101
|
Sun Y, Zhang Z, Kakkos I, Matsopoulos GK, Yuan J, Suckling J, Xu L, Cao S, Chen W, Hu X, Li T, Sim K, Qi P, Sun Y. Inferring the Individual Psychopathologic Deficits with Structural Connectivity in a Longitudinal Cohort of Schizophrenia. IEEE J Biomed Health Inform 2022; 26:2536-2546. [PMID: 34982705 DOI: 10.1109/jbhi.2021.3139701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The prediction of schizophrenia-related psychopathologic deficits is exceedingly important in the fields of psychiatry and clinical practice. However, objective association of the brain structure alterations to the illness clinical symptoms is challenging. Although, schizophrenia has been characterized as a brain dysconnectivity syndrome, evidence accounting for neuroanatomical network alterations remain scarce. Moreover, the absence of generalized connectome biomarkers for the assessment of illness progression further perplexes the prediction of long-term symptom severity. In this paper, a combination of individualized prediction models with quantitative graph theoretical analysis was adopted, providing a comprehensive appreciation of the extent to which the brain network properties are affected over time in schizophrenia. Specifically, Connectome-based Prediction Models were employed on Structural Connectivity (SC) features, efficiently capturing individual network-related differences, while identifying the anatomical connectivity disturbances contributing to the prediction of psychopathological deficits. Our results demonstrated distinctions among widespread cortical circuits responsible for different domains of symptoms, indicating the complex neural mechanisms underlying schizophrenia. Furthermore, the generated models were able to significantly predict changes of symptoms using SC features at follow-up, while the preserved SC features suggested an association with improved positive and overall symptoms. Moreover, cross-sectional significant deficits were observed in network efficiency and a progressive aberration of global integration in patients compared to healthy controls, representing a group-consensus pathological map, while supporting the dysconnectivity hypothesis.
Collapse
|
102
|
Shen X, Jiang F, Fang X, Yan W, Xie S, Zhang R. Cognitive dysfunction and cortical structural abnormalities in first-episode drug-naïve schizophrenia patients with auditory verbal hallucination. Front Psychiatry 2022; 13:998807. [PMID: 36186860 PMCID: PMC9523744 DOI: 10.3389/fpsyt.2022.998807] [Citation(s) in RCA: 2] [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: 07/20/2022] [Accepted: 08/24/2022] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE The current study aimed to examine the cognitive profiles and cortical structural alterations in first-episode drug-naïve schizophrenia with AVH (auditory verbal hallucination). METHODS Cortical structural parameters including cortical thickness and local gyrification index (LGI) estimated using FreeSurfer as well as cognitive performance assessed on the MATRICS Consensus Cognitive Battery (MCCB) were acquired from 78 schizophrenia patients with AVH, 74 schizophrenia patients without AVH (non-AVH), and 76 healthy controls (HC). Hoffman Auditory Hallucination Rating Scale (HAHRS) was applied to assess the severity of AVH. RESULTS The results revealed extensive deficits in all cognitive domains among AVH, non-AVH, and HC groups. Compared to non-AVH group, the AVH group showed poorer performance on visual learning and verbal learning domains. There were six brain regions with cortical thinning in the right hemisphere of inferior temporal gyrus, superior temporal gyrus, lateral orbito frontal cortex, rostral anterior cingulate cortex, supramarginal gyrus and insula, and two brain regions with increased LGI in the left hemisphere of superior parietal gyrus and the right hemisphere of caudal anterior cingulate cortex on AVH group relative to non-AVH group. Correlation analysis revealed that the cortical thickness in the right hemisphere of lateral orbito frontal cortex was negatively correlated with the severity of AVH in schizophrenia patients with AVH. CONCLUSION Visual learning, verbal learning dysfunction, and specific disruption of cortical structure may characterize schizophrenia patients with AVH during early stages of the disorder. Right lateral orbito frontal cortical deficits may be the pathological mechanisms underlying AVH in first-episode drug-naïve schizophrenia.
Collapse
Affiliation(s)
- Xuran Shen
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Fuli Jiang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyu Fang
- 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
| | - Shiping Xie
- Department of Psychiatry, 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
| |
Collapse
|
103
|
Xue K, Chen J, Wei Y, Chen Y, Han S, Wang C, Zhang Y, Song X, Cheng J. Altered dynamic functional connectivity of auditory cortex and medial geniculate nucleus in first-episode, drug-naïve schizophrenia patients with and without auditory verbal hallucinations. Front Psychiatry 2022; 13:963634. [PMID: 36159925 PMCID: PMC9489854 DOI: 10.3389/fpsyt.2022.963634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/18/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND AND OBJECTIVE As a key feature of schizophrenia, auditory verbal hallucination (AVH) is causing concern. Altered dynamic functional connectivity (dFC) patterns involving in auditory related regions were rarely reported in schizophrenia patients with AVH. The goal of this research was to find out the dFC abnormalities of auditory related regions in first-episode, drug-naïve schizophrenia patients with and without AVH using resting state functional magnetic resonance imaging (rs-fMRI). METHODS A total of 107 schizophrenia patients with AVH, 85 schizophrenia patients without AVH (NAVH) underwent rs-fMRI examinations, and 104 healthy controls (HC) were matched. Seed-based dFC of the primary auditory cortex (Heschl's gyrus, HES), auditory association cortex (AAC, including Brodmann's areas 22 and 42), and medial geniculate nucleus (MGN) was conducted to build a whole-brain dFC diagram, then inter group comparison and correlation analysis were performed. RESULTS In comparison to the NAVH and HC groups, the AVH group showed increased dFC from left ACC to the right middle temporal gyrus and right middle occipital gyrus, decreased dFC from left HES to the left superior occipital gyrus, left cuneus gyrus, left precuneus gyrus, decreased dFC from right HES to the posterior cingulate gyrus, and decreased dFC from left MGN to the bilateral calcarine gyrus, bilateral cuneus gyrus, bilateral lingual gyrus. The Auditory Hallucination Rating Scale (AHRS) was significantly positively correlated with the dFC values of cluster 1 (bilateral calcarine gyrus, cuneus gyrus, lingual gyrus, superior occipital gyrus, precuneus gyrus, and posterior cingulate gyrus) using left AAC seed, cluster 2 (right middle temporal gyrus and right middle occipital gyrus) using left AAC seed, cluster 1 (bilateral calcarine gyrus, cuneus gyrus, lingual gyrus, superior occipital gyrus, precuneus gyrus and posterior cingulate gyrus) using right AAC seed and cluster 2 (posterior cingulate gyrus) using right HES seed in the AVH group. In both AVH and NAVH groups, a significantly negative correlation is also found between the dFC values of cluster 2 (posterior cingulate gyrus) using the right HES seed and the PANSS negative sub-scores. CONCLUSIONS The present findings demonstrate that schizophrenia patients with AVH showed multiple abnormal dFC regions using auditory related cortex and nucleus as seeds, particularly involving the occipital lobe, default mode network (DMN), and middle temporal lobe, implying that the different dFC patterns of auditory related areas could provide a neurological mechanism of AVH in schizophrenia.
Collapse
Affiliation(s)
- Kangkang Xue
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingli Chen
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yarui Wei
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuan Chen
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shaoqiang Han
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Caihong Wang
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yong Zhang
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xueqin Song
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
104
|
Dupuy M, Abdallah M, Swendsen J, N’Kaoua B, Chanraud S, Schweitzer P, Fatseas M, Serre F, Barse E, Auriacombe M, Misdrahi D. Real-time cognitive performance and positive symptom expression in schizophrenia. Eur Arch Psychiatry Clin Neurosci 2022; 272:415-425. [PMID: 34287696 PMCID: PMC8938338 DOI: 10.1007/s00406-021-01296-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 07/04/2021] [Indexed: 11/28/2022]
Abstract
Deficits in cognitive functions are frequent in schizophrenia and are often conceptualized as stable characteristics of this disorder. However, cognitive capacities may fluctuate over the course of a day and it is unknown if such variation may be linked to the dynamic expression of psychotic symptoms. This investigation used Ecological Momentary Assessment (EMA) to provide mobile tests of cognitive functions and positive symptoms in real time. Thirty-three individuals with schizophrenia completed five EMA assessments per day for a one-week period that included real-time assessments of cognitive performance and psychotic symptoms. A subsample of patients and 31 healthy controls also completed a functional MRI examination. Relative to each individual's average score, moments of worsened cognitive performance on the mobile tests were associated with an increased probability of positive symptom occurrence over subsequent hours of the day (coef = 0.06, p < 0.05), adjusting for the presence of psychotic symptoms at the moment of mobile test administration. These prospective associations varied as a function of graph theory indices in MRI analyses. These findings demonstrate that cognitive performance is prospectively linked to psychotic symptom expression in daily life, and that underlying brain markers may be observed in the Executive Control Network. While the potential causal nature of this association remains to be investigated, our results offer promising prospects for a better understanding of the underlying mechanisms of symptom expression in schizophrenia.
Collapse
Affiliation(s)
- Maud Dupuy
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), University of Bordeaux/CNRS-UMR 5287, Bordeaux, France.
| | - Majd Abdallah
- Institut de Neurosciences Cognitives et Intégratives d’Aquitaine (INCIA), University of Bordeaux/CNRS-UMR 5287, Bordeaux, France
| | - Joel Swendsen
- Institut de Neurosciences Cognitives et Intégratives d’Aquitaine (INCIA), University of Bordeaux/CNRS-UMR 5287, Bordeaux, France ,EPHE, PSL Research University, Paris, France
| | - Bernard N’Kaoua
- Handicap, Activity, Cognition, Health, Inserm/University of Bordeaux, Talence, France
| | - Sandra Chanraud
- Institut de Neurosciences Cognitives et Intégratives d’Aquitaine (INCIA), University of Bordeaux/CNRS-UMR 5287, Bordeaux, France ,EPHE, PSL Research University, Paris, France
| | - Pierre Schweitzer
- Institut de Neurosciences Cognitives et Intégratives d’Aquitaine (INCIA), University of Bordeaux/CNRS-UMR 5287, Bordeaux, France
| | - Melina Fatseas
- Institut de Neurosciences Cognitives et Intégratives d’Aquitaine (INCIA), University of Bordeaux/CNRS-UMR 5287, Bordeaux, France ,CHU Bordeaux, Bordeaux, France
| | - Fuschia Serre
- Addiction and Neuropsychiatry (SANPSY), University of Bordeaux, CNRS USR 3413 – Sleep, Bordeaux, France
| | - Elodie Barse
- Institut de Neurosciences Cognitives et Intégratives d’Aquitaine (INCIA), University of Bordeaux/CNRS-UMR 5287, Bordeaux, France ,EPHE, PSL Research University, Paris, France
| | - Marc Auriacombe
- Addiction and Neuropsychiatry (SANPSY), University of Bordeaux, CNRS USR 3413 – Sleep, Bordeaux, France ,CH Charles Perrens, Bordeaux, France ,CHU Bordeaux, Bordeaux, France
| | - David Misdrahi
- Institut de Neurosciences Cognitives et Intégratives d’Aquitaine (INCIA), University of Bordeaux/CNRS-UMR 5287, Bordeaux, France ,CH Charles Perrens, Bordeaux, France
| |
Collapse
|
105
|
Lin G, Wu Z, Chen B, Zhang M, Wang Q, Liu M, Zhang S, Yang M, Ning Y, Zhong X. Altered Microstate Dynamics and Spatial Complexity in Late-Life Schizophrenia. Front Psychiatry 2022; 13:907802. [PMID: 35832599 PMCID: PMC9271628 DOI: 10.3389/fpsyt.2022.907802] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Resting-state EEG microstate and omega complexity analyses have been widely used to explore deviant brain function in various neuropsychiatric disorders. This study aimed to investigate the features of microstate dynamics and spatial complexity in patients with late-life schizophrenia (LLS). METHOD Microstate and omega complexity analyses were performed on resting-state EEG data from 39 in patients with LLS and compared with 40 elderly normal controls (NCs). RESULT The duration of microstate classes A and D were significantly higher in patients with LLS compared with NCs. The occurrence of microstate classes A, B, and C was significantly lower in patients with LLS compared with NCs. LLS patients have a lower time coverage of microstate class A and a higher time coverage of class D than NCs. Transition probabilities from microstate class A to B and from class A to C were significantly lower in patients with LLS compared with NCs. Transition probabilities between microstate class B and D were significantly higher in patients with LLS compared with NCs. Global omega complexity and anterior omega complexity were significantly higher in patients with LLS compared with NCs. CONCLUSION This study revealed an altered pattern of microstate dynamics and omega complexity in patients with LLS. This may reflect the disturbed neural basis underlying LLS and enhance the understanding of the pathophysiology of LLS.
Collapse
Affiliation(s)
- Gaohong Lin
- Center for Geriatric Neuroscience, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhangying Wu
- Center for Geriatric Neuroscience, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ben Chen
- Center for Geriatric Neuroscience, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Min Zhang
- Center for Geriatric Neuroscience, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qiang Wang
- Center for Geriatric Neuroscience, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Meiling Liu
- Center for Geriatric Neuroscience, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Si Zhang
- Center for Geriatric Neuroscience, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingfeng Yang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yuping Ning
- Center for Geriatric Neuroscience, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China.,The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Xiaomei Zhong
- Center for Geriatric Neuroscience, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
106
|
Aberrant maturation and connectivity of prefrontal cortex in schizophrenia-contribution of NMDA receptor development and hypofunction. Mol Psychiatry 2022; 27:731-743. [PMID: 34163013 PMCID: PMC8695640 DOI: 10.1038/s41380-021-01196-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023]
Abstract
The neurobiology of schizophrenia involves multiple facets of pathophysiology, ranging from its genetic basis over changes in neurochemistry and neurophysiology, to the systemic level of neural circuits. Although the precise mechanisms associated with the neuropathophysiology remain elusive, one essential aspect is the aberrant maturation and connectivity of the prefrontal cortex that leads to complex symptoms in various stages of the disease. Here, we focus on how early developmental dysfunction, especially N-methyl-D-aspartate receptor (NMDAR) development and hypofunction, may lead to the dysfunction of both local circuitry within the prefrontal cortex and its long-range connectivity. More specifically, we will focus on an "all roads lead to Rome" hypothesis, i.e., how NMDAR hypofunction during development acts as a convergence point and leads to local gamma-aminobutyric acid (GABA) deficits and input-output dysconnectivity in the prefrontal cortex, which eventually induce cognitive and social deficits. Many outstanding questions and hypothetical mechanisms are listed for future investigations of this intriguing hypothesis that may lead to a better understanding of the aberrant maturation and connectivity associated with the prefrontal cortex.
Collapse
|
107
|
Stuke H. Markers of muscarinic deficit for individualized treatment in schizophrenia. Front Psychiatry 2022; 13:1100030. [PMID: 36699495 PMCID: PMC9868756 DOI: 10.3389/fpsyt.2022.1100030] [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: 11/16/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
Recent clinical studies have shown that agonists at muscarinic acetylcholine receptors effectively reduce schizophrenia symptoms. It is thus conceivable that, for the first time, a second substance class of procholinergic antipsychotics could become established alongside the usual antidopaminergic antipsychotics. In addition, various basic science studies suggest that there may be a subgroup of schizophrenia in which hypofunction of muscarinic acetylcholine receptors is of etiological importance. This could represent a major opportunity for individualized treatment of schizophrenia if markers can be identified that predict response to procholinergic vs. antidopaminergic interventions. In this perspective, non-response to antidopaminergic antipsychotics, specific symptom patterns like visual hallucinations and strong disorganization, the presence of antimuscarinic antibodies, ERP markers such as mismatch negativity, and radiotracers are presented as possible in vivo markers of muscarinic deficit and thus potentially of response to procholinergic therapeutics. Finally, open questions and further research steps are outlined.
Collapse
Affiliation(s)
- Heiner Stuke
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, Berlin, Germany
| |
Collapse
|
108
|
di Hou M, Santoro V, Biondi A, Shergill SS, Premoli I. A systematic review of TMS and neurophysiological biometrics in patients with schizophrenia. J Psychiatry Neurosci 2021; 46:E675-E701. [PMID: 34933940 PMCID: PMC8695525 DOI: 10.1503/jpn.210006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 08/06/2021] [Accepted: 09/06/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Transcranial magnetic stimulation can be combined with electromyography (TMS-EMG) and electroencephalography (TMS-EEG) to evaluate the excitatory and inhibitory functions of the cerebral cortex in a standardized manner. It has been postulated that schizophrenia is a disorder of functional neural connectivity underpinned by a relative imbalance of excitation and inhibition. The aim of this review was to provide a comprehensive overview of TMS-EMG and TMS-EEG research in schizophrenia, focused on excitation or inhibition, connectivity, motor cortical plasticity and the effect of antipsychotic medications, symptom severity and illness duration on TMS-EMG and TMS-EEG indices. METHODS We searched PsycINFO, Embase and Medline, from database inception to April 2020, for studies that included TMS outcomes in patients with schizophrenia. We used the following combination of search terms: transcranial magnetic stimulation OR tms AND interneurons OR glutamic acid OR gamma aminobutyric acid OR neural inhibition OR pyramidal neurons OR excita* OR inhibit* OR GABA* OR glutam* OR E-I balance OR excitation-inhibition balance AND schizoaffective disorder* OR Schizophrenia OR schizophreni*. RESULTS TMS-EMG and TMS-EEG measurements revealed deficits in excitation or inhibition, functional connectivity and motor cortical plasticity in patients with schizophrenia. Increased duration of the cortical silent period (a TMS-EMG marker of γ-aminobutyric acid B receptor activity) with clozapine was a relatively consistent finding. LIMITATIONS Most of the studies used patients with chronic schizophrenia and medicated patients, employed cross-sectional group comparisons and had small sample sizes. CONCLUSION TMS-EMG and TMS-EEG offer an opportunity to develop a novel and improved understanding of the physiologic processes that underlie schizophrenia and to assess the therapeutic effect of antipsychotic medications. In the future, these techniques may also help predict disease progression and further our understanding of the excitatory/inhibitory balance and its implications for mechanisms that underlie treatment-resistant schizophrenia.
Collapse
Affiliation(s)
- Meng di Hou
- From the Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK (Hou, Shergill); the Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK (Santoro, Biondi, Premoli); and the Kent and Medway Medical School, Canterbury, UK (Shergill)
| | | | | | | | | |
Collapse
|
109
|
Robles DA, Boreland AJ, Pang ZP, Zahn JD. A Cerebral Organoid Connectivity Apparatus to Model Neuronal Tract Circuitry. MICROMACHINES 2021; 12:1574. [PMID: 34945423 PMCID: PMC8706388 DOI: 10.3390/mi12121574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 11/17/2022]
Abstract
Mental disorders have high prevalence, but the efficacy of existing therapeutics is limited, in part, because the pathogenic mechanisms remain enigmatic. Current models of neural circuitry include animal models and post-mortem brain tissue, which have allowed enormous progress in understanding the pathophysiology of mental disorders. However, these models limit the ability to assess the functional alterations in short-range and long-range network connectivity between brain regions that are implicated in many mental disorders, e.g., schizophrenia and autism spectrum disorders. This work addresses these limitations by developing an in vitro model of the human brain that models the in vivo cerebral tract environment. In this study, microfabrication and stem cell differentiation techniques were combined to develop an in vitro cerebral tract model that anchors human induced pluripotent stem cell-derived cerebral organoids (COs) and provides a scaffold to promote the formation of a functional connecting neuronal tract. Two designs of a Cerebral Organoid Connectivity Apparatus (COCA) were fabricated using SU-8 photoresist. The first design contains a series of spikes which anchor the CO to the COCA (spiked design), whereas the second design contains flat supporting structures with open holes in a grid pattern to anchor the organoids (grid design); both designs allow effective media exchange. Morphological and functional analyses reveal the expression of key neuronal markers as well as functional activity and signal propagation along cerebral tracts connecting CO pairs. The reported in vitro models enable the investigation of critical neural circuitry involved in neurodevelopmental processes and has the potential to help devise personalized and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Denise A. Robles
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA;
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, USA; (A.J.B.); (Z.P.P.)
| | - Andrew J. Boreland
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, USA; (A.J.B.); (Z.P.P.)
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Zhiping P. Pang
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, USA; (A.J.B.); (Z.P.P.)
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
- Pediatrics, Robert Wood Johnson Medical School, Rutgers University, One Robert Wood Johnson Place, MEB, New Brunswick, NJ 08903, USA
| | - Jeffrey D. Zahn
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA;
| |
Collapse
|
110
|
Maly IV, Morales MJ, Pletnikov MV. Astrocyte Bioenergetics and Major Psychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2021; 26:173-227. [PMID: 34888836 DOI: 10.1007/978-3-030-77375-5_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ongoing research continues to add new elements to the emerging picture of involvement of astrocyte energy metabolism in the pathophysiology of major psychiatric disorders, including schizophrenia, mood disorders, and addictions. This review outlines what is known about the energy metabolism in astrocytes, the most numerous cell type in the brain, and summarizes the recent work on how specific perturbations of astrocyte bioenergetics may contribute to the neuropsychiatric conditions. The role of astrocyte energy metabolism in mental health and disease is reviewed on the organism, organ, and cell level. Data arising from genomic, metabolomic, in vitro, and neurobehavioral studies is critically analyzed to suggest future directions in research and possible metabolism-focused therapeutic interventions.
Collapse
Affiliation(s)
- Ivan V Maly
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| | - Michael J Morales
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| | - Mikhail V Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA.
| |
Collapse
|
111
|
Shao X, Liao Y, Gu L, Chen W, Tang J. The Etiology of Auditory Hallucinations in Schizophrenia: From Multidimensional Levels. Front Neurosci 2021; 15:755870. [PMID: 34858129 PMCID: PMC8632545 DOI: 10.3389/fnins.2021.755870] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/14/2021] [Indexed: 11/25/2022] Open
Abstract
Enormous efforts have been made to unveil the etiology of auditory hallucinations (AHs), and multiple genetic and neural factors have already been shown to have their own roles. Previous studies have shown that AHs in schizophrenia vary from those in other disorders, suggesting that they have unique features and possibly distinguishable mechanisms worthy of further investigation. In this review, we intend to offer a comprehensive summary of current findings related to AHs in schizophrenia from aspects of genetics and transcriptome, neurophysiology (neurometabolic and electroencephalogram studies), and neuroimaging (structural and functional magnetic resonance imaging studies and transcriptome–neuroimaging association study). Main findings include gene polymorphisms, glutamate level change, electroencephalographic alterations, and abnormalities of white matter fasciculi, cortical structure, and cerebral activities, especially in multiple regions, including auditory and language networks. More solid and comparable research is needed to replicate and integrate ongoing findings from multidimensional levels.
Collapse
Affiliation(s)
- Xu Shao
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanhui Liao
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lin Gu
- RIKEN AIP, Tokyo, Japan.,Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Wei Chen
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinsong Tang
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
112
|
Stauffer EM, Bethlehem RAI, Warrier V, Murray GK, Romero-Garcia R, Seidlitz J, Bullmore ET. Grey and white matter microstructure is associated with polygenic risk for schizophrenia. Mol Psychiatry 2021; 26:7709-7718. [PMID: 34462574 PMCID: PMC8872982 DOI: 10.1038/s41380-021-01260-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 02/07/2023]
Abstract
Recent discovery of approximately 270 common genetic variants associated with schizophrenia has enabled polygenic risk scores (PRS) to be measured in the population. We hypothesized that normal variation in PRS would be associated with magnetic resonance imaging (MRI) phenotypes of brain morphometry and tissue composition. We used the largest extant genome-wide association dataset (N = 69,369 cases and N = 236,642 healthy controls) to measure PRS for schizophrenia in a large sample of adults from the UK Biobank (Nmax = 29,878) who had multiple micro- and macrostructural MRI metrics measured at each of 180 cortical areas, seven subcortical structures, and 15 major white matter tracts. Linear mixed-effect models were used to investigate associations between PRS and brain structure at global and regional scales, controlled for multiple comparisons. Polygenic risk was significantly associated with reduced neurite density index (NDI) at global brain scale, at 149 cortical regions, five subcortical structures, and 14 white matter tracts. Other microstructural parameters, e.g., fractional anisotropy, that were correlated with NDI were also significantly associated with PRS. Genetic effects on multiple MRI phenotypes were co-located in temporal, cingulate, and prefrontal cortical areas, insula, and hippocampus. Post-hoc bidirectional Mendelian randomization analyses provided preliminary evidence in support of a causal relationship between (reduced) thalamic NDI and (increased) risk of schizophrenia. Risk-related reduction in NDI is plausibly indicative of reduced density of myelinated axons and dendritic arborization in large-scale cortico-subcortical networks. Cortical, subcortical, and white matter microstructure may be linked to the genetic mechanisms of schizophrenia.
Collapse
Affiliation(s)
- Eva-Maria Stauffer
- Department of Psychiatry, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK.
| | - Richard A I Bethlehem
- Department of Psychiatry, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Varun Warrier
- Department of Psychiatry, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Graham K Murray
- Department of Psychiatry, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Trust, Elizabeth House, Fulbourn Hospital, Cambridge, UK
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD, Australia
| | - Rafael Romero-Garcia
- Department of Psychiatry, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Jakob Seidlitz
- Department of Child and Adolescent Psychiatry and Behavioral Science, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward T Bullmore
- Department of Psychiatry, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Trust, Elizabeth House, Fulbourn Hospital, Cambridge, UK
| |
Collapse
|
113
|
Zuccoli GS, Reis-de-Oliveira G, Garbes B, Falkai P, Schmitt A, Nakaya HI, Martins-de-Souza D. Linking proteomic alterations in schizophrenia hippocampus to NMDAr hypofunction in human neurons and oligodendrocytes. Eur Arch Psychiatry Clin Neurosci 2021; 271:1579-1586. [PMID: 33751207 DOI: 10.1007/s00406-021-01248-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 02/24/2021] [Indexed: 12/12/2022]
Abstract
Glutamatergic neurotransmission dysfunction and the early involvement of the hippocampus have been proposed to be important aspects of the pathophysiology of schizophrenia. Here, we performed proteomic analysis of hippocampus postmortem samples from schizophrenia patients as well as neural cells-neurons and oligodendrocytes-treated with MK-801, an NMDA receptor antagonist. There were similarities in processes such as oxidative stress and apoptotic process when comparing hippocampus samples with MK-801-treated neurons, and in proteins synthesis when comparing hippocampus samples with MK-801-treated oligodendrocytes. This reveals that studying the effects of glutamatergic dysfunction in different neural cells can contribute to a better understanding of what it is observed in schizophrenia patients' postmortem brains.
Collapse
Affiliation(s)
- Giuliana S Zuccoli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Guilherme Reis-de-Oliveira
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Bruna Garbes
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig Maximillian University of Munich (LMU), Munich, Germany
| | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, Ludwig Maximillian University of Munich (LMU), Munich, Germany
| | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil.
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil. .,Instituto Nacional de Biomarcadores Em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Científico E Tecnológico, São Paulo, Brasil. .,Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, Brazil. .,D'Or Institute for Research and Education (IDOR), São Paulo, Brazil.
| |
Collapse
|
114
|
Muthesius A, Grothey F, Cunningham C, Hölzer S, Vogeley K, Schultz J. Preserved metacognition despite impaired perception of intentionality cues in schizophrenia. SCHIZOPHRENIA RESEARCH-COGNITION 2021; 27:100215. [PMID: 34692428 PMCID: PMC8517602 DOI: 10.1016/j.scog.2021.100215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 11/03/2022]
Abstract
Social cognition and metacognition are frequently impaired in schizophrenia, and these impairments complicate recovery. Recent work suggests that different aspects of metacognition may not be impaired to the same degree. Furthermore, metacognition and the cognitive capacity being monitored need not be similarly impaired. Here, we assessed performance in detecting cues of intentional behaviour as well as metacognition about detecting those cues in schizophrenia. Thirty patients and controls categorized animations of moving dots into those displaying a dyadic interaction demonstrating a chase or no chase and indicated their confidence in these judgments. Perception and metacognition were assessed using signal detection theoretic measures, which were analysed using frequentist and Bayesian statistics. Patients showed a deficit compared to controls in detecting intentionality cues, but showed preserved metacognitive performance into this task. Our study reveals a selective deficit in the perception of intentionality cues, but preserved metacognitive insight into the validity of this perception. It thus appears that impairment of metacognition in schizophrenia varies across cognitive domains - metacognition should not be considered a monolithic stone that is either impaired or unimpaired.
Collapse
Affiliation(s)
- Ana Muthesius
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Farina Grothey
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Carter Cunningham
- Masters in Neuroscience Program, Medical Faculty, University of Bonn, Bonn, Germany
| | - Susanne Hölzer
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Kai Vogeley
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine, Cognitive Neuroscience (INM-3), Research Centre Jülich, Jülich, Germany
| | - Johannes Schultz
- Center for Economics and Neuroscience, University of Bonn, Bonn, Germany.,Institute of Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
115
|
Understanding complex functional wiring patterns in major depressive disorder through brain functional connectome. Transl Psychiatry 2021; 11:526. [PMID: 34645783 PMCID: PMC8513388 DOI: 10.1038/s41398-021-01646-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/20/2021] [Accepted: 09/29/2021] [Indexed: 02/06/2023] Open
Abstract
Brain function relies on efficient communications between distinct brain systems. The pathology of major depressive disorder (MDD) damages functional brain networks, resulting in cognitive impairment. Here, we reviewed the associations between brain functional connectome changes and MDD pathogenesis. We also highlighted the utility of brain functional connectome for differentiating MDD from other similar psychiatric disorders, predicting recurrence and suicide attempts in MDD, and evaluating treatment responses. Converging evidence has now linked aberrant brain functional network organization in MDD to the dysregulation of neurotransmitter signaling and neuroplasticity, providing insights into the neurobiological mechanisms of the disease and antidepressant efficacy. Widespread connectome dysfunctions in MDD patients include multiple, large-scale brain networks as well as local disturbances in brain circuits associated with negative and positive valence systems and cognitive functions. Although the clinical utility of the brain functional connectome remains to be realized, recent findings provide further promise that research in this area may lead to improved diagnosis, treatments, and clinical outcomes of MDD.
Collapse
|
116
|
N-methyl-d-aspartate receptor antagonism modulates P300 event-related potentials and associated activity in salience and central executive networks. Pharmacol Biochem Behav 2021; 211:173287. [PMID: 34653398 DOI: 10.1016/j.pbb.2021.173287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 11/21/2022]
Abstract
Impairments in auditory information processing in schizophrenia as indexed electrophysiologically by P300 deficits during novelty (P3a) and target (P3b) processing are linked to N -methyl- D -aspartate receptor (NMDAR) dysfunction. This study in 14 healthy volunteers examined the effects of a subanesthetic dose of the NMDAR antagonist ketamine on P300 and their relationship to psychomimetic symptoms and cortical source activity (with eLORETA). Ketamine reduced early (e- P3a) and late (l-P3a) novelty P300 at sensor (scalp)-level and at source-level in the salience network. Increases in dissociation symptoms were negatively correlated with ketamine-induced P3b changes, at sensor-level and source-level, in both salience and central executive networks. These P3a alterations during novelty processing, and the symptom-related P3b changes during target processing support a model of NMDAR hypofunction underlying disrupted auditory attention in schizophrenia.
Collapse
|
117
|
Yang M, Gao S, Xiong W, Zhang XY. Sex-differential associations between cognitive impairments and white matter abnormalities in first episode and drug-naïve schizophrenia. Early Interv Psychiatry 2021; 15:1179-1187. [PMID: 33058544 DOI: 10.1111/eip.13059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/24/2020] [Accepted: 09/26/2020] [Indexed: 11/30/2022]
Abstract
AIM Previous evidence has suggested that schizophrenia patients may display sex differences in cognitive impairments and cognitive impairments are related to disrupted white matter (WM) microstructure. The current research aims to address the intriguing possibility for the sex-specific association between cognitive deficits and WM abnormalities in first-episode and drug-naïve schizophrenia. METHODS Cognitive performance on the Measurement and Treatment Research to Improve Cognition in Schizophrenia (MATRICS) Consensus Cognitive Battery (MCCB) was measured in 39 FEND patients (females:males = 23:16) and 30 healthy controls (females:males = 17:13), together with whole-brain WM fractional anisotropy (FA) values determined using voxel-based diffusion tensor imaging. Correlations between cognitive performance and FA values were assessed. RESULTS Patients performed significantly worse than healthy controls in the total score and most of the subscores of MCCB. Female patients displayed better cognitive performance than male patients on the Trail Making A Test, the Hopkins Verbal Learning Test and the Spatial Span Test in the Wechsler Memory Scale. More importantly, sex-differential association between cognitive performance and FA values was found in patients, but not in healthy controls. In particular, FA values in the cerebellum were negatively correlated with the continuous performance and digital sequence scores in male patients but positively correlated with the performance on the Spatial Span Test in the Wechsler Memory Scale in female patients. CONCLUSIONS These findings suggest sex-specific neurobiological substrates involved in cognitive deficits in early-onset schizophrenia and have important implications for differentially targeted interventions between males and females.
Collapse
Affiliation(s)
- Mi Yang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for NeuroInformation, University of Electronic Science and Technology of China, Chengdu, China.,School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.,The Fourth People's Hospital of Chengdu, Chengdu, China
| | - Shan Gao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for NeuroInformation, University of Electronic Science and Technology of China, Chengdu, China
| | - Weisen Xiong
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for NeuroInformation, University of Electronic Science and Technology of China, Chengdu, China.,School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.,The Fourth People's Hospital of Chengdu, Chengdu, China
| | - Xiang Yang Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
118
|
Arnatkeviciute A, Fulcher BD, Bellgrove MA, Fornito A. Imaging Transcriptomics of Brain Disorders. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2021; 2:319-331. [PMID: 36324650 PMCID: PMC9616271 DOI: 10.1016/j.bpsgos.2021.10.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/06/2021] [Accepted: 10/11/2021] [Indexed: 01/05/2023] Open
Abstract
Noninvasive neuroimaging is a powerful tool for quantifying diverse aspects of brain structure and function in vivo, and it has been used extensively to map the neural changes associated with various brain disorders. However, most neuroimaging techniques offer only indirect measures of underlying pathological mechanisms. The recent development of anatomically comprehensive gene expression atlases has opened new opportunities for studying the transcriptional correlates of noninvasively measured neural phenotypes, offering a rich framework for evaluating pathophysiological hypotheses and putative mechanisms. Here, we provide an overview of some fundamental methods in imaging transcriptomics and outline their application to understanding brain disorders of neurodevelopment, adulthood, and neurodegeneration. Converging evidence indicates that spatial variations in gene expression are linked to normative changes in brain structure during age-related maturation and neurodegeneration that are in part associated with cell-specific gene expression markers of gene expression. Transcriptional correlates of disorder-related neuroimaging phenotypes are also linked to transcriptionally dysregulated genes identified in ex vivo analyses of patient brains. Modeling studies demonstrate that spatial patterns of gene expression are involved in regional vulnerability to neurodegeneration and the spread of disease across the brain. This growing body of work supports the utility of transcriptional atlases in testing hypotheses about the molecular mechanism driving disease-related changes in macroscopic neuroimaging phenotypes.
Collapse
Affiliation(s)
- Aurina Arnatkeviciute
- Turner Institute for Brain and Mental Health, School of Psychological Science, Monash University, Melbourne, Victoria, Australia
- Address correspondence to Aurina Arnatkeviciute, Ph.D
| | - Ben D. Fulcher
- School of Physics, The University of Sydney, Camperdown, New South Wales, Australia
| | - Mark A. Bellgrove
- Turner Institute for Brain and Mental Health, School of Psychological Science, Monash University, Melbourne, Victoria, Australia
| | - Alex Fornito
- Turner Institute for Brain and Mental Health, School of Psychological Science, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
119
|
Wang Z, Wang H, Mwansisya TE, Sheng Y, Shan B, Liu Z, Xue Z, Chen X. The integrity of the white matter in first-episode schizophrenia patients with auditory verbal hallucinations: An atlas-based DTI analysis. Psychiatry Res Neuroimaging 2021; 315:111328. [PMID: 34260985 DOI: 10.1016/j.pscychresns.2021.111328] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 05/31/2021] [Accepted: 07/02/2021] [Indexed: 11/30/2022]
Abstract
Auditory verbal hallucination (AVH) is one of the most remarkable symptoms of schizophrenia, with great impact on patients' lives and unclear pathogenesis. Neuroimaging studies have indicated that the development of AVHs is associated with white matter alteration, however, there are still inconsistencies in specific findings across previous investigations. The present study aimed to investigate the characteristics of the microstructural integrity of white matter (WM) in first-episode schizophrenia patients who experience auditory hallucinations. Atlas-based Diffusion Tensor Imaging (DTI) analysis was performed to evaluate the white matter integrity in 37 first-episode schizophrenia patients with AVH, 60 schizophrenia patients without AVH, and 50 healthy controls. Compared with the healthy controls group, AVH showed decreased mean fractional anisotropy (FA) in the genu and body of corpus callosum, right posterior corona radiata, left superior corona radiata, left external capsule, right superior fronto-occipital fasciculus, and higher mean diffusivity (MD) in genu of corpus callosum and left fornix and stria terminalis; whereas the nAVH group showed a much more significant reduction of FA and increased MD in broader brain regions. In addition, a significant positive correlation between FA and the severity of AVHs was observed in right posterior corona radiate. These observations collectively demonstrated that a certain degree of preserved fronto-temporal and interhemispheric connectivity in the early stage of schizophrenia might be associated with the brain capability to generate AVHs.
Collapse
Affiliation(s)
- Zhiyu Wang
- School of Public Health, Central South University, Changsha, China; Department of Communicable Disease Prevention and Management, Centers for Disease Control and Prevention(CDC) of Changsha City, Changsha, China
| | - Hui Wang
- Department of Geriatrics, The First Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, China
| | | | - Yaoyao Sheng
- National Clinical Research Center for Mental Disorders, and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Baoci Shan
- Key Laboratory of Nuclear Analysis, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Zhening Liu
- National Clinical Research Center for Mental Disorders, and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Psychiatry and Mental Health, National Technology Institute of Psychiatry, Changsha, China; Institute of Mental Health, Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhimin Xue
- National Clinical Research Center for Mental Disorders, and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Psychiatry and Mental Health, National Technology Institute of Psychiatry, Changsha, China; Institute of Mental Health, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xudong Chen
- National Clinical Research Center for Mental Disorders, and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Psychiatry and Mental Health, National Technology Institute of Psychiatry, Changsha, China; Institute of Mental Health, Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
120
|
Chamberland M, Genc S, Tax CMW, Shastin D, Koller K, Raven EP, Cunningham A, Doherty J, van den Bree MBM, Parker GD, Hamandi K, Gray WP, Jones DK. Detecting microstructural deviations in individuals with deep diffusion MRI tractometry. NATURE COMPUTATIONAL SCIENCE 2021; 1:598-606. [PMID: 35865756 PMCID: PMC7613101 DOI: 10.1038/s43588-021-00126-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 08/09/2021] [Indexed: 06/15/2023]
Abstract
Most diffusion magnetic resonance imaging studies of disease rely on statistical comparisons between large groups of patients and healthy participants to infer altered tissue states in the brain; however, clinical heterogeneity can greatly challenge their discriminative power. There is currently an unmet need to move away from the current approach of group-wise comparisons to methods with the sensitivity to detect altered tissue states at the individual level. This would ultimately enable the early detection and interpretation of microstructural abnormalities in individual patients, an important step towards personalized medicine in translational imaging. To this end, Detect was developed to advance diffusion magnetic resonance imaging tractometry towards single-patient analysis. By operating on the manifold of white-matter pathways and learning normative microstructural features, our framework captures idiosyncrasies in patterns along white-matter pathways. Our approach paves the way from traditional group-based comparisons to true personalized radiology, taking microstructural imaging from the bench to the bedside.
Collapse
Affiliation(s)
- Maxime Chamberland
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
- Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, the Netherlands
| | - Sila Genc
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | - Chantal M. W. Tax
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Dmitri Shastin
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
- Department of Neuroscience, University Hospital of Wales (UHW), Cardiff, UK
| | - Kristin Koller
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | - Erika P. Raven
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York, NY, USA
| | - Adam Cunningham
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Joanne Doherty
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Marianne B. M. van den Bree
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Greg D. Parker
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | - Khalid Hamandi
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
- Department of Neuroscience, University Hospital of Wales (UHW), Cardiff, UK
- Brain Repair and Intracranial Neurotherapeutics (BRAIN) Unit, School of Medicine, Cardiff University, Cardiff, UK
| | - William P. Gray
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
- Department of Neuroscience, University Hospital of Wales (UHW), Cardiff, UK
- Brain Repair and Intracranial Neurotherapeutics (BRAIN) Unit, School of Medicine, Cardiff University, Cardiff, UK
| | - Derek K. Jones
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| |
Collapse
|
121
|
Chopra S, Francey SM, O’Donoghue B, Sabaroedin K, Arnatkeviciute A, Cropley V, Nelson B, Graham J, Baldwin L, Tahtalian S, Yuen HP, Allott K, Alvarez-Jimenez M, Harrigan S, Pantelis C, Wood SJ, McGorry P, Fornito A. Functional Connectivity in Antipsychotic-Treated and Antipsychotic-Naive Patients With First-Episode Psychosis and Low Risk of Self-harm or Aggression: A Secondary Analysis of a Randomized Clinical Trial. JAMA Psychiatry 2021; 78:994-1004. [PMID: 34160595 PMCID: PMC8223142 DOI: 10.1001/jamapsychiatry.2021.1422] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
IMPORTANCE Altered functional connectivity (FC) is a common finding in resting-state functional magnetic resonance imaging (rs-fMRI) studies of people with psychosis, yet how FC disturbances evolve in the early stages of illness, and how antipsychotic treatment influences these disturbances, remains unknown. OBJECTIVE To investigate longitudinal FC changes in antipsychotic-naive and antipsychotic-treated patients with first-episode psychosis (FEP). DESIGN, SETTING, AND PARTICIPANTS This secondary analysis of a triple-blind, randomized clinical trial was conducted over a 5-year recruitment period between April 2008 and December 2016 with 59 antipsychotic-naive patients with FEP receiving either a second-generation antipsychotic or a placebo pill over a treatment period of 6 months. Participants were required to have low suicidality and aggression, to have a duration of untreated psychosis of less than 6 months, and to be living in stable accommodations with social support. Both FEP groups received intensive psychosocial therapy. A healthy control group was also recruited. Participants completed rs-fMRI scans at baseline, 3 months, and 12 months. Data were analyzed from May 2019 to August 2020. INTERVENTIONS Resting-state functional MRI was used to probe brain FC. Patients received either a second-generation antipsychotic or a matched placebo tablet. Both patient groups received a manualized psychosocial intervention. MAIN OUTCOMES AND MEASURES The primary outcomes of this analysis were to investigate (1) FC differences between patients and controls at baseline; (2) FC changes in medicated and unmedicated patients between baseline and 3 months; and (3) associations between longitudinal FC changes and clinical outcomes. An additional aim was to investigate long-term FC changes at 12 months after baseline. These outcomes were not preregistered. RESULTS Data were analyzed for 59 patients (antipsychotic medication plus psychosocial treatment: 28 [47.5%]; mean [SD] age, 19.5 [3.0] years; 15 men [53.6%]; placebo plus psychosocial treatment: 31 [52.5%]; mean [SD] age, 18.8 [2.7]; 16 men [51.6%]) and 27 control individuals (mean [SD] age, 21.9 [1.9] years). At baseline, patients showed widespread functional dysconnectivity compared with controls, with reductions predominantly affecting interactions between the default mode network, limbic systems, and the rest of the brain. From baseline to 3 months, patients receiving placebo showed increased FC principally within the same systems; some of these changes correlated with improved clinical outcomes (canonical correlation analysis R = 0.901; familywise error-corrected P = .005). Antipsychotic exposure was associated with increased FC primarily between the thalamus and the rest of the brain. CONCLUSIONS AND RELEVANCE In this secondary analysis of a clinical trial, antipsychotic-naive patients with FEP showed widespread functional dysconnectivity at baseline, followed by an early normalization of default mode network and cortical limbic dysfunction in patients receiving placebo and psychosocial intervention. Antipsychotic exposure was associated with FC changes concentrated on thalamocortical networks. TRIAL REGISTRATION ACTRN12607000608460.
Collapse
Affiliation(s)
- Sidhant Chopra
- Turner Institute for Brain and Mental Health, Monash University School of Psychological Sciences, Clayton, Victoria, Australia,Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia
| | - Shona M. Francey
- Orygen, Parkville, Victoria, Australia,Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Brian O’Donoghue
- Orygen, Parkville, Victoria, Australia,Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kristina Sabaroedin
- Turner Institute for Brain and Mental Health, Monash University School of Psychological Sciences, Clayton, Victoria, Australia,Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia
| | - Aurina Arnatkeviciute
- Turner Institute for Brain and Mental Health, Monash University School of Psychological Sciences, Clayton, Victoria, Australia
| | - Vanessa Cropley
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne & Melbourne Health, Melbourne, Victoria, Australia
| | - Barnaby Nelson
- Orygen, Parkville, Victoria, Australia,Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jessica Graham
- Orygen, Parkville, Victoria, Australia,Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Lara Baldwin
- Orygen, Parkville, Victoria, Australia,Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Steven Tahtalian
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne & Melbourne Health, Melbourne, Victoria, Australia
| | - Hok Pan Yuen
- Orygen, Parkville, Victoria, Australia,Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kelly Allott
- Orygen, Parkville, Victoria, Australia,Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Mario Alvarez-Jimenez
- Orygen, Parkville, Victoria, Australia,Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Susy Harrigan
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia,Department of Social Work, Monash University, Caulfield, Victoria, Australia
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne & Melbourne Health, Melbourne, Victoria, Australia
| | - Stephen J. Wood
- Orygen, Parkville, Victoria, Australia,Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia,University of Birmingham School of Psychology, Edgbaston, United Kingdom
| | - Patrick McGorry
- Orygen, Parkville, Victoria, Australia,Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Alex Fornito
- Turner Institute for Brain and Mental Health, Monash University School of Psychological Sciences, Clayton, Victoria, Australia,Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
122
|
Korhonen O, Zanin M, Papo D. Principles and open questions in functional brain network reconstruction. Hum Brain Mapp 2021; 42:3680-3711. [PMID: 34013636 PMCID: PMC8249902 DOI: 10.1002/hbm.25462] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/11/2021] [Accepted: 04/10/2021] [Indexed: 12/12/2022] Open
Abstract
Graph theory is now becoming a standard tool in system-level neuroscience. However, endowing observed brain anatomy and dynamics with a complex network representation involves often covert theoretical assumptions and methodological choices which affect the way networks are reconstructed from experimental data, and ultimately the resulting network properties and their interpretation. Here, we review some fundamental conceptual underpinnings and technical issues associated with brain network reconstruction, and discuss how their mutual influence concurs in clarifying the organization of brain function.
Collapse
Affiliation(s)
- Onerva Korhonen
- Department of Computer ScienceAalto University, School of ScienceHelsinki
- Centre for Biomedical TechnologyUniversidad Politécnica de MadridPozuelo de Alarcón
| | - Massimiliano Zanin
- Instituto de Física Interdisciplinar y Sistemas Complejos IFISC (CSIC‐UIB), Campus UIBPalma de MallorcaSpain
| | - David Papo
- Fondazione Istituto Italiano di TecnologiaFerrara
- Department of Neuroscience and Rehabilitation, Section of PhysiologyUniversity of FerraraFerrara
| |
Collapse
|
123
|
Abstract
Niemann-Pick disease type C (NP-C) is a severe neurovisceral lipid storage disease that results in the accumulation of unesterified cholesterol in lysosomes or endosomes. The clinical presentations of NP-C are variable which include visceral symptoms, neurologic symptoms and psychiatric symptoms. Psychosis is the most common psychiatric manifestation of NP-C and is indistinguishable from a typical psychosis presentation of schizophrenia. The common psychotic presentations in NP-C include visual hallucinations, delusions, auditory hallucinations and thought disorders. Psychosis symptoms are more common in adult or adolescent-onset forms compared with pediatric-onset forms. The underlying pathophysiology of psychosis in NP-C is most probably due to dysconnectivity particularly between frontotemporal connectivity and subcortical structures. NP-C sometimes is mistaken for schizophrenia which causes delay in treatment due to lack of awareness and literature review. This review aims to summarize the relevant case reports on psychosis symptoms in NP-C and discuss the genetics and pathophysiology underlying the condition.
Collapse
Affiliation(s)
- Leong Tung Ong
- Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
124
|
The Imbalanced Plasticity Hypothesis of Schizophrenia-Related Psychosis: A Predictive Perspective. COGNITIVE, AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2021; 21:679-697. [PMID: 34050524 DOI: 10.3758/s13415-021-00911-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/28/2021] [Indexed: 02/06/2023]
Abstract
A considerable number of studies have attempted to account for the psychotic aspects of schizophrenia in terms of the influential predictive coding (PC) hypothesis. We argue that the prediction-oriented perspective on schizophrenia-related psychosis may benefit from a mechanistic model that: 1) gives due weight to the extent to which alterations in short- and long-term synaptic plasticity determine the degree and the direction of the functional disruption that occurs in psychosis; and 2) addresses the distinction between the two central syndromes of psychosis in schizophrenia: disorganization and reality-distortion. To accomplish these goals, we propose the Imbalanced Plasticity Hypothesis - IPH, and demonstrate that it: 1) accounts for commonalities and differences between disorganization and reality distortion in terms of excessive (hyper) or insufficient (hypo) neuroplasticity, respectively; 2) provides distinct predictions in the cognitive and electrophysiological domains; and 3) is able to reconcile conflicting PC-oriented accounts of psychosis.
Collapse
|
125
|
Berdenis van Berlekom A, Notman N, Sneeboer MAM, Snijders GJLJ, Houtepen LC, Nispeling DM, He Y, Dracheva S, Hol EM, Kahn RS, de Witte LD, Boks MP. DNA methylation differences in cortical grey and white matter in schizophrenia. Epigenomics 2021; 13:1157-1169. [PMID: 34323598 PMCID: PMC8386513 DOI: 10.2217/epi-2021-0077] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/09/2021] [Indexed: 01/27/2023] Open
Abstract
Aim: Identify grey- and white-matter-specific DNA-methylation differences between schizophrenia (SCZ) patients and controls in postmortem brain cortical tissue. Materials & methods: Grey and white matter were separated from postmortem brain tissue of the superior temporal and medial frontal gyrus from SCZ (n = 10) and control (n = 11) cases. Genome-wide DNA-methylation analysis was performed using the Infinium EPIC Methylation Array (Illumina, CA, USA). Results: Four differentially methylated regions associated with SCZ status and tissue type (grey vs white matter) were identified within or near KLF9, SFXN1, SPRED2 and ALS2CL genes. Gene-expression analysis showed differential expression of KLF9 and SFXN1 in SCZ. Conclusion: Our data show distinct differences in DNA methylation between grey and white matter that are unique to SCZ, providing new leads to unravel the pathogenesis of SCZ.
Collapse
Affiliation(s)
- Amber Berdenis van Berlekom
- Department of Psychiatry, Brain Center University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Translational Neuroscience, Brain Center University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Nina Notman
- Department of Psychiatry, Brain Center University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marjolein AM Sneeboer
- Department of Psychiatry, Brain Center University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Translational Neuroscience, Brain Center University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Gijsje JLJ Snijders
- Department of Psychiatry, Brain Center University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lotte C Houtepen
- Department of Psychiatry, Brain Center University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Danny M Nispeling
- Department of Psychiatry, Brain Center University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Yujie He
- Department of Psychiatry, Brain Center University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Translational Neuroscience, Brain Center University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Stella Dracheva
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mental Illness Research, Education, & Clinical Center (VISN 2 South), James J Peters VA Medical Center, Bronx, NY, 10468, USA
| | - Elly M Hol
- Department of Translational Neuroscience, Brain Center University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - René S Kahn
- Department of Psychiatry, Brain Center University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mental Illness Research, Education, & Clinical Center (VISN 2 South), James J Peters VA Medical Center, Bronx, NY, 10468, USA
| | - Lot D de Witte
- Department of Psychiatry, Brain Center University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Marco P Boks
- Department of Psychiatry, Brain Center University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
126
|
Mastrandrea R, Piras F, Gabrielli A, Banaj N, Caldarelli G, Spalletta G, Gili T. The unbalanced reorganization of weaker functional connections induces the altered brain network topology in schizophrenia. Sci Rep 2021; 11:15400. [PMID: 34321538 PMCID: PMC8319172 DOI: 10.1038/s41598-021-94825-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/08/2021] [Indexed: 01/10/2023] Open
Abstract
Network neuroscience shed some light on the functional and structural modifications occurring to the brain associated with the phenomenology of schizophrenia. In particular, resting-state functional networks have helped our understanding of the illness by highlighting the global and local alterations within the cerebral organization. We investigated the robustness of the brain functional architecture in 44 medicated schizophrenic patients and 40 healthy comparators through an advanced network analysis of resting-state functional magnetic resonance imaging data. The networks in patients showed more resistance to disconnection than in healthy controls, with an evident discrepancy between the two groups in the node degree distribution computed along a percolation process. Despite a substantial similarity of the basal functional organization between the two groups, the expected hierarchy of healthy brains' modular organization is crumbled in schizophrenia, showing a peculiar arrangement of the functional connections, characterized by several topologically equivalent backbones. Thus, the manifold nature of the functional organization's basal scheme, together with its altered hierarchical modularity, may be crucial in the pathogenesis of schizophrenia. This result fits the disconnection hypothesis that describes schizophrenia as a brain disorder characterized by an abnormal functional integration among brain regions.
Collapse
Affiliation(s)
| | - Fabrizio Piras
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, 00179, Rome, Italy
| | - Andrea Gabrielli
- Dipartimento di Ingegneria, Università Roma Tre, 00146, Rome, Italy.,Istituto dei Sistemi Complessi (ISC)-CNR, UoS Sapienza, Dipartimento di Fisica, Università "Sapienza", 00185, Rome, Italy
| | - Nerisa Banaj
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, 00179, Rome, Italy
| | - Guido Caldarelli
- Networks Unit, IMT School for Advanced Studies, 55100, Lucca, Italy.,Istituto dei Sistemi Complessi (ISC)-CNR, UoS Sapienza, Dipartimento di Fisica, Università "Sapienza", 00185, Rome, Italy
| | - Gianfranco Spalletta
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, 00179, Rome, Italy. .,Division of Neuropsychiatry, Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Tommaso Gili
- Networks Unit, IMT School for Advanced Studies, 55100, Lucca, Italy
| |
Collapse
|
127
|
Storch S, Samantzis M, Balbi M. Driving Oscillatory Dynamics: Neuromodulation for Recovery After Stroke. Front Syst Neurosci 2021; 15:712664. [PMID: 34366801 PMCID: PMC8339272 DOI: 10.3389/fnsys.2021.712664] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/21/2021] [Indexed: 12/18/2022] Open
Abstract
Stroke is a leading cause of death and disability worldwide, with limited treatments being available. However, advances in optic methods in neuroscience are providing new insights into the damaged brain and potential avenues for recovery. Direct brain stimulation has revealed close associations between mental states and neuroprotective processes in health and disease, and activity-dependent calcium indicators are being used to decode brain dynamics to understand the mechanisms underlying these associations. Evoked neural oscillations have recently shown the ability to restore and maintain intrinsic homeostatic processes in the brain and could be rapidly deployed during emergency care or shortly after admission into the clinic, making them a promising, non-invasive therapeutic option. We present an overview of the most relevant descriptions of brain injury after stroke, with a focus on disruptions to neural oscillations. We discuss the optical technologies that are currently used and lay out a roadmap for future studies needed to inform the next generation of strategies to promote functional recovery after stroke.
Collapse
Affiliation(s)
- Sven Storch
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Montana Samantzis
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Matilde Balbi
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
128
|
Chang X, Mandl RCW, Pasternak O, Brouwer RM, Cahn W, Collin G. Diffusion MRI derived free-water imaging measures in patients with schizophrenia and their non-psychotic siblings. Prog Neuropsychopharmacol Biol Psychiatry 2021; 109:110238. [PMID: 33400942 DOI: 10.1016/j.pnpbp.2020.110238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/16/2020] [Accepted: 12/30/2020] [Indexed: 10/22/2022]
Abstract
Free-water imaging is a diffusion MRI technique that separately models water diffusion hindered by fiber tissue and water that disperses freely in the extracellular space. Studies using this technique have shown that schizophrenia is characterized by a lower level of fractional anisotropy of the tissue compartment (FAt) and higher free-water fractional volume (FW). It is unknown, however, whether such abnormalities are an expression of pre-existing (genetic) risk for schizophrenia or a manifestation of the illness. To investigate the contribution of familial risk factors to white matter abnormalities, we used the free-water imaging technique to assess FAt and FW in a large cohort of 471 participants including 161 patients with schizophrenia, 182 non-psychotic siblings, and 128 healthy controls. In this sample, patients did not show significant differences in FAt as compared to controls, but did exhibit a higher level of FW relative to both controls and siblings in the left uncinate fasciculus, superior corona radiata and fornix / stria terminalis. This increase in FW was found to be related to, though not solely explained by, ventricular enlargement. Siblings did not show significant FW abnormalities. However, siblings did show a higher level of FAt as compared to controls and patients, in line with results of a previous study on the same data using conventional DTI. Taken together, our findings suggest that extracellular free-water accumulation in patients is likely a manifestation of established disease rather than an expression of familial risk for schizophrenia and that super-normal levels of FAt in unaffected siblings may reflect a compensatory process.
Collapse
Affiliation(s)
- Xiao Chang
- Department of Psychiatry, University Medical Center Utrecht (UMCU), UMCU Brain Center, Utrecht, the Netherlands; Social, Genetic and Developmental Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
| | - René C W Mandl
- Department of Psychiatry, University Medical Center Utrecht (UMCU), UMCU Brain Center, Utrecht, the Netherlands
| | - Ofer Pasternak
- Department of Psychiatry, Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Rachel M Brouwer
- Department of Psychiatry, University Medical Center Utrecht (UMCU), UMCU Brain Center, Utrecht, the Netherlands
| | - Wiepke Cahn
- Department of Psychiatry, University Medical Center Utrecht (UMCU), UMCU Brain Center, Utrecht, the Netherlands; Altrecht Institute of Mental Health Care, Utrecht, the Netherlands
| | - Guusje Collin
- Department of Psychiatry, University Medical Center Utrecht (UMCU), UMCU Brain Center, Utrecht, the Netherlands; Department of Psychiatry, Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, USA; McGovern Institute for Brain Research, Massachusetts Institute of Technology, Boston, USA
| |
Collapse
|
129
|
Lizano P, Lutz O, Xu Y, Rubin LH, Paskowitz L, Lee AM, Eum S, Keedy SK, Hill SK, Reilly JL, Wu B, Tamminga CA, Clementz BA, Pearlson GD, Gershon ES, Keshavan MS, Sweeney JA, Bishop JR. Multivariate relationships between peripheral inflammatory marker subtypes and cognitive and brain structural measures in psychosis. Mol Psychiatry 2021; 26:3430-3443. [PMID: 33060818 PMCID: PMC8046847 DOI: 10.1038/s41380-020-00914-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022]
Abstract
Elevations in peripheral inflammatory markers have been reported in patients with psychosis. Whether this represents an inflammatory process defined by individual or subgroups of markers is unclear. Further, relationships between peripheral inflammatory marker elevations and brain structure, cognition, and clinical features of psychosis remain unclear. We hypothesized that a pattern of plasma inflammatory markers, and an inflammatory subtype established from this pattern, would be elevated across the psychosis spectrum and associated with cognition and brain structural alterations. Clinically stable psychosis probands (Schizophrenia spectrum, n = 79; Psychotic Bipolar disorder, n = 61) and matched healthy controls (HC, n = 60) were assessed for 15 peripheral inflammatory markers, cortical thickness, subcortical volume, cognition, and symptoms. A combination of unsupervised exploratory factor analysis and hierarchical clustering was used to identify inflammation subtypes. Levels of IL6, TNFα, VEGF, and CRP were significantly higher in psychosis probands compared to HCs, and there were marker-specific differences when comparing diagnostic groups. Individual and/or inflammatory marker patterns were associated with neuroimaging, cognition, and symptom measures. A higher inflammation subgroup was defined by elevations in a group of 7 markers in 36% of Probands and 20% of HCs. Probands in the elevated inflammatory marker group performed significantly worse on cognitive measures of visuo-spatial working memory and response inhibition, displayed elevated hippocampal, amygdala, putamen and thalamus volumes, and evidence of gray matter thickening compared to the proband group with low inflammatory marker levels. These findings specify the nature of peripheral inflammatory marker alterations in psychotic disorders and establish clinical, neurocognitive and neuroanatomic associations with increased inflammatory activation in psychosis. The identification of a specific subgroup of patients with inflammatory alteration provides a potential means for targeting treatment with anti-inflammatory medications.
Collapse
Affiliation(s)
- Paulo Lizano
- Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| | - Olivia Lutz
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Yanxun Xu
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
| | - Leah H Rubin
- Department of Neurology, Psychiatry, and Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Lyle Paskowitz
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
| | - Adam M Lee
- Department of Experimental and Clinical Pharmacology and Department of Psychiatry, University of Minnesota, Minneapolis, MN, USA
| | - Seenae Eum
- School of Pharmacy, Shenandoah University, Winchester, Virginia, USA
| | - Sarah K Keedy
- Department of Psychiatry and Behavioral Neurosciences, University of Chicago, Chicago, IL, USA
| | - S Kristian Hill
- Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - James L Reilly
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Chicago, IL, USA
| | - Baolin Wu
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Carol A Tamminga
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Brett A Clementz
- Department of Psychology, University of Georgia, Athens, GA, USA
| | | | - Elliot S Gershon
- Department of Psychiatry and Behavioral Neurosciences, University of Chicago, Chicago, IL, USA
| | - Matcheri S Keshavan
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - John A Sweeney
- Deptartment of Psychiatry, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Jeffrey R Bishop
- Department of Experimental and Clinical Pharmacology and Department of Psychiatry, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
130
|
Ding C, Zhang C, Kopp R, Kuney L, Meng Q, Wang L, Xia Y, Jiang Y, Dai R, Min S, Yao WD, Wong ML, Ruan H, Liu C, Chen C. Transcription factor POU3F2 regulates TRIM8 expression contributing to cellular functions implicated in schizophrenia. Mol Psychiatry 2021; 26:3444-3460. [PMID: 32929213 PMCID: PMC7956165 DOI: 10.1038/s41380-020-00877-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 01/17/2023]
Abstract
Schizophrenia (SCZ) is a neuropsychiatric disorder with aberrant expression of multiple genes. However, identifying its exact causal genes remains a considerable challenge. The brain-specific transcription factor POU3F2 (POU domain, class 3, transcription factor 2) has been recognized as a risk factor for SCZ, but our understanding of its target genes and pathogenic mechanisms are still limited. Here we report that POU3F2 regulates 42 SCZ-related genes in knockdown and RNA-sequencing experiments of human neural progenitor cells (NPCs). Among those SCZ-related genes, TRIM8 (Tripartite motif containing 8) is located in SCZ-associated genetic locus and is aberrantly expressed in patients with SCZ. Luciferase reporter and electrophoretic mobility shift assays (EMSA) showed that POU3F2 induces TRIM8 expression by binding to the SCZ-associated SNP (single nucleotide polymorphism) rs5011218, which affects POU3F2-binding efficiency at the promoter region of TRIM8. We investigated the cellular functions of POU3F2 and TRIM8 as they co-regulate several pathways related to neural development and synaptic function. Knocking down either POU3F2 or TRIM8 promoted the proliferation of NPCs, inhibited their neuronal differentiation, and impaired the excitatory synaptic transmission of NPC-derived neurons. These results indicate that POU3F2 regulates TRIM8 expression through the SCZ-associated SNP rs5011218, and both genes may be involved in the etiology of SCZ by regulating neural development and synaptic function.
Collapse
Affiliation(s)
- Chaodong Ding
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China,Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Chunling Zhang
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Richard Kopp
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Liz Kuney
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Qingtuan Meng
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China,Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China,Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, Guangxi, China
| | - Le Wang
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China,Child Health Institute of New Jersey, Department of Neuroscience and Cell Biology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Yan Xia
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China,Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Yi Jiang
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Rujia Dai
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Shishi Min
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China,Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Wei-Dong Yao
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Ma-Li Wong
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Hongyu Ruan
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA.
| | - Chunyu Liu
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China. .,Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA. .,School of Psychology, Shaanxi Normal University, Xi'an, Shaanxi, China.
| | - Chao Chen
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, the Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Molecular Precision Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
131
|
Xi YB, Guo F, Liu WM, Fu YF, Li JM, Wang HN, Chen FL, Cui LB, Zhu YQ, Li C, Kang XW, Li BJ, Yin H. Triple network hypothesis-related disrupted connections in schizophrenia: A spectral dynamic causal modeling analysis with functional magnetic resonance imaging. Schizophr Res 2021; 233:89-96. [PMID: 34246865 DOI: 10.1016/j.schres.2021.06.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 04/15/2021] [Accepted: 06/21/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE The symptom-related neurobiology characteristic of schizophrenia in the brain from a network perspective is still poorly understood, leading to a lack of potential biologically-based markers and difficulty identifying therapeutic targets. We aim to test the dysregulated cross-network interactions among the Salience Network (SN), Central Executive Network (CEN) and Default Mode Network (DMN) and how they contributed to different symptoms in schizophrenia patients. METHODS We examined network interactions among the SN, CEN and DMN in 76 patients with schizophrenia vs. 80 well-matched controls using dynamic causal modeling (DCM). We further analyzed the relation between network dynamics and Positive and Negative Syndrome Scale (PANSS). RESULTS We observed that the DMN, CEN and SN across healthy controls and schizophrenia patients showed several similarities within or between-network pattern in the resting state. Comparing schizophrenia to controls, SN-centered cross-network interactions were most significantly reduced. Crucially, the strength of connections from CEN subnetwork 1 to DMN subnetwork 1 was positively correlated with the Positive Score of PANSS. The connection from the DMN subnetwork 2 to CEN subnetwork 2 was negatively correlated with the Negative Score of PANSS. CONCLUSIONS Our study provides strong evidence for the dysregulation among SN, CEN and DMN in a triple-network perspective in schizophrenia. The connection between DMN and CEN could be clinically-relevant neurobiological signature of schizophrenia symptoms. Our study indicated that the description of brain triple network hypothesis could be a novel and possible bio-marker for schizophrenia.
Collapse
Affiliation(s)
- Yi-Bin Xi
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Fan Guo
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wen-Ming Liu
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yu-Fei Fu
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jia-Ming Li
- School of Biomedical Engineering, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hua-Ning Wang
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Fu-Lin Chen
- College of Life Sciences, Northwest University, Taibai North Rd 229, Xi'an, Shaanxi, China
| | - Long-Biao Cui
- Department of Clinical Psychology, School of Medical Psychology, Fourth Military Medical University, Xi'an, Shaanxi, China; The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yuan-Qiang Zhu
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chen Li
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiao-Wei Kang
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Bao-Juan Li
- School of Biomedical Engineering, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Hong Yin
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
132
|
Frässle S, Aponte EA, Bollmann S, Brodersen KH, Do CT, Harrison OK, Harrison SJ, Heinzle J, Iglesias S, Kasper L, Lomakina EI, Mathys C, Müller-Schrader M, Pereira I, Petzschner FH, Raman S, Schöbi D, Toussaint B, Weber LA, Yao Y, Stephan KE. TAPAS: An Open-Source Software Package for Translational Neuromodeling and Computational Psychiatry. Front Psychiatry 2021; 12:680811. [PMID: 34149484 PMCID: PMC8206497 DOI: 10.3389/fpsyt.2021.680811] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/10/2021] [Indexed: 12/26/2022] Open
Abstract
Psychiatry faces fundamental challenges with regard to mechanistically guided differential diagnosis, as well as prediction of clinical trajectories and treatment response of individual patients. This has motivated the genesis of two closely intertwined fields: (i) Translational Neuromodeling (TN), which develops "computational assays" for inferring patient-specific disease processes from neuroimaging, electrophysiological, and behavioral data; and (ii) Computational Psychiatry (CP), with the goal of incorporating computational assays into clinical decision making in everyday practice. In order to serve as objective and reliable tools for clinical routine, computational assays require end-to-end pipelines from raw data (input) to clinically useful information (output). While these are yet to be established in clinical practice, individual components of this general end-to-end pipeline are being developed and made openly available for community use. In this paper, we present the Translational Algorithms for Psychiatry-Advancing Science (TAPAS) software package, an open-source collection of building blocks for computational assays in psychiatry. Collectively, the tools in TAPAS presently cover several important aspects of the desired end-to-end pipeline, including: (i) tailored experimental designs and optimization of measurement strategy prior to data acquisition, (ii) quality control during data acquisition, and (iii) artifact correction, statistical inference, and clinical application after data acquisition. Here, we review the different tools within TAPAS and illustrate how these may help provide a deeper understanding of neural and cognitive mechanisms of disease, with the ultimate goal of establishing automatized pipelines for predictions about individual patients. We hope that the openly available tools in TAPAS will contribute to the further development of TN/CP and facilitate the translation of advances in computational neuroscience into clinically relevant computational assays.
Collapse
Affiliation(s)
- Stefan Frässle
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Eduardo A. Aponte
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Saskia Bollmann
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, ETH Zurich and University of Zurich, Zurich, Switzerland
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States
- Department of Radiology, Harvard Medical School, Charlestown, MA, United States
| | - Kay H. Brodersen
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
- Department of Computer Science, ETH Zurich, Zurich, Switzerland
| | - Cao T. Do
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Olivia K. Harrison
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Samuel J. Harrison
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Jakob Heinzle
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Sandra Iglesias
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Lars Kasper
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
- Techna Institute, University Health Network, Toronto, ON, Canada
| | - Ekaterina I. Lomakina
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
- Department of Computer Science, ETH Zurich, Zurich, Switzerland
| | - Christoph Mathys
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
- Interacting Minds Center, Aarhus University, Aarhus, Denmark
| | - Matthias Müller-Schrader
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Inês Pereira
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Frederike H. Petzschner
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Sudhir Raman
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Dario Schöbi
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Birte Toussaint
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Lilian A. Weber
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Yu Yao
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Klaas E. Stephan
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
133
|
Gangadin SS, Cahn W, Scheewe TW, Hulshoff Pol HE, Bossong MG. Reduced resting state functional connectivity in the hippocampus-midbrain-striatum network of schizophrenia patients. J Psychiatr Res 2021; 138:83-88. [PMID: 33836433 DOI: 10.1016/j.jpsychires.2021.03.041] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/22/2021] [Accepted: 03/19/2021] [Indexed: 11/17/2022]
Abstract
Contemporary preclinical models suggest that abnormal functioning of a brain network consisting of the hippocampus, midbrain and striatum plays a critical role in the pathophysiology of schizophrenia. Previous neuroimaging studies examined individual aspects of this model in schizophrenia patients and individuals at clinical high risk for psychosis. However, this exact preclinical brain network has not been translated to human neuroimaging studies with schizophrenia patients and therefore it is currently unknown how functioning of this network is altered in patients. Here we investigated resting state functional connectivity in the hippocampus-midbrain-striatum network of schizophrenia patients, using functional Magnetic Resonance Imaging. Based on preclinical models, a network of functionally validated brain regions comprising the anterior subiculum (SUB), limbic striatum (LS), ventral tegmental area (VTA) and associative striatum (AS) was examined in 47 schizophrenia patients and 51 healthy controls. Schizophrenia patients demonstrated significantly lower functional connectivity in this hippocampus-midbrain-striatum network compared with healthy controls (p = 0.036). Particular reductions in connectivity were found between the SUB and LS (0.002 ± 0.315 and 0.116 ± 0.224, p = 0.040) and between the VTA and AS (0.230 ± 0.268 and 0.356 ± 0.285, p = 0.026). In patients, functional connectivity was not significantly associated with positive, negative or general symptom scores. Reduced connectivity is consistent with the concept of functional brain dysconnectivity as a key feature of the disorder. Our results support the notion that functioning of the hippocampus-midbrain-striatum network is significantly altered in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Shiral S Gangadin
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands; Section of Neuropsychiatry, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, Groningen, the Netherlands
| | - Wiepke Cahn
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Thomas W Scheewe
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hilleke E Hulshoff Pol
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Matthijs G Bossong
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
134
|
Michelini G, Palumbo IM, DeYoung CG, Latzman RD, Kotov R. Linking RDoC and HiTOP: A new interface for advancing psychiatric nosology and neuroscience. Clin Psychol Rev 2021; 86:102025. [PMID: 33798996 PMCID: PMC8165014 DOI: 10.1016/j.cpr.2021.102025] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 03/11/2021] [Accepted: 03/19/2021] [Indexed: 12/14/2022]
Abstract
The Research Domain Criteria (RDoC) and the Hierarchical Taxonomy of Psychopathology (HiTOP) represent major dimensional frameworks proposing two alternative approaches to accelerate progress in the way psychopathology is studied, classified, and treated. RDoC is a research framework rooted in neuroscience aiming to further the understanding of transdiagnostic biobehavioral systems underlying psychopathology and ultimately inform future classifications. HiTOP is a dimensional classification system, derived from the observed covariation among symptoms of psychopathology and maladaptive traits, which seeks to provide more informative research and treatment targets (i.e., dimensional constructs and clinical assessments) than traditional diagnostic categories. This article argues that the complementary strengths of RDoC and HiTOP can be leveraged in order to achieve their respective goals. RDoC's biobehavioral framework may help elucidate the underpinnings of the clinical dimensions included in HiTOP, whereas HiTOP may provide psychometrically robust clinical targets for RDoC-informed research. We present a comprehensive mapping between dimensions included in RDoC (constructs and subconstructs) and HiTOP (spectra and subfactors) based on narrative review of the empirical literature. The resulting RDoC-HiTOP interface sheds light on the biobehavioral correlates of clinical dimensions and provides a broad set of dimensional clinical targets for etiological and neuroscientific research. We conclude with future directions and practical recommendations for using this interface to advance clinical neuroscience and psychiatric nosology. Ultimately, we envision that this RDoC-HiTOP interface has the potential to inform the development of a unified, dimensional, and biobehaviorally-grounded psychiatric nosology.
Collapse
Affiliation(s)
- Giorgia Michelini
- Semel Institute for Neuroscience & Human Behavior, Department of Psychiatry & Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA 90024, United States of America.
| | - Isabella M Palumbo
- Department of Psychology, Georgia State University, Atlanta, GA 30303, United States of America
| | - Colin G DeYoung
- Department of Psychology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Robert D Latzman
- Department of Psychology, Georgia State University, Atlanta, GA 30303, United States of America
| | - Roman Kotov
- Department of Psychiatry & Behavioral Health, Stony Brook University, Stony Brook, NY 11790, United States of America
| |
Collapse
|
135
|
Subeh GK, Lajber M, Patel T, Mostafa JA. Anti-N-Methyl-D-Aspartate Receptor Encephalitis: A Detailed Review of the Different Psychiatric Presentations and Red Flags to Look for in Suspected Cases. Cureus 2021; 13:e15188. [PMID: 34178509 PMCID: PMC8225684 DOI: 10.7759/cureus.15188] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Anti-N-methyl-D-aspartate receptor encephalitis is a rare autoimmune disorder that involves N-methyl-D-aspartate (NMDA) receptors. It is the most common autoimmune encephalitis, and early detection and treatment are crucial for morbidity-free recovery. Distinguishing this disorder from a primary psychiatric illness is quite challenging as this disorder classically presents with psychiatric manifestations that often resemble schizophrenic psychosis. Therefore, this review intended to scope the psychiatric manifestations this disorder could present with and dissect how they differ from primary psychiatric disorders. A PubMed database search was done. The results yielded were analyzed; eventually, 50 papers were used to review the different signs and symptoms the disease can present with, including common and rare disease presentations. Diagnostic challenges and helpful clinical clues to recognize the disorder were reviewed as well.
Collapse
Affiliation(s)
- Ghasaq K Subeh
- College of Medicine, University of Baghdad, Baghdad, IRQ.,Internal Medicine, Al-Karama Teaching Hospital, Baghdad, IRQ.,Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Mehreen Lajber
- Medical Education, Bacha Khan Medical College, Mardan, PAK.,Medical Education, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Talha Patel
- Emergency Department, East Lancashire Hospitals NHS Trust, Blackburn, GBR.,Emergency, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Jihan A Mostafa
- Psychiatry, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
136
|
Orepic P, Rognini G, Kannape OA, Faivre N, Blanke O. Sensorimotor conflicts induce somatic passivity and louden quiet voices in healthy listeners. Schizophr Res 2021; 231:170-177. [PMID: 33866262 DOI: 10.1016/j.schres.2021.03.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 02/09/2021] [Accepted: 03/28/2021] [Indexed: 02/08/2023]
Abstract
Sensorimotor conflicts are known to alter the perception of accompanying sensory signals, and deficits in sensory attenuation have been observed in schizophrenia. In the auditory domain, self-generated tones or voices (compared to tones or voices presented passively or with temporal delays) have been associated with changes in loudness perception and attenuated neural responses. It has been argued that for sensory signals to be attenuated, predicted and sensory consequences must have a consistent spatiotemporal relationship, between button presses and reafferent signals, via predictive sensory signaling, a process altered in schizophrenia. Here, we investigated auditory sensory attenuation for a series of morphed voices while healthy participants applied sensorimotor stimulations that had no spatiotemporal relationship to the voice stimuli and that have been shown to induce mild psychosis-like phenomena. In two independent groups of participants, we report a loudening of silent voices and found this effect only during maximal sensorimotor conflicts (versus several control conditions). Importantly, conflicting sensorimotor stimulation also induced a mild psychosis-like state in the form of somatic passivity and participants who experienced stronger passivity lacked the sensorimotor loudening effect. We argue that this conflict-related sensorimotor loudness amplification may represent a reduction of auditory self-attenuation that is lacking in participants experiencing a concomitant mild psychosis-like state. We interpret our results within the framework of the comparator model of sensorimotor control, and discuss the implications of our findings regarding passivity experiences and hallucinations in schizophrenia.
Collapse
Affiliation(s)
- Pavo Orepic
- Laboratory of Cognitive Neuroscience, Center for Neuroprosthetics and Brain Mind Institute, Faculty of Life Sciences, Swiss Federal Institute of Technology (EPFL), Switzerland
| | - Giulio Rognini
- Laboratory of Cognitive Neuroscience, Center for Neuroprosthetics and Brain Mind Institute, Faculty of Life Sciences, Swiss Federal Institute of Technology (EPFL), Switzerland
| | - Oliver Alan Kannape
- Laboratory of Cognitive Neuroscience, Center for Neuroprosthetics and Brain Mind Institute, Faculty of Life Sciences, Swiss Federal Institute of Technology (EPFL), Switzerland
| | - Nathan Faivre
- Univ. Grenoble Alpes, Univ. Savoie Mont Blanc, CNRS, LPNC, 38000 Grenoble, France
| | - Olaf Blanke
- Laboratory of Cognitive Neuroscience, Center for Neuroprosthetics and Brain Mind Institute, Faculty of Life Sciences, Swiss Federal Institute of Technology (EPFL), Switzerland; Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
137
|
Schöbi D, Homberg F, Frässle S, Endepols H, Moran RJ, Friston KJ, Tittgemeyer M, Heinzle J, Stephan KE. Model-based prediction of muscarinic receptor function from auditory mismatch negativity responses. Neuroimage 2021; 237:118096. [PMID: 33940149 DOI: 10.1016/j.neuroimage.2021.118096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 01/09/2023] Open
Abstract
Drugs affecting neuromodulation, for example by dopamine or acetylcholine, take centre stage among therapeutic strategies in psychiatry. These neuromodulators can change both neuronal gain and synaptic plasticity and therefore affect electrophysiological measures. An important goal for clinical diagnostics is to exploit this effect in the reverse direction, i.e., to infer the status of specific neuromodulatory systems from electrophysiological measures. In this study, we provide proof-of-concept that the functional status of cholinergic (specifically muscarinic) receptors can be inferred from electrophysiological data using generative (dynamic causal) models. To this end, we used epidural EEG recordings over two auditory cortical regions during a mismatch negativity (MMN) paradigm in rats. All animals were treated, across sessions, with muscarinic receptor agonists and antagonists at different doses. Together with a placebo condition, this resulted in five levels of muscarinic receptor status. Using a dynamic causal model - embodying a small network of coupled cortical microcircuits - we estimated synaptic parameters and their change across pharmacological conditions. The ensuing parameter estimates associated with (the neuromodulation of) synaptic efficacy showed both graded muscarinic effects and predictive validity between agonistic and antagonistic pharmacological conditions. This finding illustrates the potential utility of generative models of electrophysiological data as computational assays of muscarinic function. In application to EEG data of patients from heterogeneous spectrum diseases, e.g. schizophrenia, such models might help identify subgroups of patients that respond differentially to cholinergic treatments. SIGNIFICANCE STATEMENT: In psychiatry, the vast majority of pharmacological treatments affect actions of neuromodulatory transmitters, e.g. dopamine or acetylcholine. As treatment is largely trial-and-error based, one of the goals for computational psychiatry is to construct mathematical models that can serve as "computational assays" and infer the status of specific neuromodulatory systems in individual patients. This translational neuromodeling strategy has great promise for electrophysiological data in particular but requires careful validation. The present study demonstrates that the functional status of cholinergic (muscarinic) receptors can be inferred from electrophysiological data using dynamic causal models of neural circuits. While accuracy needs to be enhanced and our results must be replicated in larger samples, our current results provide proof-of-concept for computational assays of muscarinic function using EEG.
Collapse
Affiliation(s)
- Dario Schöbi
- Translational Neuromodeling Unit, Institute for Biomedical Engineering, University of Zurich & Swiss Institute of Technology (ETH Zurich), Wilfriedstrasse 6, 8032, Zurich, Switzerland
| | - Fabienne Homberg
- Boston Scientific Medizintechnik GmbH, Daniel-Goldbach-Strasse 17-27, 40880 Ratingen, Germany
| | - Stefan Frässle
- Translational Neuromodeling Unit, Institute for Biomedical Engineering, University of Zurich & Swiss Institute of Technology (ETH Zurich), Wilfriedstrasse 6, 8032, Zurich, Switzerland
| | - Heike Endepols
- Preclinical Imaging Group, Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50923 Cologne, Germany
| | - Rosalyn J Moran
- Department of Neuroimaging, Institute for Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London Se5 8AF, UK
| | - Karl J Friston
- Wellcome Centre for Human Neuroimaging, University College London, 12 Queen Square, London, WC1N, 3AR, UK
| | - Marc Tittgemeyer
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Cluster of Excellence in Cellular Stress and Aging associated Disease (CECAD), 50931 Cologne, Germany
| | - Jakob Heinzle
- Translational Neuromodeling Unit, Institute for Biomedical Engineering, University of Zurich & Swiss Institute of Technology (ETH Zurich), Wilfriedstrasse 6, 8032, Zurich, Switzerland.
| | - Klaas Enno Stephan
- Translational Neuromodeling Unit, Institute for Biomedical Engineering, University of Zurich & Swiss Institute of Technology (ETH Zurich), Wilfriedstrasse 6, 8032, Zurich, Switzerland; Wellcome Centre for Human Neuroimaging, University College London, 12 Queen Square, London, WC1N, 3AR, UK; Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| |
Collapse
|
138
|
Northoff G, Gomez-Pilar J. Overcoming Rest-Task Divide-Abnormal Temporospatial Dynamics and Its Cognition in Schizophrenia. Schizophr Bull 2021; 47:751-765. [PMID: 33305324 PMCID: PMC8661394 DOI: 10.1093/schbul/sbaa178] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Schizophrenia is a complex psychiatric disorder exhibiting alterations in spontaneous and task-related cerebral activity whose relation (termed "state dependence") remains unclear. For unraveling their relationship, we review recent electroencephalographic (and a few functional magnetic resonance imaging) studies in schizophrenia that assess and compare both rest/prestimulus and task states, ie, rest/prestimulus-task modulation. Results report reduced neural differentiation of task-related activity from rest/prestimulus activity across different regions, neural measures, cognitive domains, and imaging modalities. Together, the findings show reduced rest/prestimulus-task modulation, which is mediated by abnormal temporospatial dynamics of the spontaneous activity. Abnormal temporospatial dynamics, in turn, may lead to abnormal prediction, ie, predictive coding, which mediates cognitive changes and psychopathological symptoms, including confusion of internally and externally oriented cognition. In conclusion, reduced rest/prestimulus-task modulation in schizophrenia provides novel insight into the neuronal mechanisms that connect task-related changes to cognitive abnormalities and psychopathological symptoms.
Collapse
Affiliation(s)
- Georg Northoff
- Mental Health Center/7th Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Mind, Brain Imaging and Neuroethics, Institute of Mental Health Research, Royal Ottawa Healthcare Group, University of Ottawa, Ottawa ON, Canada
| | - Javier Gomez-Pilar
- Biomedical Engineering Group, University of Valladolid, Valladolid, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Valladolid, Spain
| |
Collapse
|
139
|
Multimodal assessment of white matter microstructure in antipsychotic-naïve schizophrenia patients and confounding effects of recreational drug use. Brain Imaging Behav 2021; 15:36-48. [PMID: 31909444 DOI: 10.1007/s11682-019-00230-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cerebral white matter (WM) aberrations in schizophrenia have been linked to multiple neurobiological substrates but the underlying mechanisms remain unknown. Moreover, antipsychotic treatment and substance use constitute potential confounders. Multimodal studies using diffusion tensor imaging (DTI) and magnetization transfer imaging (MTI) may provide deeper insight into the whole brain WM pathophysiology in schizophrenia. We combined DTI and MTI to investigate WM integrity in 51 antipsychotic-naïve, first-episode schizophrenia patients and 55 matched healthy controls, using 3 T magnetic resonance imaging (MRI). Psychopathology was assessed with the positive and negative syndrome scale (PANSS). A whole brain partial least squares correlation (PLSC) method was used to conjointly analyze DTI-derived measures (fractional anisotropy (FA), axial diffusivity (AD), radial diffusivity (RD), mode of anisotropy (MO)) and the magnetization transfer ratio (MTR) to identify group differences, and associations with psychopathology. In secondary analyses, we excluded recreational substance users from both groups resulting in 34 patients and 51 healthy controls. The primary PLSC group difference analysis identified a significant pattern of lower FA, AD, MO and higher RD in patients (p = 0.04). This pattern suggests disorganized WM microstructure in patients. The secondary PLSC group difference analysis without recreational substance users revealed a significant pattern of lower FA and higher AD, RD, MO, MTR in patients (p = 0.04). This pattern in the substance free patients is consistent with higher extracellular free-water concentrations, which may reflect neuroinflammation. No significant associations with psychopathology were observed. Recreational substance use appears to be a confounding issue, which calls for attention in future WM studies.
Collapse
|
140
|
Wang LL, Sun X, Chiu CD, Leung PWL, Chan RCK, So SHW. Altered cortico-striatal functional connectivity in people with high levels of schizotypy: A longitudinal resting-state study. Asian J Psychiatr 2021; 58:102621. [PMID: 33676189 DOI: 10.1016/j.ajp.2021.102621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/02/2021] [Accepted: 02/20/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE OF THE RESEARCH Cortico-striatal functional connectivity has been implicated in the neuropathology of schizophrenia. However, the longitudinal relationship between the cortico-striatal connectivity and schizotypy remains unknown. We examined the resting-state fMRI connectivity in 27 individuals with a high level of schizotypy and 20 individuals with a low level of schizotypy at baseline and 18 months later. Correlations between changes in cortico-striatal connectivity and changes in schizotypy scores over time were examined. PRINCIPAL RESULTS We found both increased and decreased cortico-striatal connectivity in individuals with a high level of schizotypy at baseline. Over time, these individuals showed improvement in both the negative and positive schizotypal domains. Changes in striatal-insula connectivity were positively correlated with changes in positive schizotypy from baseline to follow-up. MAJOR CONCLUSIONS Our results suggested impaired cortico-striatal connectivity in individuals with a high level of schizotypy. The dysconnectivity mainly involves the dorsal striatum. The connectivity between the dorsal striatum and the insula may be a putative marker for temporal changes in positive schizotypy.
Collapse
Affiliation(s)
- Ling-Ling Wang
- Neuropsychology and Applied Cognitive Neuroscience Laboratory, CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, The University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoqi Sun
- Department of Psychology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Chui-De Chiu
- Department of Psychology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Patrick W L Leung
- Department of Psychology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Raymond C K Chan
- Neuropsychology and Applied Cognitive Neuroscience Laboratory, CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, The University of Chinese Academy of Sciences, Beijing, China.
| | - Suzanne H W So
- Department of Psychology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
141
|
Zhuo C, Li G, Lin X, Jiang D, Xu Y, Tian H, Wang W, Song X. Strategies to solve the reverse inference fallacy in future MRI studies of schizophrenia: a review. Brain Imaging Behav 2021; 15:1115-1133. [PMID: 32304018 PMCID: PMC8032587 DOI: 10.1007/s11682-020-00284-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Few advances in schizophrenia research have been translated into clinical practice, despite 60 years of serum biomarkers studies and 50 years of genetic studies. During the last 30 years, neuroimaging studies on schizophrenia have gradually increased, partly due to the beautiful prospect that the pathophysiology of schizophrenia could be explained entirely by the Human Connectome Project (HCP). However, the fallacy of reverse inference has been a critical problem of the HCP. For this reason, there is a dire need for new strategies or research "bridges" to further schizophrenia at the biological level. To understand the importance of research "bridges," it is vital to examine the strengths and weaknesses of the recent literature. Hence, in this review, our team has summarized the recent literature (1995-2018) about magnetic resonance imaging (MRI) of schizophrenia in terms of regional and global structural and functional alterations. We have also provided a new proposal that may supplement the HCP for studying schizophrenia. As postulated, despite the vast number of MRI studies in schizophrenia, the lack of homogeneity between the studies, along with the relatedness of schizophrenia with other neurological disorders, has hindered the study of schizophrenia. In addition, the reverse inference cannot be used to diagnose schizophrenia, further limiting the clinical impact of findings from medical imaging studies. We believe that multidisciplinary technologies may be used to develop research "bridges" to further investigate schizophrenia at the single neuron or neuron cluster levels. We have postulated about future strategies for overcoming the current limitations and establishing the research "bridges," with an emphasis on multimodality imaging, molecular imaging, neuron cluster signals, single transmitter biomarkers, and nanotechnology. These research "bridges" may help solve the reverse inference fallacy and improve our understanding of schizophrenia for future studies.
Collapse
Affiliation(s)
- Chuanjun Zhuo
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, 450000, Zhengzhou, China.
- Department of Psychiatry Pattern Recognition, Department of Genetics Laboratory of Schizophrenia, School of Mental Health, Jining Medical University, 272119, Jining, China.
- Department of Psychiatry, Wenzhou Seventh People's Hospital, 325000, Wenzhou, China.
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China.
- MDT Center for Cognitive Impairment and Sleep Disorders, First Hospital of Shanxi Medical University, 030001, Taiyuan, China.
- Department of Psychiatric-Neuroimaging-Genetics and Co-Morbidity Laboratory (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center, Tianjin Medical University Mental Health Teaching Hospital, 300222, Tianjin, China.
- Biological Psychiatry of Co-collaboration Laboratory of China and Canada, Xiamen Xianyue Hospital, University of Alberta, Xiamen Xianyue Hospital, 361000, Xiamen, China.
- Department of Psychiatry, Tianjin Medical University, 300075, Tianjin, China.
- Psychiatric-Neuroimaging-Genetics-Comorbidity Laboratory (PNGC_Lab), Tianjin Anding Hospital, Department of Psychiatry, Tianjin Mental Health Centre, Mental Health Teaching Hospital of Tianjin Medical University, Shanxi Medical University, 300222, Tianjin, China.
| | - Gongying Li
- Department of Psychiatry Pattern Recognition, Department of Genetics Laboratory of Schizophrenia, School of Mental Health, Jining Medical University, 272119, Jining, China
| | - Xiaodong Lin
- Department of Psychiatry, Wenzhou Seventh People's Hospital, 325000, Wenzhou, China
| | - Deguo Jiang
- Department of Psychiatry, Wenzhou Seventh People's Hospital, 325000, Wenzhou, China
| | - Yong Xu
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
- MDT Center for Cognitive Impairment and Sleep Disorders, First Hospital of Shanxi Medical University, 030001, Taiyuan, China
| | - Hongjun Tian
- Department of Psychiatric-Neuroimaging-Genetics and Co-Morbidity Laboratory (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center, Tianjin Medical University Mental Health Teaching Hospital, 300222, Tianjin, China
| | - Wenqiang Wang
- Biological Psychiatry of Co-collaboration Laboratory of China and Canada, Xiamen Xianyue Hospital, University of Alberta, Xiamen Xianyue Hospital, 361000, Xiamen, China
| | - Xueqin Song
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, 450000, Zhengzhou, China
- Psychiatric-Neuroimaging-Genetics-Comorbidity Laboratory (PNGC_Lab), Tianjin Anding Hospital, Department of Psychiatry, Tianjin Mental Health Centre, Mental Health Teaching Hospital of Tianjin Medical University, Shanxi Medical University, 300222, Tianjin, China
| |
Collapse
|
142
|
Abstract
This paper introduces a new construct, the 'pivotal mental state', which is defined as a hyper-plastic state aiding rapid and deep learning that can mediate psychological transformation. We believe this new construct bears relevance to a broad range of psychological and psychiatric phenomena. We argue that pivotal mental states serve an important evolutionary function, that is, to aid psychological transformation when actual or perceived environmental pressures demand this. We cite evidence that chronic stress and neurotic traits are primers for a pivotal mental state, whereas acute stress can be a trigger. Inspired by research with serotonin 2A receptor agonist psychedelics, we highlight how activity at this particular receptor can robustly and reliably induce pivotal mental states, but we argue that the capacity for pivotal mental states is an inherent property of the human brain itself. Moreover, we hypothesize that serotonergic psychedelics hijack a system that has evolved to mediate rapid and deep learning when its need is sensed. We cite a breadth of evidences linking stress via a variety of inducers, with an upregulated serotonin 2A receptor system (e.g. upregulated availability of and/or binding to the receptor) and acute stress with 5-HT release, which we argue can activate this primed system to induce a pivotal mental state. The pivotal mental state model is multi-level, linking a specific molecular gateway (increased serotonin 2A receptor signaling) with the inception of a hyper-plastic brain and mind state, enhanced rate of associative learning and the potential mediation of a psychological transformation.
Collapse
Affiliation(s)
- Ari Brouwer
- Centre for Psychedelic Research, Imperial College London, London, United Kingdom
| | | |
Collapse
|
143
|
Prolonged deficit of low gamma oscillations in the peri-infarct cortex of mice after stroke. Exp Neurol 2021; 341:113696. [PMID: 33727098 DOI: 10.1016/j.expneurol.2021.113696] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 03/04/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023]
Abstract
Days and weeks after an ischemic stroke, the peri-infarct area adjacent to the necrotic tissue exhibits very intense synaptic reorganization aimed at regaining lost functions. In order to enhance functional recovery, it is important to understand the mechanisms supporting neural repair and neuroplasticity in the cortex surrounding the lesion. Brain oscillations of the local field potential (LFP) are rhythmic fluctuations of neuronal excitability that synchronize neuronal activity to organize information processing and plasticity. Although the oscillatory activity of the brain has been probed after stroke in both animals and humans using electroencephalography (EEG), the latter is ineffective to precisely map the oscillatory changes in the peri-infarct zone where synaptic plasticity potential is high. Here, we worked on the hypothesis that the brain oscillatory system is altered in the surviving peri-infarct cortex, which may slow down the functional repair and reduce the recovery. In order to document the relevance of this hypothesis, oscillatory power was measured at various distances from the necrotic core at 7 and 21 days after a permanent cortical ischemia induced in mice. Delta and theta oscillations remained at a normal power in the peri-infarct cortex, in contrast to low gamma oscillations that displayed a gradual decrease, when approaching the border of the lesion. A broadband increase of power was also observed in the homotopic contralateral sites. Thus, the proximal peri-infarct cortex could become a target of therapeutic interventions applied to correct the oscillatory regimen in order to boost post-stroke functional recovery.
Collapse
|
144
|
Hämäläinen M, Huang M, Bowyer SM. Magnetoencephalography Signal Processing, Forward Modeling, Magnetoencephalography Inverse Source Imaging, and Coherence Analysis. Neuroimaging Clin N Am 2021; 30:125-143. [PMID: 32336402 DOI: 10.1016/j.nic.2020.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Magnetoencephalography (MEG) is a noninvasive functional imaging technique for the brain. MEG directly measures the magnetic signal due to neuronal activation in gray matter with high spatial localization accuracy. The first part of this article covers the overall concepts of MEG and the forward and inverse modeling techniques. It is followed by examples of analyzing evoked and resting-state MEG signals using a high-resolution MEG source imaging technique. Next, different techniques for connectivity and network analysis are reviewed with examples showing connectivity estimates from resting-state and epileptic activity.
Collapse
Affiliation(s)
- Matti Hämäläinen
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, 149 13th Street, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA, USA
| | - Mingxiong Huang
- Department of Radiology, UCSD Radiology Imaging Lab, University of California, San Diego, 3510 Dunhill Street, San Diego, CA 92121, USA
| | - Susan M Bowyer
- Department of Neurology, MEG Lab, Henry Ford Hospital, 2799 West Grand Boulevard, CFP 079, Detroit, MI 48202, USA; Wayne State University School of Medicine, Detroit, MI, USA; Department of Physics, Oakland University, Rochester, MI, USA.
| |
Collapse
|
145
|
Masychev K, Ciprian C, Ravan M, Reilly JP, MacCrimmon D. Advanced Signal Processing Methods for Characterization of Schizophrenia. IEEE Trans Biomed Eng 2021; 68:1123-1130. [PMID: 33656984 DOI: 10.1109/tbme.2020.3011842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Schizophrenia is a severe mental disorder associated with nerobiological deficits. Auditory oddball P300 have been found to be one of the most consistent markers of schizophrenia. The goal of this study is to find quantitative features that can objectively distinguish patients with schizophrenia (SCZs) from healthy controls (HCs) based on their recorded auditory odd-ball P300 electroencephalogram (EEG) data. METHODS Using EEG dataset, we develop a machine learning (ML) algorithm to distinguish 57 SCZs from 66 HCs. The proposed ML algorithm has three steps. In the first step, a brain source localization (BSL) procedure using the linearly constrained minimum variance (LCMV) beamforming approach is employed on EEG signals to extract source waveforms from 30 specified brain regions. In the second step, a method for estimating effective connectivity, referred to as symbolic transfer entropy (STE), is applied to the source waveforms. In the third step the ML algorithm is applied to the STE connectivity matrix to determine whether a set of features can be found that successfully discriminate SCZ from HC. RESULTS The findings revealed that the SCZs have significantly higher effective connectivity compared to HCs and the selected STE features could achieve an accuracy of 92.68%, with a sensitivity of 92.98% and specificity of 92.42%. CONCLUSION The findings imply that the extracted features are from the regions that are mainly affected by SCZ and can be used to distinguish SCZs from HCs. SIGNIFICANCE The proposed ML algorithm may prove to be a promising tool for the clinical diagnosis of schizophrenia.
Collapse
|
146
|
Chan MMY, Yau SSY, Han YMY. The neurobiology of prefrontal transcranial direct current stimulation (tDCS) in promoting brain plasticity: A systematic review and meta-analyses of human and rodent studies. Neurosci Biobehav Rev 2021; 125:392-416. [PMID: 33662444 DOI: 10.1016/j.neubiorev.2021.02.035] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/05/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022]
Abstract
The neurobiological mechanisms underlying prefrontal transcranial direct current stimulation (tDCS) remain elusive. Randomized, sham-controlled trials in humans and rodents applying in vivo prefrontal tDCS were included to explore whether prefrontal tDCS modulates resting-state and event-related functional connectivity, neural oscillation and synaptic plasticity. Fifty studies were included in the systematic review and 32 in the meta-analyses. Neuroimaging meta-analysis indicated anodal prefrontal tDCS significantly enhanced bilateral median cingulate activity [familywise error (FWE)-corrected p < .005]; meta-regression revealed a positive relationship between changes in median cingulate activity after tDCS and current density (FWE-corrected p < .005) as well as electric current strength (FWE-corrected p < .05). Meta-analyses of electroencephalography and magnetoencephalography data revealed nonsignificant changes (ps > .1) in both resting-state and event-related oscillatory power across all frequency bands. Applying anodal tDCS over the rodent hippocampus/prefrontal cortex enhanced long-term potentiation and brain-derived neurotrophic factor expression in the stimulated brain regions (ps <.005). Evidence supporting prefrontal tDCS administration is preliminary; more methodologically consistent studies evaluating its effects on cognitive function that include brain activity measurements are needed.
Collapse
Affiliation(s)
- Melody M Y Chan
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Sonata S Y Yau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Yvonne M Y Han
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong SAR, China.
| |
Collapse
|
147
|
Grove TB, Lasagna CA, Martínez-Cancino R, Pamidighantam P, Deldin PJ, Tso IF. Neural Oscillatory Abnormalities During Gaze Processing in Schizophrenia: Evidence of Reduced Theta Phase Consistency and Inter-areal Theta-Gamma Coupling. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2021; 6:370-379. [PMID: 33160880 PMCID: PMC7917157 DOI: 10.1016/j.bpsc.2020.08.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Abnormal gaze discrimination in schizophrenia (SZ) is associated with impairment in social functioning, but the neural mechanisms remain unclear. Evidence suggests that local neural oscillations and inter-areal communication through neural synchronization are critical physiological mechanisms supporting basic and complex cognitive processes. The roles of these mechanisms in abnormal gaze processing in SZ have not been investigated. The present study examined local neural oscillations and connectivity between anterior and bilateral posterior brain areas during gaze processing. METHODS During electroencephalography recording, 28 participants with SZ and 34 healthy control participants completed a gaze discrimination task. Time-frequency decomposition of electroencephalography data was used to examine neural oscillatory power and intertrial phase consistency at bilateral posterior and midline anterior scalp sites. In addition, connectivity between these anterior and posterior sites, in terms of cross-frequency coupling between theta phase and gamma amplitude, was examined using the Kullback-Leibler Modulation Index. RESULTS Participants with SZ showed reduced total power of theta-band activity relative to healthy control participants at all sites examined. This group difference could be accounted for by reduced intertrial phase consistency of theta activity in SZ participants, which was related to reduced gaze discrimination accuracy in SZ. In addition, SZ participants exhibited reduced Kullback-Leibler indexing, both feedforward and feedback connectivity, between the posterior and anterior sites. CONCLUSIONS These findings suggest that abnormal theta phase consistency and dysconnection between posterior face processing and anterior areas may underlie gaze processing deficits in SZ.
Collapse
Affiliation(s)
- Tyler B Grove
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan
| | - Carly A Lasagna
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan
| | - Ramón Martínez-Cancino
- Swartz Center for Computational Neuroscience, University of California San Diego, La Jolla, California
| | | | - Patricia J Deldin
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Michigan, Ann Arbor, Michigan
| | - Ivy F Tso
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan; Department of Psychology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
148
|
Barone A, Signoriello S, Latte G, Vellucci L, Giordano G, Avagliano C, Buonaguro EF, Marmo F, Tomasetti C, Iasevoli F, de Bartolomeis A. Modulation of glutamatergic functional connectivity by a prototypical antipsychotic: Translational inference from a postsynaptic density immediate-early gene-based network analysis. Behav Brain Res 2021; 404:113160. [PMID: 33577880 DOI: 10.1016/j.bbr.2021.113160] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 01/30/2021] [Accepted: 01/31/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Although extensively studied, the effect of antipsychotics is not completely understood at a network level. We tested the hypothesis that acute administration of haloperidol would modulate functional connectivity of brain regions relevant to schizophrenia pathophysiology. To assess putative changes in brain network properties and regional interactivity, we studied the expression of Homer1a, an Immediate Early Gene (IEG) demonstrated to be induced by antipsychotic administration and coding for a protein involved in glutamatergic synapses remodeling. METHODS Sprague-Dawley rats (n = 26) assigned to vehicle (VEH; NaCl 0.9%) or haloperidol (HAL; 0.8 mg/kg) were included in the network analysis. Homer1a mRNA induction was evaluated by in situ hybridization. Signal intensity analysis was performed in 33 Regions of Interest (ROIs) in the cortex, the caudate putamen, and the nucleus accumbens. A signal correlation analysis was performed, computing all possible pairwise Pearson correlations among ROIs in the two groups. Two networks were generated for HAL and VEH groups, and their properties and topography were explored. RESULTS VEH and HAL networks showed qualitative differences in global efficiency and clustering coefficient. The HAL network showed enhanced interactivity between cortical and striatal regions, and within caudate putamen subdivisions. On the other hand, it exhibited reduced inter-correlations between cingulate cortex and anterior insula and caudate putamen and nucleus accumbens. Moreover, haloperidol was able to modulate centrality of crucial functional hubs. These preclinical results corroborate and expand the clinical evidence that antipsychotics may modulate specific brain network properties and disease-related circuits' interactivity.
Collapse
Affiliation(s)
- Annarita Barone
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science, and Odontostomatology, University of Naples Federico II, 80131, Napoli, Italy
| | - Simona Signoriello
- Medical Statistics Unit, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Gianmarco Latte
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science, and Odontostomatology, University of Naples Federico II, 80131, Napoli, Italy
| | - Licia Vellucci
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science, and Odontostomatology, University of Naples Federico II, 80131, Napoli, Italy
| | - Giuseppe Giordano
- Department of Social and Political Studies, University of Salerno, 84084, Fisciano, SA, Italy
| | - Camilla Avagliano
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science, and Odontostomatology, University of Naples Federico II, 80131, Napoli, Italy
| | - Elisabetta F Buonaguro
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science, and Odontostomatology, University of Naples Federico II, 80131, Napoli, Italy
| | - Federica Marmo
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science, and Odontostomatology, University of Naples Federico II, 80131, Napoli, Italy
| | - Carmine Tomasetti
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science, and Odontostomatology, University of Naples Federico II, 80131, Napoli, Italy
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science, and Odontostomatology, University of Naples Federico II, 80131, Napoli, Italy
| | - Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science, and Odontostomatology, University of Naples Federico II, 80131, Napoli, Italy.
| |
Collapse
|
149
|
Chen J, Müller VI, Dukart J, Hoffstaedter F, Baker JT, Holmes AJ, Vatansever D, Nickl-Jockschat T, Liu X, Derntl B, Kogler L, Jardri R, Gruber O, Aleman A, Sommer IE, Eickhoff SB, Patil KR. Intrinsic Connectivity Patterns of Task-Defined Brain Networks Allow Individual Prediction of Cognitive Symptom Dimension of Schizophrenia and Are Linked to Molecular Architecture. Biol Psychiatry 2021; 89:308-319. [PMID: 33357631 PMCID: PMC7770333 DOI: 10.1016/j.biopsych.2020.09.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Despite the marked interindividual variability in the clinical presentation of schizophrenia, the extent to which individual dimensions of psychopathology relate to the functional variability in brain networks among patients remains unclear. Here, we address this question using network-based predictive modeling of individual psychopathology along 4 data-driven symptom dimensions. Follow-up analyses assess the molecular underpinnings of predictive networks by relating them to neurotransmitter-receptor distribution patterns. METHODS We investigated resting-state functional magnetic resonance imaging data from 147 patients with schizophrenia recruited at 7 sites. Individual expression along negative, positive, affective, and cognitive symptom dimensions was predicted using a relevance vector machine based on functional connectivity within 17 meta-analytic task networks following repeated 10-fold cross-validation and leave-one-site-out analyses. Results were validated in an independent sample. Networks robustly predicting individual symptom dimensions were spatially correlated with density maps of 9 receptors/transporters from prior molecular imaging in healthy populations. RESULTS Tenfold and leave-one-site-out analyses revealed 5 predictive network-symptom associations. Connectivity within theory of mind, cognitive reappraisal, and mirror neuron networks predicted negative, positive, and affective symptom dimensions, respectively. Cognitive dimension was predicted by theory of mind and socioaffective default networks. Importantly, these predictions generalized to the independent sample. Intriguingly, these two networks were positively associated with D1 receptor and serotonin reuptake transporter densities as well as dopamine synthesis capacity. CONCLUSIONS We revealed a robust association between intrinsic functional connectivity within networks for socioaffective processes and the cognitive dimension of psychopathology. By investigating the molecular architecture, this work links dopaminergic and serotonergic systems with the functional topography of brain networks underlying cognitive symptoms in schizophrenia.
Collapse
Affiliation(s)
- Ji Chen
- Institute of Neuroscience and Medicine: Brain and Behavior (INM-7), Research Center Jülich, Jülich, Germany; Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Veronika I. Müller
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany,Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Juergen Dukart
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany,Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Felix Hoffstaedter
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany,Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Justin T. Baker
- Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA 02478,Department of Psychiatry, Harvard Medical School, Boston, MA 02114
| | - Avram J. Holmes
- Department of Psychology, Yale University, New Haven, CT 06520
| | - Deniz Vatansever
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 200433, Shanghai, PR China
| | - Thomas Nickl-Jockschat
- Iowa Neuroscience Institute & Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Xiaojin Liu
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany,Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Birgit Derntl
- Department of Psychiatry and Psychotherapy, Medical School, University of Tübingen, Germany
| | - Lydia Kogler
- Department of Psychiatry and Psychotherapy, Medical School, University of Tübingen, Germany
| | - Renaud Jardri
- Univ Lille, INSERM U1172, Lille Neuroscience & Cognition Centre, Plasticity & SubjectivitY team & CHU Lille, Fontan Hospital, CURE platform, Lille, France
| | - Oliver Gruber
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, Heidelberg University, Germany
| | - André Aleman
- Department of Neuroscience, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Iris E. Sommer
- Department of Biomedical Science of Cells and Systems, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Simon B. Eickhoff
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany,Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany,Correspondence should be addressed to: Simon B. Eickhoff, Institute of Systems Neuroscience, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany & Institute of Neuroscience and Medicine, Brain and Behaviour (INM-7), Research Center Jülich, 52428 Jülich, Germany. Tel: +49 2461 61 1791; .; Ji Chen, Institute of Systems Neuroscience, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany & Institute of Neuroscience and Medicine, Brain and Behaviour (INM-7), Research Center Jülich, 52428 Jülich, Germany. Tel: +49 2461 61 85334;
| | - Kaustubh R. Patil
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany,Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
150
|
Elyamany O, Leicht G, Herrmann CS, Mulert C. Transcranial alternating current stimulation (tACS): from basic mechanisms towards first applications in psychiatry. Eur Arch Psychiatry Clin Neurosci 2021; 271:135-156. [PMID: 33211157 PMCID: PMC7867505 DOI: 10.1007/s00406-020-01209-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022]
Abstract
Transcranial alternating current stimulation (tACS) is a unique form of non-invasive brain stimulation. Sinusoidal alternating electric currents are delivered to the scalp to affect mostly cortical neurons. tACS is supposed to modulate brain function and, in turn, cognitive processes by entraining brain oscillations and inducing long-term synaptic plasticity. Therefore, tACS has been investigated in cognitive neuroscience, but only recently, it has been also introduced in psychiatric clinical trials. This review describes current concepts and first findings of applying tACS as a potential therapeutic tool in the field of psychiatry. The current understanding of its mechanisms of action is explained, bridging cellular neuronal activity and the brain network mechanism. Revisiting the relevance of altered brain oscillations found in six major psychiatric disorders, putative targets for the management of mental disorders using tACS are discussed. A systematic literature search on PubMed was conducted to report findings of the clinical studies applying tACS in patients with psychiatric conditions. In conclusion, the initial results may support the feasibility of tACS in clinical psychiatric populations without serious adverse events. Moreover, these results showed the ability of tACS to reset disturbed brain oscillations, and thus to improve behavioural outcomes. In addition to its potential therapeutic role, the reactivity of the brain circuits to tACS could serve as a possible tool to determine the diagnosis, classification or prognosis of psychiatric disorders. Future double-blind randomised controlled trials are necessary to answer currently unresolved questions. They may aim to detect response predictors and control for various confounding factors.
Collapse
Affiliation(s)
- Osama Elyamany
- Centre of Psychiatry, Justus-Liebig University, Klinikstrasse 36, 35392, Giessen, Hessen, Germany
- Centre for Mind, Brain and Behaviour (CMBB), University of Marburg and Justus-Liebig University Giessen, Marburg, Germany
| | - Gregor Leicht
- Department of Psychiatry and Psychotherapy, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph S Herrmann
- Experimental Psychology Lab, Centre for Excellence "Hearing4all," European Medical School, University of Oldenburg, Oldenburg, Lower Saxony, Germany
- Research Centre Neurosensory Science, University of Oldenburg, Oldenburg, Lower Saxony, Germany
| | - Christoph Mulert
- Centre of Psychiatry, Justus-Liebig University, Klinikstrasse 36, 35392, Giessen, Hessen, Germany.
- Centre for Mind, Brain and Behaviour (CMBB), University of Marburg and Justus-Liebig University Giessen, Marburg, Germany.
| |
Collapse
|