, PR China,To whom correspondence should be addressed; University of Electronic Science and Technology of China, Second North Jianshe Road, Chengdu 610054, PR China; tel: 86-28-83201018, fax: 86-28-83208238, e-mail:
| |
Collapse
|
418
|
Zhou Z, Wang K, Tang J, Wei D, Song L, Peng Y, Fu Y, Qiu J. Cortical thickness distinguishes between major depression and schizophrenia in adolescents. BMC Psychiatry 2021; 21:361. [PMID: 34284747 PMCID: PMC8293570 DOI: 10.1186/s12888-021-03373-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 07/08/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Early diagnosis of adolescent psychiatric disorder is crucial for early intervention. However, there is extensive comorbidity between affective and psychotic disorders, which increases the difficulty of precise diagnoses among adolescents. METHODS We obtained structural magnetic resonance imaging scans from 150 adolescents, including 67 and 47 patients with major depressive disorder (MDD) and schizophrenia (SCZ), as well as 34 healthy controls (HC) to explore whether psychiatric disorders could be identified using a machine learning technique. Specifically, we used the support vector machine and the leave-one-out cross-validation method to distinguish among adolescents with MDD and SCZ and healthy controls. RESULTS We found that cortical thickness was a classification feature of a) MDD and HC with 79.21% accuracy where the temporal pole had the highest weight; b) SCZ and HC with 69.88% accuracy where the left superior temporal sulcus had the highest weight. Notably, adolescents with MDD and SCZ could be classified with 62.93% accuracy where the right pars triangularis had the highest weight. CONCLUSIONS Our findings suggest that cortical thickness may be a critical biological feature in the diagnosis of adolescent psychiatric disorders. These findings might be helpful to establish an early prediction model for adolescents to better diagnose psychiatric disorders.
Collapse
Affiliation(s)
- Zheyi Zhou
- grid.419897.a0000 0004 0369 313XKey Laboratory of Cognition and Personality (SWU), Ministry of Education, Chongqing, 400715 China ,grid.263906.8Faculty of Psychology, Southwest University, No.2 Tiansheng Road, Beibei District, Chongqing, 400715 China
| | - Kangcheng Wang
- grid.410585.d0000 0001 0495 1805Faculty of Psychology, Shandong Normal University, Jinan, 250014 Shandong China
| | - Jinxiang Tang
- grid.452206.7Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, No.1, Yixueyuan Road, Yuzhong District, Chongqing, 400016 China ,Sleep and Psychology Center, The Bishan Hospital of Chongqing, Chongqing, 402760 China
| | - Dongtao Wei
- grid.419897.a0000 0004 0369 313XKey Laboratory of Cognition and Personality (SWU), Ministry of Education, Chongqing, 400715 China ,grid.263906.8Faculty of Psychology, Southwest University, No.2 Tiansheng Road, Beibei District, Chongqing, 400715 China
| | - Li Song
- grid.419897.a0000 0004 0369 313XKey Laboratory of Cognition and Personality (SWU), Ministry of Education, Chongqing, 400715 China ,grid.263906.8Faculty of Psychology, Southwest University, No.2 Tiansheng Road, Beibei District, Chongqing, 400715 China
| | - Yadong Peng
- grid.452206.7Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, No.1, Yixueyuan Road, Yuzhong District, Chongqing, 400016 China ,Department of Psychology, Chongqing Health Center for Women and Children, Chongqing, 401147 China
| | - Yixiao Fu
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, No.1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Jiang Qiu
- Key Laboratory of Cognition and Personality (SWU), Ministry of Education, Chongqing, 400715, China. .,Faculty of Psychology, Southwest University, No.2 Tiansheng Road, Beibei District, Chongqing, 400715, China. .,Collaborative Innovation Center of Assessment Toward Basic Education Quality, Southwest University Branch, Beijing Normal University, Beijing, 100875, China.
| |
Collapse
|
419
|
Zhu X, Ward J, Cullen B, Lyall DM, Strawbridge RJ, Lyall LM, Smith DJ. Phenotypic and genetic associations between anhedonia and brain structure in UK Biobank. Transl Psychiatry 2021; 11:395. [PMID: 34282121 PMCID: PMC8289859 DOI: 10.1038/s41398-021-01522-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023] Open
Abstract
Anhedonia is a core symptom of multiple psychiatric disorders and has been associated with alterations in brain structure. Genome-wide association studies suggest that anhedonia is heritable, with a polygenic architecture, but few studies have explored the association between genetic loading for anhedonia-indexed by polygenic risk scores for anhedonia (PRS-anhedonia)-and structural brain imaging phenotypes. Here, we investigated how anhedonia and PRS-anhedonia were associated with brain structure within the UK Biobank cohort. Brain measures (including total grey/white matter volumes, subcortical volumes, cortical thickness (CT) and white matter integrity) were analysed using linear mixed models in relation to anhedonia and PRS-anhedonia in 19,592 participants (9225 males; mean age = 62.6 years, SD = 7.44). We found that state anhedonia was significantly associated with reduced total grey matter volume (GMV); increased total white matter volume (WMV); smaller volumes in thalamus and nucleus accumbens; reduced CT within the paracentral cortex, the opercular part of inferior frontal gyrus, precentral cortex, insula and rostral anterior cingulate cortex; and poorer integrity of many white matter tracts. PRS-anhedonia was associated with reduced total GMV; increased total WMV; reduced white matter integrity; and reduced CT within the parahippocampal cortex, superior temporal gyrus and insula. Overall, both state anhedonia and PRS-anhedonia were associated with individual differences in multiple brain structures, including within reward-related circuits. These associations may represent vulnerability markers for psychopathology relevant to a range of psychiatric disorders.
Collapse
Affiliation(s)
- Xingxing Zhu
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK.
| | - Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Breda Cullen
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Donald M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Rona J Strawbridge
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Health Data Research (HDR), Glasgow, UK
| | - Laura M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Daniel J Smith
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
- Division of Psychiatry, Kennedy Tower, Royal Edinburgh Hospital, Edinburgh, UK
| |
Collapse
|
420
|
Reduced cortical thickness of the paracentral lobule in at-risk mental state individuals with poor 1-year functional outcomes. Transl Psychiatry 2021; 11:396. [PMID: 34282119 PMCID: PMC8289863 DOI: 10.1038/s41398-021-01516-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/26/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
Although widespread cortical thinning centered on the fronto-temporal regions in schizophrenia has been reported, the findings in at-risk mental state (ARMS) patients have been inconsistent. In addition, it remains unclear whether abnormalities of cortical thickness (CT) in ARMS individuals, if present, are related to their functional decline irrespective of future psychosis onset. In this multicenter study in Japan, T1-weighted magnetic resonance imaging was performed at baseline in 107 individuals with ARMS, who were subdivided into resilient (77, good functional outcome) and non-resilient (13, poor functional outcome) groups based on the change in Global Assessment of Functioning scores during 1-year follow-up, and 104 age- and sex-matched healthy controls recruited at four scanning sites. We measured the CT of the entire cortex and performed group comparisons using FreeSurfer software. The relationship between the CT and cognitive functioning was examined in an ARMS subsample (n = 70). ARMS individuals as a whole relative to healthy controls exhibited a significantly reduced CT, predominantly in the fronto-temporal regions, which was partly associated with cognitive impairments, and an increased CT in the left parietal and right occipital regions. Compared with resilient ARMS individuals, non-resilient ARMS individuals exhibited a significantly reduced CT of the right paracentral lobule. These findings suggest that ARMS individuals partly share CT abnormalities with patients with overt schizophrenia, potentially representing general vulnerability to psychopathology, and also support the role of cortical thinning in the paracentral lobule as a predictive biomarker for short-term functional decline in the ARMS population.
Collapse
|
421
|
Ye H, Zalesky A, Lv J, Loi SM, Cetin-Karayumak S, Rathi Y, Tian Y, Pantelis C, Di Biase MA. Network Analysis of Symptom Comorbidity in Schizophrenia: Relationship to Illness Course and Brain White Matter Microstructure. Schizophr Bull 2021; 47:1156-1167. [PMID: 33693887 PMCID: PMC8266579 DOI: 10.1093/schbul/sbab015] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
INTRODUCTION Recent network-based analyses suggest that schizophrenia symptoms are intricately connected and interdependent, such that central symptoms can activate adjacent symptoms and increase global symptom burden. Here, we sought to identify key clinical and neurobiological factors that relate to symptom organization in established schizophrenia. METHODS A symptom comorbidity network was mapped for a broad constellation of symptoms measured in 642 individuals with a schizophrenia-spectrum disorder. Centrality analyses were used to identify hub symptoms. The extent to which each patient's symptoms formed clusters in the comorbidity network was quantified with cluster analysis and used to predict (1) clinical features, including illness duration and psychosis (positive symptom) severity and (2) brain white matter microstructure, indexed by the fractional anisotropy (FA), in a subset (n = 296) of individuals with diffusion-weighted imaging (DWI) data. RESULTS Global functioning, substance use, and blunted affect were the most central symptoms within the symptom comorbidity network. Symptom profiles for some patients formed highly interconnected clusters, whereas other patients displayed unrelated and disconnected symptoms. Stronger clustering among an individual's symptoms was significantly associated with shorter illness duration (t = 2.7; P = .0074), greater psychosis severity (ie, positive symptoms expression) (t = -5.5; P < 0.0001) and lower fractional anisotropy in fibers traversing the cortico-cerebellar-thalamic-cortical circuit (r = .59, P < 0.05). CONCLUSION Symptom network structure varies over the course of schizophrenia: symptom interactions weaken with increasing illness duration and strengthen during periods of high positive symptom expression. Reduced white matter coherence relates to stronger symptom clustering, and thus, may underlie symptom cascades and global symptomatic burden in individuals with schizophrenia.
Collapse
Affiliation(s)
- Hua Ye
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, Victoria, Australia
| | - Andrew Zalesky
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, Victoria, Australia
- Melbourne School of Engineering, The University of Melbourne, Parkville, Victoria, Australia
| | - Jinglei Lv
- School of Biomedical Engineering & Brain and Mind Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Samantha M Loi
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, Victoria, Australia
| | | | - Yogesh Rathi
- Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Ye Tian
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, Victoria, Australia
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, Victoria, Australia
| | - Maria A Di Biase
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, Victoria, Australia
- Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
422
|
Yamaguchi H, Hashimoto Y, Sugihara G, Miyata J, Murai T, Takahashi H, Honda M, Hishimoto A, Yamashita Y. Three-Dimensional Convolutional Autoencoder Extracts Features of Structural Brain Images With a "Diagnostic Label-Free" Approach: Application to Schizophrenia Datasets. Front Neurosci 2021; 15:652987. [PMID: 34305514 PMCID: PMC8294943 DOI: 10.3389/fnins.2021.652987] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/07/2021] [Indexed: 01/17/2023] Open
Abstract
There has been increasing interest in performing psychiatric brain imaging studies using deep learning. However, most studies in this field disregard three-dimensional (3D) spatial information and targeted disease discrimination, without considering the genetic and clinical heterogeneity of psychiatric disorders. The purpose of this study was to investigate the efficacy of a 3D convolutional autoencoder (3D-CAE) for extracting features related to psychiatric disorders without diagnostic labels. The network was trained using a Kyoto University dataset including 82 patients with schizophrenia (SZ) and 90 healthy subjects (HS) and was evaluated using Center for Biomedical Research Excellence (COBRE) datasets, including 71 SZ patients and 71 HS. We created 16 3D-CAE models with different channels and convolutions to explore the effective range of hyperparameters for psychiatric brain imaging. The number of blocks containing two convolutional layers and one pooling layer was set, ranging from 1 block to 4 blocks. The number of channels in the extraction layer varied from 1, 4, 16, and 32 channels. The proposed 3D-CAEs were successfully reproduced into 3D structural magnetic resonance imaging (MRI) scans with sufficiently low errors. In addition, the features extracted using 3D-CAE retained the relation to clinical information. We explored the appropriate hyperparameter range of 3D-CAE, and it was suggested that a model with 3 blocks may be related to extracting features for predicting the dose of medication and symptom severity in schizophrenia.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Information Medicine, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Tokyo, Japan.,Department of Psychiatry, School of Medicine, Yokohama City University, Yokohama, Japan
| | - Yuki Hashimoto
- Department of Information Medicine, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Tokyo, Japan
| | - Genichi Sugihara
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Jun Miyata
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshiya Murai
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidehiko Takahashi
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Manabu Honda
- Department of Information Medicine, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Tokyo, Japan
| | - Akitoyo Hishimoto
- Department of Psychiatry, School of Medicine, Yokohama City University, Yokohama, Japan
| | - Yuichi Yamashita
- Department of Information Medicine, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Tokyo, Japan
| |
Collapse
|
423
|
Associations between long-term psychosis risk, probabilistic category learning, and attenuated psychotic symptoms with cortical surface morphometry. Brain Imaging Behav 2021; 16:91-106. [PMID: 34218406 DOI: 10.1007/s11682-021-00479-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2021] [Indexed: 10/20/2022]
Abstract
Neuroimaging studies have consistently found structural cortical abnormalities in individuals with schizophrenia, especially in structural hubs. However, it is unclear what abnormalities predate psychosis onset and whether abnormalities are related to behavioral performance and symptoms associated with psychosis risk. Using surface-based morphometry, we examined cortical volume, gyrification, and thickness in a psychosis risk group at long-term risk for developing a psychotic disorder (n = 18; i.e., extreme positive schizotypy plus interview-rated attenuated psychotic symptoms [APS]) and control group (n = 19). Overall, the psychosis risk group exhibited cortical abnormalities in multiple structural hub regions, with abnormalities associated with poorer probabilistic category learning, a behavioral measure strongly associated with psychosis risk. For instance, the psychosis risk group had hypogyria in a right posterior midcingulate cortical hub and left superior parietal cortical hub, as well as decreased volume in a right pericalcarine hub. Morphometric measures in all of these regions were also associated with poorer probabilistic category learning. In addition to decreased right pericalcarine volume, the psychosis risk group exhibited a number of other structural abnormalities in visual network structural hub regions, consistent with previous evidence of visual perception deficits in psychosis risk. Further, severity of APS hallucinations, delusional ideation, and suspiciousness/persecutory ideas were associated with gyrification abnormalities, with all domains associated with hypogyria of the right lateral orbitofrontal cortex. Thus, current results suggest that structural abnormalities, especially in structural hubs, are present in psychosis risk and are associated both with poor learning on a psychosis risk-related task and with APS severity.
Collapse
|
424
|
Lubczyńska MJ, Muetzel RL, El Marroun H, Hoek G, Kooter IM, Thomson EM, Hillegers M, Vernooij MW, White T, Tiemeier H, Guxens M. Air pollution exposure during pregnancy and childhood and brain morphology in preadolescents. ENVIRONMENTAL RESEARCH 2021; 198:110446. [PMID: 33221303 DOI: 10.1016/j.envres.2020.110446] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Studies investigating the relationship between exposure to air pollution and brain development using magnetic resonance images are emerging. However, most studies have focused only on prenatal exposures, and have included a limited selection of pollutants. Here, we aim to expand the current knowledge by studying pregnancy and childhood exposure to a wide selection of pollutants, and brain morphology in preadolescents. METHODS We used data from 3133 preadolescents from a birth cohort from Rotterdam, the Netherlands (enrollment: 2002-2006). Concentrations of nitrogen oxides, coarse, fine, and ultrafine particles, and composition of fine particles were estimated for participant's home addresses in pregnancy and childhood, using land use regression models. Structural brain images were obtained at age 9-12 years. We assessed the relationships of air pollution exposure, with brain volumes, and surface-based morphometric data, adjusting for socioeconomic and life-style characteristics, using single as well as multi-pollutant approach. RESULTS No associations were observed between air pollution exposures and global volumes of total brain, and cortical and subcortical grey matter. However, we found associations between higher pregnancy and childhood air pollution exposures with smaller corpus callosum, smaller hippocampus, larger amygdala, smaller nucleus accumbens, and larger cerebellum (e.g. -69.2mm3 hippocampal volume [95%CI -129.1 to -9.3] per 1ng/m3 increase in pregnancy exposure to polycyclic aromatic hydrocarbons). Higher pregnancy exposure to air pollution was associated with smaller cortical thickness while higher childhood exposure was associated with predominantly larger cortical surface area. CONCLUSION Higher pregnancy or childhood exposure to several air pollutants was associated with altered volume of several brain structures, as well as with cortical thickness and surface area. Associations showed some similarity to delayed maturation and effects of early-life stress.
Collapse
Affiliation(s)
- Małgorzata J Lubczyńska
- ISGlobal, Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain; Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Spain
| | - Ryan L Muetzel
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands; The Generation R Study Group, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Hanan El Marroun
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands; Department of Psychology, Education and Child Studies, Erasmus School of Social and Behavioural Sciences, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Gerard Hoek
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Ingeborg M Kooter
- Department of Circular Economy & Environment, Netherlands Organisation for Applied Scientific Research, Utrecht, the Netherlands
| | - Errol M Thomson
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Canada; Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Manon Hillegers
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands; Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Tonya White
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands; Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands; Department of Social and Behavioral Science, Harvard T.H. Chan School of Public Health, Boston, USA
| | - Mònica Guxens
- ISGlobal, Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain; Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Spain; Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands.
| |
Collapse
|
425
|
Chopra S, Fornito A, Francey SM, O'Donoghue B, Cropley V, Nelson B, Graham J, Baldwin L, Tahtalian S, Yuen HP, Allott K, Alvarez-Jimenez M, Harrigan S, Sabaroedin K, Pantelis C, Wood SJ, McGorry P. Differentiating the effect of antipsychotic medication and illness on brain volume reductions in first-episode psychosis: A Longitudinal, Randomised, Triple-blind, Placebo-controlled MRI Study. Neuropsychopharmacology 2021; 46:1494-1501. [PMID: 33637835 PMCID: PMC8209146 DOI: 10.1038/s41386-021-00980-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/17/2021] [Accepted: 01/20/2021] [Indexed: 01/31/2023]
Abstract
Changes in brain volume are a common finding in Magnetic Resonance Imaging (MRI) studies of people with psychosis and numerous longitudinal studies suggest that volume deficits progress with illness duration. However, a major unresolved question concerns whether these changes are driven by the underlying illness or represent iatrogenic effects of antipsychotic medication. In this study, 62 antipsychotic-naïve patients with first-episode psychosis (FEP) received either a second-generation antipsychotic (risperidone or paliperidone) or a placebo pill over a treatment period of 6 months. Both FEP groups received intensive psychosocial therapy. A healthy control group (n = 27) was also recruited. Structural MRI scans were obtained at baseline, 3 months and 12 months. Our primary aim was to differentiate illness-related brain volume changes from medication-related changes within the first 3 months of treatment. We secondarily investigated long-term effects at the 12-month timepoint. From baseline to 3 months, we observed a significant group x time interaction in the pallidum (p < 0.05 FWE-corrected), such that patients receiving antipsychotic medication showed increased volume, patients on placebo showed decreased volume, and healthy controls showed no change. Across the entire patient sample, a greater increase in pallidal grey matter volume over 3 months was associated with a greater reduction in symptom severity. Our findings indicate that psychotic illness and antipsychotic exposure exert distinct and spatially distributed effects on brain volume. Our results align with prior work in suggesting that the therapeutic efficacy of antipsychotic medications may be primarily mediated through their effects on the basal ganglia.
Collapse
Affiliation(s)
- Sidhant Chopra
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia.
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia.
| | - Alex Fornito
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
| | - Shona M Francey
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Brian O'Donoghue
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Vanessa Cropley
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne & Melbourne Health, Melbourne, VIC, Australia
| | - Barnaby Nelson
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Jessica Graham
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Lara Baldwin
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Steven Tahtalian
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne & Melbourne Health, Melbourne, VIC, Australia
| | - Hok Pan Yuen
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Kelly Allott
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Mario Alvarez-Jimenez
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Susy Harrigan
- Department of Social Work, Monash University, Clayton, VIC, Australia
- Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia
| | - Kristina Sabaroedin
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne & Melbourne Health, Melbourne, VIC, Australia
- The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J Wood
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
- School of Psychology, University Birmingham, Edgbaston, UK
| | - Patrick McGorry
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
426
|
Jalbrzikowski M, Hayes RA, Wood SJ, Nordholm D, Zhou JH, Fusar-Poli P, Uhlhaas PJ, Takahashi T, Sugranyes G, Kwak YB, Mathalon DH, Katagiri N, Hooker CI, Smigielski L, Colibazzi T, Via E, Tang J, Koike S, Rasser PE, Michel C, Lebedeva I, Hegelstad WTV, de la Fuente-Sandoval C, Waltz JA, Mizrahi R, Corcoran CM, Resch F, Tamnes CK, Haas SS, Lemmers-Jansen ILJ, Agartz I, Allen P, Amminger GP, Andreassen OA, Atkinson K, Bachman P, Baeza I, Baldwin H, Bartholomeusz CF, Borgwardt S, Catalano S, Chee MWL, Chen X, Cho KIK, Cooper RE, Cropley VL, Dolz M, Ebdrup BH, Fortea A, Glenthøj LB, Glenthøj BY, de Haan L, Hamilton HK, Harris MA, Haut KM, He Y, Heekeren K, Heinz A, Hubl D, Hwang WJ, Kaess M, Kasai K, Kim M, Kindler J, Klaunig MJ, Koppel A, Kristensen TD, Kwon JS, Lawrie SM, Lee J, León-Ortiz P, Lin A, Loewy RL, Ma X, McGorry P, McGuire P, Mizuno M, Møller P, Moncada-Habib T, Muñoz-Samons D, Nelson B, Nemoto T, Nordentoft M, Omelchenko MA, Oppedal K, Ouyang L, Pantelis C, Pariente JC, Raghava JM, Reyes-Madrigal F, Roach BJ, Røssberg JI, Rössler W, Salisbury DF, Sasabayashi D, Schall U, Schiffman J, Schlagenhauf F, Schmidt A, Sørensen ME, et alJalbrzikowski M, Hayes RA, Wood SJ, Nordholm D, Zhou JH, Fusar-Poli P, Uhlhaas PJ, Takahashi T, Sugranyes G, Kwak YB, Mathalon DH, Katagiri N, Hooker CI, Smigielski L, Colibazzi T, Via E, Tang J, Koike S, Rasser PE, Michel C, Lebedeva I, Hegelstad WTV, de la Fuente-Sandoval C, Waltz JA, Mizrahi R, Corcoran CM, Resch F, Tamnes CK, Haas SS, Lemmers-Jansen ILJ, Agartz I, Allen P, Amminger GP, Andreassen OA, Atkinson K, Bachman P, Baeza I, Baldwin H, Bartholomeusz CF, Borgwardt S, Catalano S, Chee MWL, Chen X, Cho KIK, Cooper RE, Cropley VL, Dolz M, Ebdrup BH, Fortea A, Glenthøj LB, Glenthøj BY, de Haan L, Hamilton HK, Harris MA, Haut KM, He Y, Heekeren K, Heinz A, Hubl D, Hwang WJ, Kaess M, Kasai K, Kim M, Kindler J, Klaunig MJ, Koppel A, Kristensen TD, Kwon JS, Lawrie SM, Lee J, León-Ortiz P, Lin A, Loewy RL, Ma X, McGorry P, McGuire P, Mizuno M, Møller P, Moncada-Habib T, Muñoz-Samons D, Nelson B, Nemoto T, Nordentoft M, Omelchenko MA, Oppedal K, Ouyang L, Pantelis C, Pariente JC, Raghava JM, Reyes-Madrigal F, Roach BJ, Røssberg JI, Rössler W, Salisbury DF, Sasabayashi D, Schall U, Schiffman J, Schlagenhauf F, Schmidt A, Sørensen ME, Suzuki M, Theodoridou A, Tomyshev AS, Tor J, Værnes TG, Velakoulis D, Venegoni GD, Vinogradov S, Wenneberg C, Westlye LT, Yamasue H, Yuan L, Yung AR, van Amelsvoort TAMJ, Turner JA, van Erp TGM, Thompson PM, Hernaus D. Association of Structural Magnetic Resonance Imaging Measures With Psychosis Onset in Individuals at Clinical High Risk for Developing Psychosis: An ENIGMA Working Group Mega-analysis. JAMA Psychiatry 2021; 78:753-766. [PMID: 33950164 PMCID: PMC8100913 DOI: 10.1001/jamapsychiatry.2021.0638] [Show More Authors] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/04/2021] [Indexed: 01/10/2023]
Abstract
Importance The ENIGMA clinical high risk (CHR) for psychosis initiative, the largest pooled neuroimaging sample of individuals at CHR to date, aims to discover robust neurobiological markers of psychosis risk. Objective To investigate baseline structural neuroimaging differences between individuals at CHR and healthy controls as well as between participants at CHR who later developed a psychotic disorder (CHR-PS+) and those who did not (CHR-PS-). Design, Setting, and Participants In this case-control study, baseline T1-weighted magnetic resonance imaging (MRI) data were pooled from 31 international sites participating in the ENIGMA Clinical High Risk for Psychosis Working Group. CHR status was assessed using the Comprehensive Assessment of At-Risk Mental States or Structured Interview for Prodromal Syndromes. MRI scans were processed using harmonized protocols and analyzed within a mega-analysis and meta-analysis framework from January to October 2020. Main Outcomes and Measures Measures of regional cortical thickness (CT), surface area, and subcortical volumes were extracted from T1-weighted MRI scans. Independent variables were group (CHR group vs control group) and conversion status (CHR-PS+ group vs CHR-PS- group vs control group). Results Of the 3169 included participants, 1428 (45.1%) were female, and the mean (SD; range) age was 21.1 (4.9; 9.5-39.9) years. This study included 1792 individuals at CHR and 1377 healthy controls. Using longitudinal clinical information, 253 in the CHR-PS+ group, 1234 in the CHR-PS- group, and 305 at CHR without follow-up data were identified. Compared with healthy controls, individuals at CHR exhibited widespread lower CT measures (mean [range] Cohen d = -0.13 [-0.17 to -0.09]), but not surface area or subcortical volume. Lower CT measures in the fusiform, superior temporal, and paracentral regions were associated with psychosis conversion (mean Cohen d = -0.22; 95% CI, -0.35 to 0.10). Among healthy controls, compared with those in the CHR-PS+ group, age showed a stronger negative association with left fusiform CT measures (F = 9.8; P < .001; q < .001) and left paracentral CT measures (F = 5.9; P = .005; q = .02). Effect sizes representing lower CT associated with psychosis conversion resembled patterns of CT differences observed in ENIGMA studies of schizophrenia (ρ = 0.35; 95% CI, 0.12 to 0.55; P = .004) and individuals with 22q11.2 microdeletion syndrome and a psychotic disorder diagnosis (ρ = 0.43; 95% CI, 0.20 to 0.61; P = .001). Conclusions and Relevance This study provides evidence for widespread subtle, lower CT measures in individuals at CHR. The pattern of CT measure differences in those in the CHR-PS+ group was similar to those reported in other large-scale investigations of psychosis. Additionally, a subset of these regions displayed abnormal age associations. Widespread disruptions in CT coupled with abnormal age associations in those at CHR may point to disruptions in postnatal brain developmental processes.
Collapse
Affiliation(s)
- Maria Jalbrzikowski
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rebecca A Hayes
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Stephen J Wood
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
- Orygen, Melbourne, Australia
- School of Psychology, University of Birmingham, Birmingham, United Kingdom
| | - Dorte Nordholm
- Copenhagen Research Center for Mental Health, Mental Health Center Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Juan H Zhou
- Center for Sleep and Cognition, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Center for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Paolo Fusar-Poli
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- EPIC Lab, Department of Psychosis Studies, King's College London, London, United Kingdom
| | - Peter J Uhlhaas
- Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom
| | - Tsutomu Takahashi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Gisela Sugranyes
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neuroscience, 2017SGR-881, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Universitat de Barcelona, Barcelona, Spain
| | - Yoo Bin Kwak
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, Republic of Korea
| | - Daniel H Mathalon
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco
- San Francisco Veterans Affairs Health Care System, San Francisco, California
| | - Naoyuki Katagiri
- Department of Neuropsychiatry, Toho University School of Medicine, Tokyo, Japan
| | - Christine I Hooker
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois
| | - Lukasz Smigielski
- Department of Child and Adolescent Psychiatry, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Tiziano Colibazzi
- Department of Psychiatry, Columbia University, New York, New York
- New York State Psychiatric Institute, New York
| | - Esther Via
- Child and Adolescent Mental Health Research Group, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Child and Adolescent Psychiatry and Psychology Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Jinsong Tang
- Department of Psychiatry, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Neurobiology of Zhejiang Province, School of Medicine, Zhejiang University Hangzhou, Hangzhou, China
| | - Shinsuke Koike
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and Sciences, The University of Tokyo, Tokyo, Japan
- The University of Tokyo Institute for Diversity and Adaptation of Human Mind, Tokyo, Japan
| | - Paul E Rasser
- Priority Centre for Brain and Mental Health Research, The University of Newcastle, Newcastle, Australia
- Priority Research Centre for Stroke and Brain Injury, The University of Newcastle, Newcastle, Australia
| | - Chantal Michel
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | | | - Wenche Ten Velden Hegelstad
- Faculty of Social Sciences, University of Stavanger, Stavanger, Norway
- TIPS Centre for Clinical Research in Psychosis, Stavanger University Hospital, Stavanger, Norway
| | | | - James A Waltz
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore
| | - Romina Mizrahi
- Douglas Research Center, Montreal, Quebec, Canada
- McGill University, Department of Psychiatry, Montreal, Quebec, Canada
| | - Cheryl M Corcoran
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
- Mental Illness Research, Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, New York, New York
| | - Franz Resch
- Clinic for Child and Adolescent Psychiatry, University Hospital of Heidelberg, Heidelberg, Germany
| | - Christian K Tamnes
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- PROMENTA Research Center, Department of Psychology, University of Oslo, Oslo, Norway
| | - Shalaila S Haas
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Imke L J Lemmers-Jansen
- Faculty of Behavioural and Movement Sciences, Department of Clinical, Neuro and Developmental Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Ingrid Agartz
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Paul Allen
- Department of Psychology, University of Roehampton, London, United Kingdom
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - G Paul Amminger
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
- Orygen, Melbourne, Australia
| | - Ole A Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kimberley Atkinson
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter Bachman
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Inmaculada Baeza
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neuroscience, 2017SGR-881, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Universitat de Barcelona, Barcelona, Spain
| | - Helen Baldwin
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- NIHR Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, United Kingdom
| | - Cali F Bartholomeusz
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
- Orygen, Melbourne, Australia
| | - Stefan Borgwardt
- Department of Psychiatry, University of Basel, Basel, Switzerland
- Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| | - Sabrina Catalano
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael W L Chee
- Center for Sleep and Cognition, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Xiaogang Chen
- National Clinical Research Center for Mental Disorders and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Kang Ik K Cho
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rebecca E Cooper
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Melbourne, Australia
| | - Vanessa L Cropley
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Melbourne, Australia
- Centre for Mental Health, Faculty of Health, Arts and Design, School of Health Sciences, Swinburne University, Melbourne, Australia
| | - Montserrat Dolz
- Child and Adolescent Mental Health Research Group, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Child and Adolescent Psychiatry and Psychology Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Bjørn H Ebdrup
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Adriana Fortea
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neuroscience, Hospital Clinic Barcelona, Fundació Clínic Recerca Biomèdica, Universitat de Barcelona, Barcelona, Spain
| | - Louise Birkedal Glenthøj
- Copenhagen Research Center for Mental Health, Mental Health Center Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Birte Y Glenthøj
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lieuwe de Haan
- Department of Psychiatry, Amsterdam University Medical Centre, Amsterdam, the Netherlands
- Arkin, Amsterdam, the Netherlands
| | - Holly K Hamilton
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco
- San Francisco Veterans Affairs Health Care System, San Francisco, California
| | - Mathew A Harris
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Kristen M Haut
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois
| | - Ying He
- National Clinical Research Center for Mental Disorders and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Karsten Heekeren
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Psychiatry and Psychotherapy I, LVR-Hospital Cologne, Cologne, Germany
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin, Berlin, Germany
| | - Daniela Hubl
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Wu Jeong Hwang
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, Republic of Korea
| | - Michael Kaess
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
- Department of Child and Adolescent Psychiatry, Center of Psychosocial Medicine, University of Heidelberg, Heidelberg, Germany
| | - Kiyoto Kasai
- The University of Tokyo Institute for Diversity and Adaptation of Human Mind, Tokyo, Japan
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- The International Research Center for Neurointelligence at The University of Tokyo Institutes for Advanced Study, The University of Tokyo, Tokyo, Japan
| | - Minah Kim
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jochen Kindler
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Mallory J Klaunig
- Department of Psychology, University of Maryland, Baltimore County, Baltimore
| | - Alex Koppel
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Tina D Kristensen
- Copenhagen Research Center for Mental Health, Mental Health Center Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
| | - Jun Soo Kwon
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Stephen M Lawrie
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Jimmy Lee
- Department of Psychosis, Institute of Mental Health, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Pablo León-Ortiz
- Laboratory of Experimental Psychiatry, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Ashleigh Lin
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Rachel L Loewy
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco
| | - Xiaoqian Ma
- National Clinical Research Center for Mental Disorders and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Patrick McGorry
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
- Orygen, Melbourne, Australia
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Masafumi Mizuno
- Department of Neuropsychiatry, Toho University School of Medicine, Tokyo, Japan
| | - Paul Møller
- Department for Mental Health Research and Development, Division of Mental Health and Addiction, Vestre Viken Hospital Trust, Lier, Norway
| | - Tomas Moncada-Habib
- Laboratory of Experimental Psychiatry, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Daniel Muñoz-Samons
- Child and Adolescent Mental Health Research Group, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Child and Adolescent Psychiatry and Psychology Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Barnaby Nelson
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
- Orygen, Melbourne, Australia
| | - Takahiro Nemoto
- Department of Neuropsychiatry, Toho University School of Medicine, Tokyo, Japan
| | - Merete Nordentoft
- Copenhagen Research Center for Mental Health, Mental Health Center Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Ketil Oppedal
- Stavanger Medical Imaging Laboratory, Department of Radiology, Stavanger University Hospital, Stavanger, Norway
| | - Lijun Ouyang
- National Clinical Research Center for Mental Disorders and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Melbourne, Australia
- Centre for Mental Health, Faculty of Health, Arts and Design, School of Health Sciences, Swinburne University, Melbourne, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Jose C Pariente
- Magnetic Resonance Imaging Core Facility, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Jayachandra M Raghava
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
- Department of Clinical Physiology, Nuclear Medicine and PET, Functional Imaging Unit, University of Copenhagen, Glostrup, Denmark
| | - Francisco Reyes-Madrigal
- Laboratory of Experimental Psychiatry, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Brian J Roach
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco
- San Francisco Veterans Affairs Health Care System, San Francisco, California
| | - Jan I Røssberg
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Wulf Rössler
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin, Berlin, Germany
| | - Dean F Salisbury
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Daiki Sasabayashi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Ulrich Schall
- Priority Centre for Brain and Mental Health Research, The University of Newcastle, Newcastle, Australia
- Priority Research Centre Grow Up Well, The University of Newcastle, Newcastle, Australia
| | - Jason Schiffman
- Department of Psychology, University of Maryland, Baltimore County, Baltimore
- Department of Psychological Science, University of California, Irvine
| | - Florian Schlagenhauf
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin, Berlin, Germany
| | - Andre Schmidt
- Department of Psychiatry, University of Basel, Basel, Switzerland
| | - Mikkel E Sørensen
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
| | - Michio Suzuki
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Anastasia Theodoridou
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | | - Jordina Tor
- Child and Adolescent Mental Health Research Group, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Child and Adolescent Psychiatry and Psychology Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Tor G Værnes
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Early Intervention in Psychosis Advisory Unit for South-East Norway, TIPS Sør-Øst, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Dennis Velakoulis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Melbourne, Australia
- Neuropsychiatry, The Royal Melbourne Hospital, Melbourne, Australia
| | - Gloria D Venegoni
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Faculty of Health Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Sophia Vinogradov
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis
| | - Christina Wenneberg
- Copenhagen Research Center for Mental Health, Mental Health Center Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lars T Westlye
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Hidenori Yamasue
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu City, Japan
| | - Liu Yuan
- National Clinical Research Center for Mental Disorders and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Alison R Yung
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
- Orygen, Melbourne, Australia
- School of Health Sciences, University of Manchester, Manchester, United Kingdom
| | - Thérèse A M J van Amelsvoort
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Faculty of Health Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | | | - Theo G M van Erp
- Center for the Neurobiology of Learning and Memory, Irvine, California
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California, Irvine
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging and Informatics, Keck School of Medicine of USC, University of Southern California, Los Angeles
| | - Dennis Hernaus
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Faculty of Health Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
427
|
Individual deviations from normative models of brain structure in a large cross-sectional schizophrenia cohort. Mol Psychiatry 2021; 26:3512-3523. [PMID: 32963336 PMCID: PMC8329928 DOI: 10.1038/s41380-020-00882-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 08/21/2020] [Accepted: 09/04/2020] [Indexed: 12/12/2022]
Abstract
The heterogeneity of schizophrenia has defied efforts to derive reproducible and definitive anatomical maps of structural brain changes associated with the disorder. We aimed to map deviations from normative ranges of brain structure for individual patients and evaluate whether the loci of individual deviations recapitulated group-average brain maps of schizophrenia pathology. For each of 48 white matter tracts and 68 cortical regions, normative percentiles of variation in fractional anisotropy (FA) and cortical thickness (CT) were established using diffusion-weighted and structural MRI from healthy adults (n = 195). Individuals with schizophrenia (n = 322) were classified as either within the normative range for healthy individuals of the same age and sex (5-95% percentiles), infra-normal (<5% percentile) or supra-normal (>95% percentile). Repeating this classification for each tract and region yielded a deviation map for each individual. Compared to the healthy comparison group, the schizophrenia group showed widespread reductions in FA and CT, involving virtually all white matter tracts and cortical regions. Paradoxically, however, no more than 15-20% of patients deviated from the normative range for any single tract or region. Furthermore, 79% of patients showed infra-normal deviations for at least one locus (healthy individuals: 59 ± 2%, p < 0.001). Thus, while infra-normal deviations were common among patients, their anatomical loci were highly inconsistent between individuals. Higher polygenic risk for schizophrenia associated with a greater number of regions with infra-normal deviations in CT (r = -0.17, p = 0.006). We conclude that anatomical loci of schizophrenia-related changes are highly heterogeneous across individuals to the extent that group-consensus pathological maps are not representative of most individual patients. Normative modeling can aid in parsing schizophrenia heterogeneity and guiding personalized interventions.
Collapse
|
428
|
De Peri L, Deste G, Vita A. Strucutural brain imaging at the onset of schizophrenia:What have we learned and what have we missed. Psychiatry Res 2021; 301:113962. [PMID: 33945963 DOI: 10.1016/j.psychres.2021.113962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 11/28/2022]
Abstract
Over the past 50 years, the application of structural neuroimaging techniques to schizophrenia research has added relevant information about the pathophysiology of the disorder. Several lines of investigation gave strong evidence that schizophrenia is associated with multiple subtle brain abnormalities that involve both cerebral grey and white matter volumes and structure. The time of onset and longitudinal course of brain morphological abnormalities support the notion that brain pathology of schizophrenia has a neurodevelopmental component and a progressive course, although several confounders of brain changes should be carefully taken into account. Brain anomalies detected before and close to the onset of schizophrenia are likely to be unrelated to confounders of brain changes such as antipsychotic drug treatment, duration of illness or illicit substance abuse, i.e. they related to the pathological process of the disorder per se. Nonetheless, clinically useful diagnostic or prognostic biomarkers have not derived from neuroimaging studies and this is likely related to the neurobiological heterogeneity of the disorder. Thus, there is the compelling need to set new methodological standards for developing innovative hypothesis-driven studies to overcome what we have missed to date in neuroimaging research in schizophrenia.
Collapse
Affiliation(s)
- Luca De Peri
- Cantonal Psychiatric Clinic, Cantonal Socio-Psychiatric Association, Mendrisio, Switzerland
| | - Giacomo Deste
- Department of Mental Health, Spedali Civili Hospital, Brescia, Italy
| | - Antonio Vita
- Department of Mental Health, Spedali Civili Hospital, Brescia, Italy; Department of Clinical and Experimentale Sciences, University of Brescia, Italy.
| |
Collapse
|
429
|
Ramsay IS, Fryer S, Roach BJ, Boos A, Fisher M, Loewy R, Ford JM, Vinogradov S, Mathalon DH. Response to targeted cognitive training may be neuroprotective in patients with early schizophrenia. Psychiatry Res Neuroimaging 2021; 312:111285. [PMID: 33865147 PMCID: PMC8137670 DOI: 10.1016/j.pscychresns.2021.111285] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/18/2021] [Accepted: 04/06/2021] [Indexed: 01/07/2023]
Abstract
Individuals with schizophrenia exhibit widespread cortical thinning associated with illness severity and deficits in cognition. However, intact cortical thickness (CTh) may serve as a protective factor. The current study sought to examine changes in CTh in response to auditory targeted cognitive training (TCT) in individuals with recent onset schizophrenia. Participants underwent MRI scanning and a cognitive assessment before and after being randomly assigned to 40 h of either TCT (N = 21) or a computer games control condition (CG; N = 22) over 16 weeks. Groups did not differ at baseline on demographic variables or measures of CTh. At the level of group averages, neither group showed significant pre-post changes in CTh in any brain region. However, changes in CTh related to individual differences in treatment outcome, as improved global cognition in the TCT group corresponded to reduced cortical thinning in frontal, temporal, parietal, and occipital lobes. These relationships were not observed in the CG group. The current findings suggest that TCT may be neuroprotective in early schizophrenia, such that individuals who improved in response to training also showed a reduction in cortical thinning that may be otherwise hastened due to age and illness.
Collapse
Affiliation(s)
- Ian S Ramsay
- University of Minnesota, Department of Psychiatry and Behavioral Sciences, USA.
| | - Susanna Fryer
- University of California, San Francisco, Department of Psychiatry and Behavioral Science, USA; San Francisco Veterans Affairs Medical Center, USA
| | - Brian J Roach
- University of California, San Francisco, Department of Psychiatry and Behavioral Science, USA; San Francisco Veterans Affairs Medical Center, USA
| | - Alison Boos
- University of California, San Francisco, Department of Psychiatry and Behavioral Science, USA; San Francisco Veterans Affairs Medical Center, USA
| | - Melissa Fisher
- University of Minnesota, Department of Psychiatry and Behavioral Sciences, USA
| | - Rachel Loewy
- University of California, San Francisco, Department of Psychiatry and Behavioral Science, USA; San Francisco Veterans Affairs Medical Center, USA
| | - Judith M Ford
- University of California, San Francisco, Department of Psychiatry and Behavioral Science, USA; San Francisco Veterans Affairs Medical Center, USA
| | - Sophia Vinogradov
- University of Minnesota, Department of Psychiatry and Behavioral Sciences, USA
| | - Daniel H Mathalon
- University of California, San Francisco, Department of Psychiatry and Behavioral Science, USA; San Francisco Veterans Affairs Medical Center, USA
| |
Collapse
|
430
|
Rodriguez-Perez N, Ayesa-Arriola R, Ortiz-García de la Foz V, Setien-Suero E, Tordesillas-Gutierrez D, Crespo-Facorro B. Long term cortical thickness changes after a first episode of non- affective psychosis: The 10 year follow-up of the PAFIP cohort. Prog Neuropsychopharmacol Biol Psychiatry 2021; 108:110180. [PMID: 33212193 DOI: 10.1016/j.pnpbp.2020.110180] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/28/2020] [Accepted: 11/12/2020] [Indexed: 12/25/2022]
Abstract
Cortical thickness has been widely studied in individuals with schizophrenia and, in particular, first-episode psychosis. Abnormalities have been described, although there is, to date, a lack of consensus regarding changes across time and correlations with clinical and functional outcomes of the illness. One hundred and twenty-three first-episode psychosis patients and 74 healthy volunteers were subjected to magnetic resonance imaging scans and clinical and functional assessments by different scales at four consecutive visits during a 10 year follow-up period. Linear mixed effects models were applied to our data to compute cortical thickness changes over time in (1) schizophrenia patients versus healthy controls and (2) in patients with good versus poor functional outcome. The associations between cortical thickness percentage changes and clinical and functional status at 10 years were also assessed. The patients presented a thinner cortex than the controls at baseline (b's = -0.06; q ≤ 0.00023) with non-significant coefficients for the interaction term (follow-up time x group) (b's = -0.001; q ≥ 0.681). Poor functioning patients presented statistically significant coefficients for the interaction term (follow-up time x functionality) (left: b = -0.005, q = 0.019; right: b = -0.005, q = 0.022). In contrast, no correlations were found between cortical thickness measurements and clinical variables at 10 years. Overall, there were widespread thickness anomalies in first-episode psychosis patients across cortical regions that remained stable across time. Progressive thickness changes were related to patient functional outcomes, with progressive and steeper cortical thinning in patients with worse functional outcomes and a stabilization in those with better outcomes.
Collapse
Affiliation(s)
- Noelia Rodriguez-Perez
- Hospital Universitario Virgen del Rocío, Department of Psychiatry, Instituto de Investigación Sanitaria de Sevilla, IBiS, Sevilla, Spain; CIBERSAM, Centro Investigación Biomédica en Red Salud Mental, Madrid, Spain.
| | - Rosa Ayesa-Arriola
- University Hospital Marqués de Valdecilla, IDIVAL, Department of Psychiatry, School of Medicine, University of Cantabria, Santander, Spain
| | - Victor Ortiz-García de la Foz
- University Hospital Marqués de Valdecilla, IDIVAL, Department of Psychiatry, School of Medicine, University of Cantabria, Santander, Spain
| | - Esther Setien-Suero
- University Hospital Marqués de Valdecilla, IDIVAL, Department of Psychiatry, School of Medicine, University of Cantabria, Santander, Spain
| | - Diana Tordesillas-Gutierrez
- CIBERSAM, Centro Investigación Biomédica en Red Salud Mental, Madrid, Spain; Neuroimaging Unit, Technological Facilities, IDIVAL, Santander, Spain
| | - Benedicto Crespo-Facorro
- Hospital Universitario Virgen del Rocío, Department of Psychiatry, Instituto de Investigación Sanitaria de Sevilla, IBiS, Sevilla, Spain; CIBERSAM, Centro Investigación Biomédica en Red Salud Mental, Madrid, Spain; University of Sevilla, Sevilla, Spain.
| | | |
Collapse
|
431
|
Song W, Qian W, Wang W, Yu S, Lin GN. Mendelian randomization studies of brain MRI yield insights into the pathogenesis of neuropsychiatric disorders. BMC Genomics 2021; 22:342. [PMID: 34078268 PMCID: PMC8171058 DOI: 10.1186/s12864-021-07661-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 04/29/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Observational studies have identified various associations between neuroimaging alterations and neuropsychiatric disorders. However, whether such associations could truly reflect causal relations remains still unknown. RESULTS Here, we leveraged genome-wide association studies (GWAS) summary statistics for (1) 11 psychiatric disorders (sample sizes varied from n = 9,725 to 1,331,010); (2) 110 diffusion tensor imaging (DTI) measurement (sample size n = 17,706); (3) 101 region-of-interest (ROI) volumes, and investigate the causal relationship between brain structures and neuropsychiatric disorders by two-sample Mendelian randomization. Among all DTI-Disorder combinations, we observed a significant causal association between the superior longitudinal fasciculus (SLF) and the risk of Anorexia nervosa (AN) (Odds Ratio [OR] = 0.62, 95 % confidence interval: 0.50 ~ 0.76, P = 6.4 × 10- 6). Similar significant associations were also observed between the body of the corpus callosum (fractional anisotropy) and Alzheimer's disease (OR = 1.07, 95 % CI: 1.03 ~ 1.11, P = 4.1 × 10- 5). By combining all observations, we found that the overall p-value for DTI - Disorder associations was significantly elevated compared to the null distribution (Kolmogorov-Smirnov P = 0.009, inflation factor λ = 1.37), especially for DTI - Bipolar disorder (BP) (λ = 2.64) and DTI - AN (λ = 1.82). In contrast, for ROI-Disorder combinations, we only found a significant association between the brain region of pars triangularis and Schizophrenia (OR = 0.48, 95 % CI: 0.34 ~ 0.69, P = 5.9 × 10- 5) and no overall p-value elevation for ROI-Disorder analysis compared to the null expectation. CONCLUSIONS As a whole, we show that SLF degeneration may be a risk factor for AN, while DTI variations could be causally related to some neuropsychiatric disorders, such as BP and AN. Also, the white matter structure might have a larger impact on neuropsychiatric disorders than subregion volumes.
Collapse
Affiliation(s)
- Weichen Song
- Shanghai Mental Health Center, School of Biomedical Engineering, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, 200030, Shanghai, China
| | - Wei Qian
- Shanghai Mental Health Center, School of Biomedical Engineering, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, 200030, Shanghai, China
| | - Weidi Wang
- Shanghai Mental Health Center, School of Biomedical Engineering, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, 200030, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, 200030, Shanghai, China
| | - Shunying Yu
- Shanghai Mental Health Center, School of Biomedical Engineering, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, 200030, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, 200030, Shanghai, China
| | - Guan Ning Lin
- Shanghai Mental Health Center, School of Biomedical Engineering, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, 200030, Shanghai, China.
- Shanghai Key Laboratory of Psychotic Disorders, 200030, Shanghai, China.
| |
Collapse
|
432
|
Abstract
Brain scientists are now capable of collecting more data in a single experiment than researchers a generation ago might have collected over an entire career. Indeed, the brain itself seems to thirst for more and more data. Such digital information not only comprises individual studies but is also increasingly shared and made openly available for secondary, confirmatory, and/or combined analyses. Numerous web resources now exist containing data across spatiotemporal scales. Data processing workflow technologies running via cloud-enabled computing infrastructures allow for large-scale processing. Such a move toward greater openness is fundamentally changing how brain science results are communicated and linked to available raw data and processed results. Ethical, professional, and motivational issues challenge the whole-scale commitment to data-driven neuroscience. Nevertheless, fueled by government investments into primary brain data collection coupled with increased sharing and community pressure challenging the dominant publishing model, large-scale brain and data science is here to stay.
Collapse
Affiliation(s)
- John Darrell Van Horn
- Department of Psychology, University of Virginia, Charlottesville, Virginia, USA
- School of Data Science, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
433
|
Moreau CA, Ching CR, Kumar K, Jacquemont S, Bearden CE. Structural and functional brain alterations revealed by neuroimaging in CNV carriers. Curr Opin Genet Dev 2021; 68:88-98. [PMID: 33812299 PMCID: PMC8205978 DOI: 10.1016/j.gde.2021.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/01/2021] [Accepted: 03/09/2021] [Indexed: 01/21/2023]
Abstract
Copy Number Variants (CNVs) are associated with elevated rates of neuropsychiatric disorders. A 'genetics-first' approach, involving the CNV effects on the brain, irrespective of clinical symptomatology, allows investigation of mechanisms underlying neuropsychiatric disorders in the general population. Recent years have seen an increasing number of larger multisite neuroimaging studies investigating the effect of CNVs on structural and functional brain endophenotypes. Alterations overlap with those found in idiopathic psychiatric conditions but effect sizes are twofold to fivefold larger. Here we review new CNV-associated structural and functional brain alterations and outline the future of neuroimaging genomics research, with particular emphasis on developing new resources for the study of high-risk CNVs and rare genomic variants.
Collapse
Affiliation(s)
- Clara A Moreau
- Sainte-Justine Hospital Research Center, Montreal, Canada; Department of Pediatrics, University of Montreal, Montreal, Canada; Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montréal, Canada; Human Genetics and Cognitive Functions, CNRS UMR 3571, Université de Paris, Institut Pasteur, Paris, France
| | - Christopher Rk Ching
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, USA
| | - Kuldeep Kumar
- Sainte-Justine Hospital Research Center, Montreal, Canada
| | - Sebastien Jacquemont
- Sainte-Justine Hospital Research Center, Montreal, Canada; Department of Pediatrics, University of Montreal, Montreal, Canada.
| | - Carrie E Bearden
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and Psychology, University of California, Los Angeles, USA.
| |
Collapse
|
434
|
Radonjić NV, Hess JL, Rovira P, Andreassen O, Buitelaar JK, Ching CRK, Franke B, Hoogman M, Jahanshad N, McDonald C, Schmaal L, Sisodiya SM, Stein DJ, van den Heuvel OA, van Erp TGM, van Rooij D, Veltman DJ, Thompson P, Faraone SV. Structural brain imaging studies offer clues about the effects of the shared genetic etiology among neuropsychiatric disorders. Mol Psychiatry 2021; 26:2101-2110. [PMID: 33456050 PMCID: PMC8440178 DOI: 10.1038/s41380-020-01002-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Genomewide association studies have found significant genetic correlations among many neuropsychiatric disorders. In contrast, we know much less about the degree to which structural brain alterations are similar among disorders and, if so, the degree to which such similarities have a genetic etiology. From the Enhancing Neuroimaging Genetics through Meta-Analysis (ENIGMA) consortium, we acquired standardized mean differences (SMDs) in regional brain volume and cortical thickness between cases and controls. We had data on 41 brain regions for: attention-deficit/hyperactivity disorder (ADHD), autism spectrum disorder (ASD), bipolar disorder (BD), epilepsy, major depressive disorder (MDD), obsessive compulsive disorder (OCD), and schizophrenia (SCZ). These data had been derived from 24,360 patients and 37,425 controls. The SMDs were significantly correlated between SCZ and BD, OCD, MDD, and ASD. MDD was positively correlated with BD and OCD. BD was positively correlated with OCD and negatively correlated with ADHD. These pairwise correlations among disorders were correlated with the corresponding pairwise correlations among disorders derived from genomewide association studies (r = 0.494). Our results show substantial similarities in sMRI phenotypes among neuropsychiatric disorders and suggest that these similarities are accounted for, in part, by corresponding similarities in common genetic variant architectures.
Collapse
Affiliation(s)
- Nevena V Radonjić
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Jonathan L Hess
- Departments of Psychiatry and of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Paula Rovira
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Ole Andreassen
- NORMENT-Institute of Clinical Medicine, Division of Mental Health and Addiction, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Jan K Buitelaar
- Radboudumc, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Christopher R K Ching
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Marina Del Rey, CA, USA
| | - Barbara Franke
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martine Hoogman
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Neda Jahanshad
- Imaging Genetics Center, Department of Neurology and Biomedical Engineering, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina Del Rey, CA, USA
| | - Carrie McDonald
- Department of Psychiatry, Center for Multimodal Imaging and Genetics (CMIG), University of California, San Diego, CA, USA
| | - Lianne Schmaal
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Orygen, The National Centre of Excellence for Youth Mental Health, Parkville, VIC, Australia
| | - Sanjay M Sisodiya
- UCL Queen Square Institute of Neurology, Department of Clinical and Experimental Epilepsy, University College London, London, UK
- Chalfont Centre for Epilepsy, Epilepsy Society, Bucks, UK
| | - Dan J Stein
- SA MRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry & Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Odile A van den Heuvel
- Department of Psychiatry and Department of Anatomy & Neurosciences, Amsterdam UMC/VUmc, Amsterdam, The Netherlands
| | - Theo G M van Erp
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA, USA
| | - Daan van Rooij
- Donders Centre for Cognitive Neuroimaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dick J Veltman
- Department of Psychiatry and Department of Anatomy & Neurosciences, Amsterdam UMC/VUmc, Amsterdam, The Netherlands
| | - Paul Thompson
- Neuro Imaging Institute for Neuroimaging and Informatics, Keck School of Medicine of the University of Southern California, Marina Del Rey, CA, USA
| | - Stephen V Faraone
- Departments of Psychiatry and of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
435
|
Kim M, Haney JR, Zhang P, Hernandez LM, Wang LK, Perez-Cano L, Loohuis LMO, de la Torre-Ubieta L, Gandal MJ. Brain gene co-expression networks link complement signaling with convergent synaptic pathology in schizophrenia. Nat Neurosci 2021; 24:799-809. [PMID: 33958802 PMCID: PMC8178202 DOI: 10.1038/s41593-021-00847-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 03/25/2021] [Indexed: 02/02/2023]
Abstract
The most significant common variant association for schizophrenia (SCZ) reflects increased expression of the complement component 4A (C4A). Yet, it remains unclear how C4A interacts with other SCZ risk genes or whether the complement system more broadly is implicated in SCZ pathogenesis. Here, we integrate several existing, large-scale genetic and transcriptomic datasets to interrogate the functional role of the complement system and C4A in the human brain. Unexpectedly, we find no significant genetic enrichment among known complement system genes for SCZ. Conversely, brain co-expression network analyses using C4A as a seed gene reveal that genes downregulated when C4A expression increases exhibit strong and specific genetic enrichment for SCZ risk. This convergent genomic signal reflects synaptic processes, is sexually dimorphic and most prominent in frontal cortical brain regions, and is accentuated by smoking. Overall, these results indicate that synaptic pathways-rather than the complement system-are the driving force conferring SCZ risk.
Collapse
Affiliation(s)
- Minsoo Kim
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jillian R Haney
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Pan Zhang
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Leanna M Hernandez
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lee-Kai Wang
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Laura Perez-Cano
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- STALICLA DDS, Barcelona, Spain
| | - Loes M Olde Loohuis
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Luis de la Torre-Ubieta
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael J Gandal
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
436
|
Increased peripheral inflammation in schizophrenia is associated with worse cognitive performance and related cortical thickness reductions. Eur Arch Psychiatry Clin Neurosci 2021; 271:595-607. [PMID: 33760971 DOI: 10.1007/s00406-021-01237-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 02/03/2021] [Indexed: 12/16/2022]
Abstract
While the biological substrates of brain and behavioural changes in persons with schizophrenia remain unclear, increasing evidence implicates that inflammation is involved. In schizophrenia, including first-episode psychosis and anti-psychotic naïve patients, there are numerous reports of increased peripheral inflammation, cognitive deficits and neuropathologies such as cortical thinning. Research defining the relationship between inflammation and schizophrenia symptomatology and neuropathology is needed. Therefore, we analysed the level of C-reactive protein (CRP), a peripheral inflammation marker, and its relationship with cognitive functioning in a cohort of 644 controls and 499 schizophrenia patients. In a subset of individuals who underwent MRI scanning (99 controls and 194 schizophrenia cases), we tested if serum CRP was associated with cortical thickness. CRP was significantly increased in schizophrenia patients compared to controls, co-varying for age, sex, overweight/obesity and diabetes (p < 0.006E-10). In schizophrenia, increased CRP was mildly associated with worse performance in attention, controlling for age, sex and education (R =- 0.15, p = 0.001). Further, increased CRP was associated with reduced cortical thickness in three regions related to attention: the caudal middle frontal, the pars opercularis and the posterior cingulate cortices, which remained significant after controlling for multiple comparisons (all p < 0.05). Together, these findings indicate that increased peripheral inflammation is associated with deficits in cognitive function and brain structure in schizophrenia, especially reduced attention and reduced cortical thickness in associated brain regions. Using CRP as a biomarker of peripheral inflammation in persons with schizophrenia may help to identify vulnerable patients and those that may benefit from adjunctive anti-inflammatory treatments.
Collapse
|
437
|
Plitman E, Bussy A, Valiquette V, Salaciak A, Patel R, Cupo L, Béland ML, Tullo S, Tardif CL, Rajah MN, Near J, Devenyi GA, Chakravarty MM. The impact of the Siemens Tim Trio to Prisma upgrade and the addition of volumetric navigators on cortical thickness, structure volume, and 1H-MRS indices: An MRI reliability study with implications for longitudinal study designs. Neuroimage 2021; 238:118172. [PMID: 34082116 DOI: 10.1016/j.neuroimage.2021.118172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 11/30/2022] Open
Abstract
Many magnetic resonance imaging (MRI) measures are being studied longitudinally to explore topics such as biomarker detection and clinical staging. A pertinent concern to longitudinal work is MRI scanner upgrades. When upgrades occur during the course of a longitudinal MRI neuroimaging investigation, there may be an impact on the compatibility of pre- and post-upgrade measures. Similarly, subject motion is another issue that may be detrimental to MRI work and embedding volumetric navigators (vNavs) within acquisition sequences has emerged as a technique that allows for prospective motion correction. Our research group recently underwent an upgrade from a Siemens MAGNETOM 3T Tim Trio system to a Siemens MAGNETOM 3T Prisma Fit system. The goals of the current work were to: 1) investigate the impact of this upgrade on commonly used structural imaging measures and proton magnetic resonance spectroscopy indices ("Prisma Upgrade protocol") and 2) examine structural imaging measures in a sequence with vNavs alongside a standard acquisition sequence ("vNav protocol"). While high reliability was observed for most of the investigated MRI outputs, suboptimal reliability was observed for certain indices. Across the scanner upgrade, increases in frontal, temporal, and cingulate cortical thickness (CT) and thalamus volume, along with decreases in parietal CT and amygdala, globus pallidus, hippocampus, and striatum volumes, were observed. No significant impact of the upgrade was found in 1H-MRS analyses. Further, CT estimates were found to be larger in MPRAGE acquisitions compared to vNav-MPRAGE acquisitions mainly within temporal areas, while the opposite was found mostly in parietal brain regions. The results from this work should be considered in longitudinal study designs and comparable prospective motion correction investigations are warranted in cases of marked head movement.
Collapse
Affiliation(s)
- Eric Plitman
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada.
| | - Aurélie Bussy
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Canada
| | - Vanessa Valiquette
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Alyssa Salaciak
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Raihaan Patel
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| | - Lani Cupo
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Canada
| | - Marie-Lise Béland
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Stephanie Tullo
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Canada
| | - Christine Lucas Tardif
- Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada; McConnell Brain Imaging Centre, Montreal Neurological Institute, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - M Natasha Rajah
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Psychology, McGill University, Montreal, Quebec, Canada; Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Jamie Near
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada; Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Gabriel A Devenyi
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - M Mallar Chakravarty
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada; Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada.
| |
Collapse
|
438
|
St Clair D, Lang B. Schizophrenia: a classic battle ground of nature versus nurture debate. Sci Bull (Beijing) 2021; 66:1037-1046. [PMID: 36654248 DOI: 10.1016/j.scib.2021.01.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/29/2020] [Accepted: 10/13/2020] [Indexed: 01/20/2023]
Abstract
Much has been learned about the etiology and pathogenesis of schizophrenia since the term was first used by Eugene Bleuler over a century ago to describe one of the most important forms of major mental illness to affect mankind. Both nature and nurture feature prominently in our understanding of the genesis of the overall risk of developing schizophrenia. We now have a firm grasp of the broad structure of the genetic architecture and several key environmental risk factors have been identified and delineated. However, much of the heritability of schizophrenia remains unexplained and the reported environmental risk factors do not explain all the variances not attributable to genetic risk factors. The biggest problem at present is that our understanding of the causal mechanisms involved is still in its infancy. In this review, we describe the extent and limits of our knowledge of the specific genetic/constitutional and non-genetic/environmental factors that contribute to the overall risk of schizophrenia. We suggest novel methods may be required to understand the almost certainly immensely complex multi-level causal mechanisms that contribute to the generation of the schizophrenia phenotype.
Collapse
Affiliation(s)
- David St Clair
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, National Clinical Research Center for Mental Disorders, Changsha 410011, China; Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK; Bio-X Life Science Research Center, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Bing Lang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, National Clinical Research Center for Mental Disorders, Changsha 410011, China; Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
439
|
Gurholt TP, Kaufmann T, Frei O, Alnæs D, Haukvik UK, van der Meer D, Moberget T, O'Connell KS, Leinhard OD, Linge J, Simon R, Smeland OB, Sønderby IE, Winterton A, Steen NE, Westlye LT, Andreassen OA. Population-based body-brain mapping links brain morphology with anthropometrics and body composition. Transl Psychiatry 2021; 11:295. [PMID: 34006848 PMCID: PMC8131380 DOI: 10.1038/s41398-021-01414-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 12/13/2022] Open
Abstract
Understanding complex body-brain processes and the interplay between adipose tissue and brain health is important for understanding comorbidity between psychiatric and cardiometabolic disorders. We investigated associations between brain structure and anthropometric and body composition measures using brain magnetic resonance imaging (MRI; n = 24,728) and body MRI (n = 4973) of generally healthy participants in the UK Biobank. We derived regional and global measures of brain morphometry using FreeSurfer and tested their association with (i) anthropometric measures, and (ii) adipose and muscle tissue measured from body MRI. We identified several significant associations with small effect sizes. Anthropometric measures showed negative, nonlinear, associations with cerebellar/cortical gray matter, and brain stem structures, and positive associations with ventricular volumes. Subcortical structures exhibited mixed effect directionality, with strongest positive association for accumbens. Adipose tissue measures, including liver fat and muscle fat infiltration, were negatively associated with cortical/cerebellum structures, while total thigh muscle volume was positively associated with brain stem and accumbens. Regional investigations of cortical area, thickness, and volume indicated widespread and largely negative associations with anthropometric and adipose tissue measures, with an opposite pattern for thigh muscle volume. Self-reported diabetes, hypertension, or hypercholesterolemia were associated with brain structure. The findings provide new insight into physiological body-brain associations suggestive of shared mechanisms between cardiometabolic risk factors and brain health. Whereas the causality needs to be determined, the observed patterns of body-brain relationships provide a foundation for understanding the underlying mechanisms linking psychiatric disorders with obesity and cardiovascular disease, with potential for the development of new prevention strategies.
Collapse
Affiliation(s)
- Tiril P Gurholt
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway.
| | - Tobias Kaufmann
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Oleksandr Frei
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Dag Alnæs
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Unn K Haukvik
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Dennis van der Meer
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Torgeir Moberget
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Kevin S O'Connell
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Olof D Leinhard
- AMRA Medical, Linköping, Sweden
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | | | - Rozalyn Simon
- AMRA Medical, Linköping, Sweden
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Olav B Smeland
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Ida E Sønderby
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Adriano Winterton
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Nils Eiel Steen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Lars T Westlye
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway
| |
Collapse
|
440
|
The gut microbiome is associated with brain structure and function in schizophrenia. Sci Rep 2021; 11:9743. [PMID: 33963227 PMCID: PMC8105323 DOI: 10.1038/s41598-021-89166-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
The effect of the gut microbiome on the central nervous system and its possible role in mental disorders have received increasing attention. However, knowledge about the relationship between the gut microbiome and brain structure and function is still very limited. Here, we used 16S rRNA sequencing with structural magnetic resonance imaging (sMRI) and resting-state functional (rs-fMRI) to investigate differences in fecal microbiota between 38 patients with schizophrenia (SZ) and 38 demographically matched normal controls (NCs) and explored whether such differences were associated with brain structure and function. At the genus level, we found that the relative abundance of Ruminococcus and Roseburia was significantly lower, whereas the abundance of Veillonella was significantly higher in SZ patients than in NCs. Additionally, the analysis of MRI data revealed that several brain regions showed significantly lower gray matter volume (GMV) and regional homogeneity (ReHo) but significantly higher amplitude of low-frequency fluctuation in SZ patients than in NCs. Moreover, the alpha diversity of the gut microbiota showed a strong linear relationship with the values of both GMV and ReHo. In SZ patients, the ReHo indexes in the right STC (r = − 0.35, p = 0.031, FDR corrected p = 0.039), the left cuneus (r = − 0.33, p = 0.044, FDR corrected p = 0.053) and the right MTC (r = − 0.34, p = 0.03, FDR corrected p = 0.052) were negatively correlated with the abundance of the genus Roseburia. Our results suggest that the potential role of the gut microbiome in SZ is related to alterations in brain structure and function. This study provides insights into the underlying neuropathology of SZ.
Collapse
|
441
|
Wang C, Oughourlian T, Tishler TA, Anwar F, Raymond C, Pham AD, Perschon A, Villablanca JP, Ventura J, Subotnik KL, Nuechterlein KH, Ellingson BM. Cortical morphometric correlational networks associated with cognitive deficits in first episode schizophrenia. Schizophr Res 2021; 231:179-188. [PMID: 33872855 DOI: 10.1016/j.schres.2021.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/09/2021] [Accepted: 04/07/2021] [Indexed: 12/14/2022]
Abstract
Schizophrenia (SCZ) is a chronic cognitive and behavioral disorder associated with abnormal cortical activity during information processing. Several brain structures associated with the seven performance domains evaluated using the MATRICS (Measurement and Treatment Research to Improve Cognition in Schizophrenia) Consensus Cognitive Battery (MCCB) have shown cortical volume loss in first episode schizophrenia (FES) patients. However, the relationship between morphological organization and MCCB performance remains unclear. Therefore, in the current observational study, high-resolution structural MRI scans were collected from 50 FES patients, and the morphometric correlation network (MCN) using cortical volume was established to characterize the cortical pattern associated with poorer MCCB performance. We also investigated topological properties, such as the modularity, the degree and the betweenness centrality. Our findings show structural volume was directly and strongly associated with the cognitive deficits of FES patients in the precuneus, anterior cingulate, and fusiform gyrus, as well as the prefrontal, parietal, and sensorimotor cortices. The medial orbitofrontal, fusiform, and superior frontal gyri were not only identified as the predominant nodes with high degree and betweenness centrality in the MCN, but they were also found to be critical in performance in several of the MCCB domains. Together, these results suggest a widespread cortical network is altered in FES patients and that performance on the MCCB domains is associated with the core pathophysiology of SCZ.
Collapse
Affiliation(s)
- Chencai Wang
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Talia Oughourlian
- Department of Psychology, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Todd A Tishler
- Dept. of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Faizan Anwar
- Dept. of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Catalina Raymond
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Alex D Pham
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Abby Perschon
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - J Pablo Villablanca
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Joseph Ventura
- Dept. of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Kenneth L Subotnik
- Dept. of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Keith H Nuechterlein
- Dept. of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America; Department of Psychology, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Benjamin M Ellingson
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America; Dept. of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America; Neuroscience Interdisciplinary Graduate Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America.
| |
Collapse
|
442
|
Lee WH, Antoniades M, Schnack HG, Kahn RS, Frangou S. Brain age prediction in schizophrenia: Does the choice of machine learning algorithm matter? Psychiatry Res Neuroimaging 2021; 310:111270. [PMID: 33714090 PMCID: PMC8056405 DOI: 10.1016/j.pscychresns.2021.111270] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/27/2022]
Abstract
Brain-predicted age difference (brainPAD) has been used in schizophrenia to assess individual-level deviation in the biological age of the patients' brain (i.e., brain-age) from normative reference brain structural datasets. There is marked inter-study variation in brainPAD in schizophrenia which is commonly attributed to sample heterogeneity. However, the potential contribution of the different machine learning algorithms used for brain-age estimation has not been systematically evaluated. Here, we aimed to assess variation in brain-age estimated by six commonly used algorithms [ordinary least squares regression, ridge regression, least absolute shrinkage and selection operator regression, elastic-net regression, linear support vector regression, and relevance vector regression] when applied to the same brain structural features from the same sample. To assess reproducibility we used data from two publically available samples of healthy individuals (n = 1092 and n = 492) and two further samples, from the Icahn School of Medicine at Mount Sinai (ISMMS) and the Center of Biomedical Research Excellence (COBRE), comprising both patients with schizophrenia (n = 90 and n = 76) and healthy individuals (n = 200 and n = 87). Performance similarity across algorithms was compared within each sample using correlation analyses and hierarchical clustering. Across all samples ordinary least squares regression, the only algorithm without a penalty term, performed markedly worse. All other algorithms showed comparable performance but they still yielded variable brain-age estimates despite being applied to the same data. Although brainPAD was consistently higher in patients with schizophrenia, it varied by algorithm from 3.8 to 5.2 years in the ISMMS sample and from to 4.5 to 11.7 years in the COBRE sample. Algorithm choice introduces variations in brain-age and may confound inter-study comparisons when assessing brainPAD in schizophrenia.
Collapse
Affiliation(s)
- Won Hee Lee
- Department of Software Convergence, Kyung Hee University, Yongin, Republic of Korea
| | - Mathilde Antoniades
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 20019, United States
| | - Hugo G Schnack
- Department of Psychiatry, UMCU Brain Center, University Medical Center Utrecht, Netherlands
| | - Rene S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 20019, United States
| | - Sophia Frangou
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 20019, United States; Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada.
| |
Collapse
|
443
|
Koshiyama D, Miyakoshi M, Joshi YB, Molina JL, Tanaka-Koshiyama K, Sprock J, Braff DL, Swerdlow NR, Light GA. Neural network dynamics underlying gamma synchronization deficits in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2021; 107:110224. [PMID: 33340619 PMCID: PMC8631608 DOI: 10.1016/j.pnpbp.2020.110224] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/06/2020] [Accepted: 12/09/2020] [Indexed: 01/09/2023]
Abstract
Gamma-band (40-Hz) activity is critical for cortico-cortical transmission and the integration of information across neural networks during sensory and cognitive processing. Patients with schizophrenia show selective reductions in the capacity to support synchronized gamma-band oscillations in response to auditory stimulation presented 40-Hz. Despite widespread application of this 40-Hz auditory steady-state response (ASSR) as a translational electroencephalographic biomarker for therapeutic development for neuropsychiatric disorders, the spatiotemporal dynamics underlying the ASSR have not been fully characterized. In this study, a novel Granger causality analysis was applied to assess the propagation of gamma oscillations in response to 40-Hz steady-state stimulation across cortical sources in schizophrenia patients (n = 426) and healthy comparison subjects (n = 293). Both groups showed multiple ASSR source interactions that were broadly distributed across brain regions. Schizophrenia patients showed distinct, hierarchically sequenced connectivity abnormalities. During the response onset interval, patients exhibited abnormal increased connectivity from the inferior frontal gyrus to the superior temporal gyrus, followed by decreased connectivity from the superior temporal to the middle cingulate gyrus. In the later portion of the ASSR response (300-500 ms), patients showed significantly increased connectivity from the superior temporal to the middle frontal gyrus followed by decreased connectivity from the left superior frontal gyrus to the right superior and middle frontal gyri. These findings highlight both the orchestration of distributed multiple sources in response to simple gamma-frequency stimulation in healthy subjects as well as the patterns of deficits in the generation and maintenance of gamma-band oscillations across the temporo-frontal sources in schizophrenia patients.
Collapse
Affiliation(s)
- Daisuke Koshiyama
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA
| | - Makoto Miyakoshi
- Swartz Center for Neural Computation, University of California San Diego, La Jolla, CA 92093-0559, USA.
| | - Yash B. Joshi
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Juan L. Molina
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA 92161, USA
| | | | - Joyce Sprock
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - David L. Braff
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Neal R. Swerdlow
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA
| | - Gregory A. Light
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093-0804, USA,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, CA 92161, USA
| |
Collapse
|
444
|
Parkes L, Moore TM, Calkins ME, Cook PA, Cieslak M, Roalf DR, Wolf DH, Gur RC, Gur RE, Satterthwaite TD, Bassett DS. Transdiagnostic dimensions of psychopathology explain individuals' unique deviations from normative neurodevelopment in brain structure. Transl Psychiatry 2021; 11:232. [PMID: 33879764 PMCID: PMC8058055 DOI: 10.1038/s41398-021-01342-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/24/2021] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
Psychopathology is rooted in neurodevelopment. However, clinical and biological heterogeneity, together with a focus on case-control approaches, have made it difficult to link dimensions of psychopathology to abnormalities of neurodevelopment. Here, using the Philadelphia Neurodevelopmental Cohort, we built normative models of cortical volume and tested whether deviations from these models better predicted psychiatric symptoms compared to raw cortical volume. Specifically, drawing on the p-factor hypothesis, we distilled 117 clinical symptom measures into six orthogonal psychopathology dimensions: overall psychopathology, anxious-misery, externalizing disorders, fear, positive psychosis symptoms, and negative psychosis symptoms. We found that multivariate patterns of deviations yielded improved out-of-sample prediction of psychopathology dimensions compared to multivariate patterns of raw cortical volume. We also found that correlations between overall psychopathology and deviations in ventromedial prefrontal, inferior temporal, and dorsal anterior cingulate cortices were stronger than those observed for specific dimensions of psychopathology (e.g., anxious-misery). Notably, these same regions are consistently implicated in a range of putatively distinct disorders. Finally, we performed conventional case-control comparisons of deviations in a group of individuals with depression and a group with attention-deficit hyperactivity disorder (ADHD). We observed spatially overlapping effects between these groups that diminished when controlling for overall psychopathology. Together, our results suggest that modeling cortical brain features as deviations from normative neurodevelopment improves prediction of psychiatric symptoms in out-of-sample testing, and that p-factor models of psychopathology may assist in separating biomarkers that are disorder-general from those that are disorder-specific.
Collapse
Affiliation(s)
- Linden Parkes
- Department of Bioengineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tyler M Moore
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Lifespan Brain Institute, University of Pennsylvania & Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Monica E Calkins
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Lifespan Brain Institute, University of Pennsylvania & Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Philip A Cook
- Department of Radiology, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Matthew Cieslak
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Lifespan Brain Institute, University of Pennsylvania & Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David R Roalf
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Lifespan Brain Institute, University of Pennsylvania & Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Daniel H Wolf
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Lifespan Brain Institute, University of Pennsylvania & Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ruben C Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Lifespan Brain Institute, University of Pennsylvania & Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Radiology, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Neurology, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Raquel E Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Lifespan Brain Institute, University of Pennsylvania & Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Radiology, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Neurology, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Theodore D Satterthwaite
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Lifespan Brain Institute, University of Pennsylvania & Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Danielle S Bassett
- Department of Bioengineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Neurology, Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Department of Electrical & Systems Engineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Physics & Astronomy, College of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Santa Fe Institute, Santa Fe, NM, 87501, USA.
| |
Collapse
|
445
|
Stein F, Meller T, Brosch K, Schmitt S, Ringwald K, Pfarr JK, Meinert S, Thiel K, Lemke H, Waltemate L, Grotegerd D, Opel N, Jansen A, Nenadić I, Dannlowski U, Krug A, Kircher T. Psychopathological Syndromes Across Affective and Psychotic Disorders Correlate With Gray Matter Volumes. Schizophr Bull 2021; 47:1740-1750. [PMID: 33860786 PMCID: PMC8530386 DOI: 10.1093/schbul/sbab037] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION More than a century of research on the neurobiological underpinnings of major psychiatric disorders (major depressive disorder [MDD], bipolar disorder [BD], schizophrenia [SZ], and schizoaffective disorder [SZA]) has been unable to identify diagnostic markers. An alternative approach is to study dimensional psychopathological syndromes that cut across categorical diagnoses. The aim of the current study was to identify gray matter volume (GMV) correlates of transdiagnostic symptom dimensions. METHODS We tested the association of 5 psychopathological factors with GMV using multiple regression models in a sample of N = 1069 patients meeting Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM-IV) criteria for MDD (n = 818), BD (n = 132), and SZ/SZA (n = 119). T1-weighted brain images were acquired with 3-Tesla magnetic resonance imaging and preprocessed with CAT12. Interactions analyses (diagnosis × psychopathological factor) were performed to test whether local GMV associations were driven by DSM-IV diagnosis. We further tested syndrome specific regions of interest (ROIs). RESULTS Whole brain analysis showed a significant negative association of the positive formal thought disorder factor with GMV in the right middle frontal gyrus, the paranoid-hallucinatory syndrome in the right fusiform, and the left middle frontal gyri. ROI analyses further showed additional negative associations, including the negative syndrome with bilateral frontal opercula, positive formal thought disorder with the left amygdala-hippocampus complex, and the paranoid-hallucinatory syndrome with the left angular gyrus. None of the GMV associations interacted with DSM-IV diagnosis. CONCLUSIONS We found associations between psychopathological syndromes and regional GMV independent of diagnosis. Our findings open a new avenue for neurobiological research across disorders, using syndrome-based approaches rather than categorical diagnoses.
Collapse
Affiliation(s)
- Frederike Stein
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany,Center for Mind Brain and Behavior, University of Marburg, Marburg, Germany,To whom correspondence should be addressed; Department of Psychiatry and Psychotherapy, Philipps-Universität Marburg, Rudolf-Bultmann-Str. 8, 35039 Marburg, Germany; tel: +49-6421-58-63831, fax: +49-6421-58-68939, e-mail:
| | - Tina Meller
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany,Center for Mind Brain and Behavior, University of Marburg, Marburg, Germany
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany,Center for Mind Brain and Behavior, University of Marburg, Marburg, Germany
| | - Simon Schmitt
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany,Center for Mind Brain and Behavior, University of Marburg, Marburg, Germany
| | - Kai Ringwald
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany,Center for Mind Brain and Behavior, University of Marburg, Marburg, Germany
| | - Julia Katharina Pfarr
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany,Center for Mind Brain and Behavior, University of Marburg, Marburg, Germany
| | - Susanne Meinert
- Department of Psychiatry University of Münster, Münster, Germany
| | - Katharina Thiel
- Department of Psychiatry University of Münster, Münster, Germany
| | - Hannah Lemke
- Department of Psychiatry University of Münster, Münster, Germany
| | - Lena Waltemate
- Department of Psychiatry University of Münster, Münster, Germany
| | | | - Nils Opel
- Department of Psychiatry University of Münster, Münster, Germany
| | - Andreas Jansen
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany,Center for Mind Brain and Behavior, University of Marburg, Marburg, Germany
| | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany,Center for Mind Brain and Behavior, University of Marburg, Marburg, Germany
| | - Udo Dannlowski
- Department of Psychiatry University of Münster, Münster, Germany
| | - Axel Krug
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany,Center for Mind Brain and Behavior, University of Marburg, Marburg, Germany,Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany,Center for Mind Brain and Behavior, University of Marburg, Marburg, Germany
| |
Collapse
|
446
|
D’Ambrosio E, Jauhar S, Kim S, Veronese M, Rogdaki M, Pepper F, Bonoldi I, Kotoula V, Kempton MJ, Turkheimer F, Kwon JS, Kim E, Howes OD. The relationship between grey matter volume and striatal dopamine function in psychosis: a multimodal 18F-DOPA PET and voxel-based morphometry study. Mol Psychiatry 2021; 26:1332-1345. [PMID: 31690805 PMCID: PMC7610423 DOI: 10.1038/s41380-019-0570-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 09/23/2019] [Accepted: 10/23/2019] [Indexed: 01/26/2023]
Abstract
A leading hypothesis for schizophrenia and related psychotic disorders proposes that cortical brain disruption leads to subcortical dopaminergic dysfunction, which underlies psychosis in the majority of patients who respond to treatment. Although supported by preclinical findings that prefrontal cortical lesions lead to striatal dopamine dysregulation, the relationship between prefrontal structural volume and striatal dopamine function has not been tested in people with psychosis. We therefore investigated the in vivo relationship between striatal dopamine synthesis capacity and prefrontal grey matter volume in treatment-responsive patients with psychosis, and compared them to treatment non-responsive patients, where dopaminergic mechanisms are not thought to be central. Forty patients with psychosis across two independent cohorts underwent 18F-DOPA PET scans to measure dopamine synthesis capacity (indexed as the influx rate constant Kicer) and structural 3T MRI. The PET, but not MR, data have been reported previously. Structural images were processed using DARTEL-VBM. GLM analyses were performed in SPM12 to test the relationship between prefrontal grey matter volume and striatal Kicer. Treatment responders showed a negative correlation between prefrontal grey matter and striatal dopamine synthesis capacity, but this was not evident in treatment non-responders. Specifically, we found an interaction between treatment response, whole striatal dopamine synthesis capacity and grey matter volume in left (pFWE corr. = 0.017) and right (pFWE corr. = 0.042) prefrontal cortex. We replicated the finding in right prefrontal cortex in the independent sample (pFWE corr. = 0.031). The summary effect size was 0.82. Our findings are consistent with the long-standing hypothesis of dysregulation of the striatal dopaminergic system being related to prefrontal cortex pathology in schizophrenia, but critically also extend the hypothesis to indicate it can be applied to treatment-responsive schizophrenia only. This suggests that different mechanisms underlie the pathophysiology of treatment-responsive and treatment-resistant schizophrenia.
Collapse
Affiliation(s)
- Enrico D’Ambrosio
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK,Psychiatric Neuroscience Group, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | - Sameer Jauhar
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK,Early Intervention Psychosis Clinical Academic Group, South London & Maudsley NHS Trust, London
| | - Seoyoung Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Maria Rogdaki
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK,Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London, W12 0NN, UK
| | - Fiona Pepper
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Ilaria Bonoldi
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Vasileia Kotoula
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Matthew J Kempton
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Jun Soo Kwon
- Department of Brain & Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea,Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Euitae Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea. .,Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Oliver D Howes
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK. .,Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London, W12 0NN, UK.
| |
Collapse
|
447
|
Karcher NR, Schiffman J, Barch DM. Environmental Risk Factors and Psychotic-like Experiences in Children Aged 9-10. J Am Acad Child Adolesc Psychiatry 2021; 60:490-500. [PMID: 32682894 PMCID: PMC7895444 DOI: 10.1016/j.jaac.2020.07.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/30/2020] [Accepted: 07/09/2020] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Research implicates environmental risk factors, including correlates of urbanicity, deprivation, and environmental toxins, in psychotic-like experiences (PLEs). The current study examined associations between several types of environmental risk factors and PLEs in school-age children, whether these associations were specific to PLEs or generalized to other psychopathology, and examined possible neural mechanisms for significant associations. METHOD The current study used cross-sectional data from 10,328 children 9-10 years old from the Adolescent Brain Cognitive Development (ABCD) Study. Hierarchical linear models examined associations between PLEs and geocoded environmental risk factors and whether associations generalized to internalizing/externalizing symptoms. Mediation models examined evidence of structural magnetic resonance imaging abnormalities (eg, intracranial volume) potentially mediating associations between PLEs and environmental risk factors. RESULTS Specific types of environmental risk factors, namely, measures of urbanicity (eg, drug offense exposure, less perception of neighborhood safety), deprivation (eg, overall deprivation, poverty rate), and lead exposure risk, were associated with PLEs. These associations showed evidence of stronger associations with PLEs than internalizing/externalizing symptoms (especially overall deprivation, poverty, drug offense exposure, and lead exposure risk). There was evidence that brain volume mediated between 11% and 25% of associations of poverty, perception of neighborhood safety, and lead exposure risk with PLEs. CONCLUSION Although in the context of cross-sectional analyses, this evidence is consistent with neural measures partially mediating the association between PLEs and environmental exposures. This study also replicated and extended recent findings of associations between PLEs and environmental exposures, finding evidence for specific associations with correlates of urbanicity, deprivation, and lead exposure risk.
Collapse
Affiliation(s)
| | - Jason Schiffman
- University of Maryland, Baltimore County; University of California, Irvine
| | | |
Collapse
|
448
|
Hong SJ, Sisk LM, Caballero C, Mekhanik A, Roy AK, Milham MP, Gee DG. Decomposing complex links between the childhood environment and brain structure in school-aged youth. Dev Cogn Neurosci 2021; 48:100919. [PMID: 33556882 PMCID: PMC7868609 DOI: 10.1016/j.dcn.2021.100919] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 10/26/2020] [Accepted: 01/18/2021] [Indexed: 12/22/2022] Open
Abstract
Childhood experiences play a profound role in conferring risk and resilience for brain and behavioral development. However, how different facets of the environment shape neurodevelopment remains largely unknown. Here we sought to decompose heterogeneous relationships between environmental factors and brain structure in 989 school-aged children from the Adolescent Brain Cognitive Development Study. We applied a cross-modal integration and clustering approach called 'Similarity Network Fusion', which combined two brain morphometrics (i.e., cortical thickness and myelin-surrogate markers), and key environmental factors (i.e., trauma exposure, neighborhood safety, school environment, and family environment) to identify homogeneous subtypes. Depending on the subtyping resolution, results identified two or five subgroups, each characterized by distinct brain structure-environment profiles. Notably, more supportive caregiving and school environments were associated with greater myelination, whereas less supportive caregiving, higher family conflict and psychopathology, and higher perceived neighborhood safety were observed with greater cortical thickness. These subtypes were highly reproducible and predicted externalizing symptoms and overall mental health problems. Our findings support the theory that distinct environmental exposures are differentially associated with alterations in structural neurodevelopment. Delineating more precise associations between risk factors, protective factors, and brain development may inform approaches to enhance risk identification and optimize interventions targeting specific experiences.
Collapse
Affiliation(s)
- Seok-Jun Hong
- Center for the Developing Brain, Child Mind Institute, New York, NY, USA; Center for Neuroscience Imaging Research, Institute for Basic Science, Sungkyunkwan University, Suwon, South Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon, South Korea
| | - Lucinda M Sisk
- Department of Psychology, Yale University, New Haven, CT, USA
| | | | - Anthony Mekhanik
- Center for the Developing Brain, Child Mind Institute, New York, NY, USA
| | - Amy K Roy
- Department of Psychology, Fordham University, Bronx, NY, USA
| | - Michael P Milham
- Center for the Developing Brain, Child Mind Institute, New York, NY, USA; Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute, Orangeburg, NY, USA
| | - Dylan G Gee
- Department of Psychology, Yale University, New Haven, CT, USA.
| |
Collapse
|
449
|
Reijonen J, Könönen M, Tuunanen P, Määttä S, Julkunen P. Atlas-informed computational processing pipeline for individual targeting of brain areas for therapeutic navigated transcranial magnetic stimulation. Clin Neurophysiol 2021; 132:1612-1621. [PMID: 34030058 DOI: 10.1016/j.clinph.2021.01.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/06/2021] [Accepted: 01/29/2021] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Navigated transcranial magnetic stimulation (nTMS) is targeted at different cortical sites for diagnostic, therapeutic, and neuroscientific purposes. Correct identification of the cortical target areas is important for achieving desired effects, but it is challenging when no direct responses arise upon target area stimulation. We aimed at utilizing atlas-based marking of cortical areas for nTMS targeting to present a convenient, rater-independent method for overlaying the individual target sites with brain anatomy. METHODS We developed a pipeline, which fits a brain atlas to the individual brain and enables visualization of the target areas during the nTMS session. We applied the pipeline to our previous nTMS data, focusing on depression and schizophrenia patients. Furthermore, we included examples of Tourette syndrome and tinnitus therapies, as well as neurosurgical and motor mappings. RESULTS In depression and schizophrenia patients, the visually selected dorsolateral prefrontal cortex (DLPFC) targets were close to the border between atlas areas A9/46 and A8. In the other areas, the atlas-based areas were in agreement with the treatment targets. CONCLUSIONS The atlas-based target areas agreed well with the cortical targets selected by experts during the treatments. SIGNIFICANCE Overlaying atlas information over the navigation view is a convenient and useful add-on for improving nTMS targeting.
Collapse
Affiliation(s)
- Jusa Reijonen
- Department of Clinical Neurophysiology, Kuopio University Hospital, Kuopio, Finland; Department of Applied Physics, University of Eastern Finland, Kuopio, Finland.
| | - Mervi Könönen
- Department of Clinical Radiology, Kuopio University Hospital, Kuopio, Finland
| | - Pasi Tuunanen
- Department of Clinical Neurophysiology, Kuopio University Hospital, Kuopio, Finland
| | - Sara Määttä
- Department of Clinical Neurophysiology, Kuopio University Hospital, Kuopio, Finland
| | - Petro Julkunen
- Department of Clinical Neurophysiology, Kuopio University Hospital, Kuopio, Finland; Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
450
|
Genetic factors influencing a neurobiological substrate for psychiatric disorders. Transl Psychiatry 2021; 11:192. [PMID: 33782385 PMCID: PMC8007575 DOI: 10.1038/s41398-021-01317-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/07/2021] [Accepted: 01/20/2021] [Indexed: 02/05/2023] Open
Abstract
A retrospective meta-analysis of magnetic resonance imaging voxel-based morphometry studies proposed that reduced gray matter volumes in the dorsal anterior cingulate and the left and right anterior insular cortex-areas that constitute hub nodes of the salience network-represent a common substrate for major psychiatric disorders. Here, we investigated the hypothesis that the common substrate serves as an intermediate phenotype to detect genetic risk variants relevant for psychiatric disease. To this end, after a data reduction step, we conducted genome-wide association studies of a combined common substrate measure in four population-based cohorts (n = 2271), followed by meta-analysis and replication in a fifth cohort (n = 865). After correction for covariates, the heritability of the common substrate was estimated at 0.50 (standard error 0.18). The top single-nucleotide polymorphism (SNP) rs17076061 was associated with the common substrate at genome-wide significance and replicated, explaining 1.2% of the common substrate variance. This SNP mapped to a locus on chromosome 5q35.2 harboring genes involved in neuronal development and regeneration. In follow-up analyses, rs17076061 was not robustly associated with psychiatric disease, and no overlap was found between the broader genetic architecture of the common substrate and genetic risk for major depressive disorder, bipolar disorder, or schizophrenia. In conclusion, our study identified that common genetic variation indeed influences the common substrate, but that these variants do not directly translate to increased disease risk. Future studies should investigate gene-by-environment interactions and employ functional imaging to understand how salience network structure translates to psychiatric disorder risk.
Collapse
|