Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 199
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Psychiatry ; 28(10): 4363-4373, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37644174

ABSTRACT

Converging evidence suggests that schizophrenia (SZ) with primary, enduring negative symptoms (i.e., Deficit SZ (DSZ)) represents a distinct entity within the SZ spectrum while the neurobiological underpinnings remain undetermined. In the largest dataset of DSZ and Non-Deficit (NDSZ), we conducted a meta-analysis of data from 1560 individuals (168 DSZ, 373 NDSZ, 1019 Healthy Controls (HC)) and a mega-analysis of a subsampled data from 944 individuals (115 DSZ, 254 NDSZ, 575 HC) collected across 9 worldwide research centers of the ENIGMA SZ Working Group (8 in the mega-analysis), to clarify whether they differ in terms of cortical morphology. In the meta-analysis, sites computed effect sizes for differences in cortical thickness and surface area between SZ and control groups using a harmonized pipeline. In the mega-analysis, cortical values of individuals with schizophrenia and control participants were analyzed across sites using mixed-model ANCOVAs. The meta-analysis of cortical thickness showed a converging pattern of widespread thinner cortex in fronto-parietal regions of the left hemisphere in both DSZ and NDSZ, when compared to HC. However, DSZ have more pronounced thickness abnormalities than NDSZ, mostly involving the right fronto-parietal cortices. As for surface area, NDSZ showed differences in fronto-parietal-temporo-occipital cortices as compared to HC, and in temporo-occipital cortices as compared to DSZ. Although DSZ and NDSZ show widespread overlapping regions of thinner cortex as compared to HC, cortical thinning seems to better typify DSZ, being more extensive and bilateral, while surface area alterations are more evident in NDSZ. Our findings demonstrate for the first time that DSZ and NDSZ are characterized by different neuroimaging phenotypes, supporting a nosological distinction between DSZ and NDSZ and point toward the separate disease hypothesis.


Subject(s)
Schizophrenia , Humans , Schizophrenia/genetics , Magnetic Resonance Imaging , Neuroimaging , Parietal Lobe , Syndrome , Cerebral Cortex/diagnostic imaging
2.
Hum Brain Mapp ; 44(6): 2620-2635, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36840728

ABSTRACT

Resting-state functional network connectivity (rsFNC) has shown utility for identifying characteristic functional brain patterns in individuals with psychiatric and mood disorders, providing a promising avenue for biomarker development. However, several factors have precluded widespread clinical adoption of rsFNC diagnostics, namely a lack of standardized approaches for capturing comparable and reproducible imaging markers across individuals, as well as the disagreement on the amount of data required to robustly detect intrinsic connectivity networks (ICNs) and diagnostically relevant patterns of rsFNC at the individual subject level. Recently, spatially constrained independent component analysis (scICA) has been proposed as an automated method for extracting ICNs standardized to a chosen network template while still preserving individual variation. Leveraging the scICA methodology, which solves the former challenge of standardized neuroimaging markers, we investigate the latter challenge of identifying a minimally sufficient data length for clinical applications of resting-state fMRI (rsfMRI). Using a dataset containing rsfMRI scans of individuals with schizophrenia and controls (M = 310) as well as simulated rsfMRI, we evaluated the robustness of ICN and rsFNC estimates at both the subject- and group-level, as well as the performance of diagnostic classification, with respect to the length of the rsfMRI time course. We found individual estimates of ICNs and rsFNC from the full-length (5 min) reference time course were sufficiently approximated with just 3-3.5 min of data (r = 0.85, 0.88, respectively), and significant differences in group-average rsFNC could be sufficiently approximated with even less data, just 2 min (r = 0.86). These results from the shorter clinical data were largely consistent with the results from validation experiments using longer time series from both simulated (30 min) and real-world (14 min) datasets, in which estimates of subject-level FNC were reliably estimated with 3-5 min of data. Moreover, in the real-world data we found rsFNC and ICN estimates generated across the full range of data lengths (0.5-14 min) more reliably matched those generated from the first 5 min of scan time than those generated from the last 5 min, suggesting increased influence of "late scan" noise factors such as fatigue or drowsiness may limit the reliability of FNC from data collected after 10+ min of scan time, further supporting the notion of shorter scans. Lastly, a diagnostic classification model trained on just 2 min of data retained 97%-98% classification accuracy relative to that of the full-length reference model. Our results suggest that, when decomposed with scICA, rsfMRI scans of just 2-5 min show good clinical utility without significant loss of individual FNC information of longer scan lengths.


Subject(s)
Brain , Magnetic Resonance Imaging , Humans , Magnetic Resonance Imaging/methods , Reproducibility of Results , Brain/diagnostic imaging , Neuroimaging , Mood Disorders , Brain Mapping/methods
3.
Hum Brain Mapp ; 43(15): 4556-4566, 2022 10 15.
Article in English | MEDLINE | ID: mdl-35762454

ABSTRACT

In this work, we focus on explicitly nonlinear relationships in functional networks. We introduce a technique using normalized mutual information (NMI) that calculates the nonlinear relationship between different brain regions. We demonstrate our proposed approach using simulated data and then apply it to a dataset previously studied by Damaraju et al. This resting-state fMRI data included 151 schizophrenia patients and 163 age- and gender-matched healthy controls. We first decomposed these data using group independent component analysis (ICA) and yielded 47 functionally relevant intrinsic connectivity networks. Our analysis showed a modularized nonlinear relationship among brain functional networks that was particularly noticeable in the sensory and visual cortex. Interestingly, the modularity appears both meaningful and distinct from that revealed by the linear approach. Group analysis identified significant differences in explicitly nonlinear functional network connectivity (FNC) between schizophrenia patients and healthy controls, particularly in the visual cortex, with controls showing more nonlinearity (i.e., higher normalized mutual information between time courses with linear relationships removed) in most cases. Certain domains, including subcortical and auditory, showed relatively less nonlinear FNC (i.e., lower normalized mutual information), whereas links between the visual and other domains showed evidence of substantial nonlinear and modular properties. Overall, these results suggest that quantifying nonlinear dependencies of functional connectivity may provide a complementary and potentially important tool for studying brain function by exposing relevant variation that is typically ignored. Beyond this, we propose a method that captures both linear and nonlinear effects in a "boosted" approach. This method increases the sensitivity to group differences compared to the standard linear approach, at the cost of being unable to separate linear and nonlinear effects.


Subject(s)
Schizophrenia , Visual Cortex , Brain/diagnostic imaging , Brain Mapping , Humans , Magnetic Resonance Imaging/methods , Rest , Schizophrenia/diagnostic imaging , Visual Cortex/diagnostic imaging
4.
Hum Brain Mapp ; 43(1): 352-372, 2022 01.
Article in English | MEDLINE | ID: mdl-34498337

ABSTRACT

Schizophrenia is associated with widespread alterations in subcortical brain structure. While analytic methods have enabled more detailed morphometric characterization, findings are often equivocal. In this meta-analysis, we employed the harmonized ENIGMA shape analysis protocols to collaboratively investigate subcortical brain structure shape differences between individuals with schizophrenia and healthy control participants. The study analyzed data from 2,833 individuals with schizophrenia and 3,929 healthy control participants contributed by 21 worldwide research groups participating in the ENIGMA Schizophrenia Working Group. Harmonized shape analysis protocols were applied to each site's data independently for bilateral hippocampus, amygdala, caudate, accumbens, putamen, pallidum, and thalamus obtained from T1-weighted structural MRI scans. Mass univariate meta-analyses revealed more-concave-than-convex shape differences in the hippocampus, amygdala, accumbens, and thalamus in individuals with schizophrenia compared with control participants, more-convex-than-concave shape differences in the putamen and pallidum, and both concave and convex shape differences in the caudate. Patterns of exaggerated asymmetry were observed across the hippocampus, amygdala, and thalamus in individuals with schizophrenia compared to control participants, while diminished asymmetry encompassed ventral striatum and ventral and dorsal thalamus. Our analyses also revealed that higher chlorpromazine dose equivalents and increased positive symptom levels were associated with patterns of contiguous convex shape differences across multiple subcortical structures. Findings from our shape meta-analysis suggest that common neurobiological mechanisms may contribute to gray matter reduction across multiple subcortical regions, thus enhancing our understanding of the nature of network disorganization in schizophrenia.


Subject(s)
Amygdala/pathology , Corpus Striatum/pathology , Hippocampus/pathology , Neuroimaging , Schizophrenia/pathology , Thalamus/pathology , Amygdala/diagnostic imaging , Corpus Striatum/diagnostic imaging , Hippocampus/diagnostic imaging , Humans , Multicenter Studies as Topic , Schizophrenia/diagnostic imaging , Thalamus/diagnostic imaging
5.
Drug Dev Res ; 82(5): 678-684, 2021 08.
Article in English | MEDLINE | ID: mdl-32394511

ABSTRACT

Tardive dyskinesia (TD) is a potentially irreversible movement disorder observed following long-term antipsychotic exposure. Its cause is unknown; however, a genetic component has been supported by studies of affected families. Dysbindin-1, encoded by the dystrobrevin-binding protein 1 DTNBP1 gene, has been associated with schizophrenia and is potentially involved in dopamine neurotransmission through its regulation of dopamine release and dopamine D2 receptor recycling, making it a candidate for investigation in TD. We investigated common variants across the DTNBP1 gene in our schizophrenia/patients with schizoaffective disorder of European ancestry. We found a number of DTNBP1 three-marker haplotypes to be associated with TD occurrence and TD severity (p < 0.05). These preliminary findings, if replicated in larger independent samples, would suggest that drugs targeting dysbindin-1 may be an option in the prevention and treatment of TD.


Subject(s)
Dysbindin/genetics , Schizophrenia/drug therapy , Schizophrenia/genetics , Tardive Dyskinesia/chemically induced , Tardive Dyskinesia/genetics , Adult , Female , Genetic Predisposition to Disease , Genotype , Haplotypes , Humans , Male
6.
NMR Biomed ; 33(6): e4294, 2020 06.
Article in English | MEDLINE | ID: mdl-32207187

ABSTRACT

The human brain is asymmetrically lateralized for certain functions (such as language processing) to regions in one hemisphere relative to the other. Asymmetries are measured with a laterality index (LI). However, traditional LI measures are limited by a lack of consensus on metrics used for its calculation. To address this limitation, source-based laterality (SBL) leverages an independent component analysis for the identification of laterality-specific alterations, identifying covarying components between hemispheres across subjects. SBL is successfully implemented with simulated data with inherent differences in laterality. SBL is then compared with a voxel-wise analysis utilizing structural data from a sample of patients with schizophrenia and controls without schizophrenia. SBL group comparisons identified three distinct temporal regions and one cerebellar region with significantly altered laterality in patients with schizophrenia relative to controls. Previous work highlights reductions in laterality (ie, reduced left gray matter volume) in patients with schizophrenia compared with controls without schizophrenia. Results from this pilot SBL project are the first, to our knowledge, to identify covarying laterality differences within discrete temporal brain regions. The authors argue SBL provides a unique focus to detect covarying laterality differences in patients with schizophrenia, facilitating the discovery of laterality aspects undetected in previous work.


Subject(s)
Functional Laterality , Schizophrenia/pathology , Schizophrenia/physiopathology , Temporal Lobe/pathology , Temporal Lobe/physiopathology , Adolescent , Adult , Brain Mapping , Computer Simulation , Female , Humans , Linear Models , Male , Middle Aged , Nerve Net/physiopathology , Statistics, Nonparametric , Young Adult
7.
Psychol Med ; 50(8): 1267-1277, 2020 06.
Article in English | MEDLINE | ID: mdl-31155012

ABSTRACT

BACKGROUND: Schizophrenia is associated with robust hippocampal volume deficits but subregion volume deficits, their associations with cognition, and contributing genes remain to be determined. METHODS: Hippocampal formation (HF) subregion volumes were obtained using FreeSurfer 6.0 from individuals with schizophrenia (n = 176, mean age ± s.d. = 39.0 ± 11.5, 132 males) and healthy volunteers (n = 173, mean age ± s.d. = 37.6 ± 11.3, 123 males) with similar mean age, gender, handedness, and race distributions. Relationships between the HF subregion volume with the largest between group difference, neuropsychological performance, and single-nucleotide polymorphisms were assessed. RESULTS: This study found a significant group by region interaction on hippocampal subregion volumes. Compared to healthy volunteers, individuals with schizophrenia had significantly smaller dentate gyrus (DG) (Cohen's d = -0.57), Cornu Ammonis (CA) 4, molecular layer of the hippocampus, hippocampal tail, and CA 1 volumes, when statistically controlling for intracranial volume; DG (d = -0.43) and CA 4 volumes remained significantly smaller when statistically controlling for mean hippocampal volume. DG volume showed the largest between group difference and significant positive associations with visual memory and speed of processing in the overall sample. Genome-wide association analysis with DG volume as the quantitative phenotype identified rs56055643 (ß = 10.8, p < 5 × 10-8, 95% CI 7.0-14.5) on chromosome 3 in high linkage disequilibrium with MOBP. Gene-based analyses identified associations between SLC25A38 and RPSA and DG volume. CONCLUSIONS: This study suggests that DG dysfunction is fundamentally involved in schizophrenia pathophysiology, that it may contribute to cognitive abnormalities in schizophrenia, and that underlying biological mechanisms may involve contributions from MOBP, SLC25A38, and RPSA.


Subject(s)
Dentate Gyrus/pathology , Schizophrenia/genetics , Schizophrenia/pathology , Adult , Case-Control Studies , Cognition , Female , Genome-Wide Association Study , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Mitochondrial Membrane Transport Proteins/genetics , Myelin Proteins/genetics , Organ Size , Receptors, Laminin/genetics , Regression Analysis , Ribosomal Proteins/genetics
8.
Neuroimage ; 184: 843-854, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30300752

ABSTRACT

Multimodal, imaging-genomics techniques offer a platform for understanding genetic influences on brain abnormalities in psychiatric disorders. Such approaches utilize the information available from both imaging and genomics data and identify their association. Particularly for complex disorders such as schizophrenia, the relationship between imaging and genomic features may be better understood by incorporating additional information provided by advanced multimodal modeling. In this study, we propose a novel framework to combine features corresponding to functional magnetic resonance imaging (functional) and single nucleotide polymorphism (SNP) data from 61 schizophrenia (SZ) patients and 87 healthy controls (HC). In particular, the features for the functional and genetic modalities include dynamic (i.e., time-varying) functional network connectivity (dFNC) features and the SNP data, respectively. The dFNC features are estimated from component time-courses, obtained using group independent component analysis (ICA), by computing sliding-window functional network connectivity, and then estimating subject specific states from this dFNC data using a k-means clustering approach. For each subject, both the functional (dFNC states) and SNP data are selected as features for a parallel ICA (pICA) based imaging-genomic framework. This analysis identified a significant association between a SNP component (defined by large clusters of functionally related SNPs statistically correlated with phenotype components) and time-varying or dFNC component (defined by clusters of related connectivity links among distant brain regions distributed across discrete dynamic states, and statistically correlated with genomic components) in schizophrenia. Importantly, the polygenetic risk score (PRS) for SZ (computed as a linearly weighted sum of the genotype profiles with weights derived from the odds ratios of the psychiatric genomics consortium (PGC)) was negatively correlated with the significant dFNC component, which were mostly present within a state that exhibited a lower occupancy rate in individuals with SZ compared with HC, hence identifying a potential dFNC imaging biomarker for schizophrenia. Taken together, the current findings provide preliminary evidence for a link between dFNC measures and genetic risk, suggesting the application of dFNC patterns as biomarkers in imaging genetic association study.


Subject(s)
Brain Mapping/methods , Brain/physiopathology , Schizophrenia/genetics , Schizophrenia/physiopathology , Adult , Cluster Analysis , Female , Genetic Predisposition to Disease , Genomics , Humans , Magnetic Resonance Imaging , Male , Neural Pathways/physiopathology , Pilot Projects , Polymorphism, Single Nucleotide , Schizophrenia/diagnostic imaging
9.
Hum Brain Mapp ; 40(10): 3058-3077, 2019 07.
Article in English | MEDLINE | ID: mdl-30884018

ABSTRACT

The brain is highly dynamic, reorganizing its activity at different interacting spatial and temporal scales, including variation within and between brain networks. The chronnectome is a model of the brain in which nodal activity and connectivity patterns change in fundamental and recurring ways over time. Most literature assumes fixed spatial nodes/networks, ignoring the possibility that spatial nodes/networks may vary in time. Here, we introduce an approach to calculate a spatially fluid chronnectome (called the spatial chronnectome for clarity), which focuses on the variations of networks coupling at the voxel level, and identify a novel set of spatially dynamic features. Results reveal transient spatially fluid interactions between intra- and internetwork relationships in which brain networks transiently merge and separate, emphasizing dynamic segregation and integration. Brain networks also exhibit distinct spatial patterns with unique temporal characteristics, potentially explaining a broad spectrum of inconsistencies in previous studies that assumed static networks. Moreover, we show anticorrelative connections to brain networks are transient as opposed to constant across the entire scan. Preliminary assessments using a multi-site dataset reveal the ability of the approach to obtain new information and nuanced alterations that remain undetected during static analysis. Patients with schizophrenia (SZ) display transient decreases in voxel-wise network coupling within visual and auditory networks, and higher intradomain coupling variability. In summary, the spatial chronnectome represents a new direction of research enabling the study of functional networks which are transient at the voxel level, and the identification of mechanisms for within- and between-subject spatial variability.


Subject(s)
Brain/physiology , Connectome/methods , Models, Neurological , Neural Pathways/physiology , Schizophrenia/physiopathology , Adult , Female , Humans , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Male , Middle Aged , Young Adult
10.
Hum Brain Mapp ; 40(13): 3795-3809, 2019 09.
Article in English | MEDLINE | ID: mdl-31099151

ABSTRACT

There is growing evidence that rather than using a single brain imaging modality to study its association with physiological or symptomatic features, the field is paying more attention to fusion of multimodal information. However, most current multimodal fusion approaches that incorporate functional magnetic resonance imaging (fMRI) are restricted to second-level 3D features, rather than the original 4D fMRI data. This trade-off is that the valuable temporal information is not utilized during the fusion step. Here we are motivated to propose a novel approach called "parallel group ICA+ICA" that incorporates temporal fMRI information from group independent component analysis (GICA) into a parallel independent component analysis (ICA) framework, aiming to enable direct fusion of first-level fMRI features with other modalities (e.g., structural MRI), which thus can detect linked functional network variability and structural covariations. Simulation results show that the proposed method yields accurate intermodality linkage detection regardless of whether it is strong or weak. When applied to real data, we identified one pair of significantly associated fMRI-sMRI components that show group difference between schizophrenia and controls in both modalities, and this linkage can be replicated in an independent cohort. Finally, multiple cognitive domain scores can be predicted by the features identified in the linked component pair by our proposed method. We also show these multimodal brain features can predict multiple cognitive scores in an independent cohort. Overall, results demonstrate the ability of parallel GICA+ICA to estimate joint information from 4D and 3D data without discarding much of the available information up front, and the potential for using this approach to identify imaging biomarkers to study brain disorders.


Subject(s)
Functional Neuroimaging/methods , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Nerve Net/pathology , Nerve Net/physiopathology , Schizophrenia/pathology , Schizophrenia/physiopathology , Adult , Clinical Trials, Phase III as Topic , Computer Simulation , Female , Humans , Male , Middle Aged , Models, Theoretical , Nerve Net/diagnostic imaging , Schizophrenia/diagnostic imaging , Young Adult
11.
Hum Brain Mapp ; 40(6): 1969-1986, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30588687

ABSTRACT

The analysis of time-varying activity and connectivity patterns (i.e., the chronnectome) using resting-state magnetic resonance imaging has become an important part of ongoing neuroscience discussions. The majority of previous work has focused on variations of temporal coupling among fixed spatial nodes or transition of the dominant activity/connectivity pattern over time. Here, we introduce an approach to capture spatial dynamics within functional domains (FDs), as well as temporal dynamics within and between FDs. The approach models the brain as a hierarchical functional architecture with different levels of granularity, where lower levels have higher functional homogeneity and less dynamic behavior and higher levels have less homogeneity and more dynamic behavior. First, a high-order spatial independent component analysis is used to approximate functional units. A functional unit is a pattern of regions with very similar functional activity over time. Next, functional units are used to construct FDs. Finally, functional modules (FMs) are calculated from FDs, providing an overall view of brain dynamics. Results highlight the spatial fluidity within FDs, including a broad spectrum of changes in regional associations, from strong coupling to complete decoupling. Moreover, FMs capture the dynamic interplay between FDs. Patients with schizophrenia show transient reductions in functional activity and state connectivity across several FDs, particularly the subcortical domain. Activity and connectivity differences convey unique information in many cases (e.g., the default mode) highlighting their complementarity information. The proposed hierarchical model to capture FD spatiotemporal variations provides new insight into the macroscale chronnectome and identifies changes hidden from existing approaches.


Subject(s)
Brain/diagnostic imaging , Models, Neurological , Adolescent , Adult , Brain/physiology , Brain Mapping/methods , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Young Adult
12.
Alzheimers Dement ; 14(9): 1137-1147, 2018 09.
Article in English | MEDLINE | ID: mdl-29782824

ABSTRACT

INTRODUCTION: Although the apolipoprotein E ε4-allele (APOE-ε4) is a susceptibility factor for Alzheimer's disease (AD) and dementia with Lewy bodies (DLB), its relationship with imaging and cognitive measures across the AD/DLB spectrum remains unexplored. METHODS: We studied 298 patients (AD = 250, DLB = 48; 38 autopsy-confirmed; NCT01800214) using neuropsychological testing, volumetric magnetic resonance imaging, and APOE genotyping to investigate the association of APOE-ε4 with hippocampal volume and learning/memory phenotypes, irrespective of diagnosis. RESULTS: Across the AD/DLB spectrum: (1) hippocampal volumes were smaller with increasing APOE-ε4 dosage (no genotype × diagnosis interaction observed), (2) learning performance as assessed by total recall scores was associated with hippocampal volumes only among APOE-ε4 carriers, and (3) APOE-ε4 carriers performed worse on long-delay free word recall. DISCUSSION: These findings provide evidence that APOE-ε4 is linked to hippocampal atrophy and learning/memory phenotypes across the AD/DLB spectrum, which could be useful as biomarkers of disease progression in therapeutic trials of mixed disease.


Subject(s)
Alzheimer Disease/genetics , Apolipoprotein E4/genetics , Hippocampus/diagnostic imaging , Learning , Lewy Body Disease/genetics , Aged , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/psychology , Endophenotypes , Female , Genetic Predisposition to Disease , Heterozygote , Hippocampus/pathology , Humans , Lewy Body Disease/diagnostic imaging , Lewy Body Disease/psychology , Magnetic Resonance Imaging , Male , Neuropsychological Tests , Organ Size
13.
Int J Neuropsychopharmacol ; 20(1): 40-49, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27927736

ABSTRACT

Background: QUALIFY was a 28-week, randomized, open-label, head-to-head trial that assessed improvements across multiple measures in stable patients with schizophrenia with aripiprazole once-monthly 400 mg vs paliperidone palmitate. Methods: Secondary effectiveness assessments included physician-rated readiness for work using the Work Readiness Questionnaire, the Clinical Global Impression-Severity and Clinical Global Impression-Improvement scales, and quality of life with the rater-blinded Heinrichs-Carpenter Quality of Life Scale. Patients assessed their treatment satisfaction and quality of life with Subjective Well-Being under Neuroleptic Treatment-short version and Tolerability and Quality of Life questionnaires. Results: Odds of being ready for work at week 28 were significantly higher with aripiprazole once-monthly 400 mg vs paliperidone palmitate (adjusted odds ratio, 2.67; 95% CI, 1.39-5.14; P=.003). Aripiprazole once-monthly 400 mg produced numerically or significantly greater improvements from baseline vs paliperidone palmitate in all Quality of Life Scale items. With aripiprazole once-monthly 400 mg vs paliperidone palmitate at week 28, there were significantly more Clinical Global Impression-Severity and Clinical Global Impression-Improvement responders (adjusted odds ratio, 2.26; P=.010, and 2.51; P=.0032) and significantly better Clinical Global Impression-Improvement scores (least squares mean treatment difference, -0.326; 95% CI, -0.60 to -0.05; P=.020). Numerically larger improvements with aripiprazole once-monthly 400 mg vs paliperidone palmitate were observed for patient-rated scales Subjective Well-Being under Neuroleptic Treatment-short version and Tolerability and Quality of Life. Partial correlations were strongest among clinician-rated and among patient-rated scales but poorest between clinician and patient-rated scales. Conclusions: Consistently greater improvements were observed with aripiprazole once-monthly 400 mg vs paliperidone palmitate across all measures. Partial correlations between scales demonstrate the multidimensionality of various measures of improvement. More patients on aripiprazole once-monthly 400 mg were deemed ready to work by the study end. Trial registry: National Institutes of Health registry, NCT01795547, https://clinicaltrials.gov/ct2/results?id=NCT01795547).


Subject(s)
Antipsychotic Agents/therapeutic use , Aripiprazole/therapeutic use , Paliperidone Palmitate/therapeutic use , Schizophrenia/drug therapy , Adult , Antipsychotic Agents/adverse effects , Aripiprazole/adverse effects , Employment , Female , Humans , Male , Paliperidone Palmitate/adverse effects , Patient Satisfaction , Psychiatric Status Rating Scales , Quality of Life , Severity of Illness Index , Single-Blind Method , Surveys and Questionnaires , Treatment Outcome
14.
Hum Psychopharmacol ; 32(1)2017 01.
Article in English | MEDLINE | ID: mdl-28120489

ABSTRACT

OBJECTIVE: Tardive dyskinesia (TD) is a motor side effect that may develop after long-term antipsychotic treatment. Schizophrenia has recently been associated with the Neurexin-1 (NRXN1) gene that codes for a cell adhesion molecule in synaptic communication. METHODS: This study examined five NRXN1 single-nucleotide polymorphisms (SNPs) for possible association with the occurrence and severity of TD in 178 schizophrenia patients of European ancestry. RESULTS: We did not find these SNPs to be significantly associated with TD. CONCLUSIONS: More research is needed with additional SNPs and in bigger samples before we can completely rule out the role of NRXN1 in TD.


Subject(s)
Antipsychotic Agents/adverse effects , Cell Adhesion Molecules, Neuronal/genetics , Genetic Association Studies/methods , Nerve Tissue Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Schizophrenia/genetics , Tardive Dyskinesia/genetics , Adult , Calcium-Binding Proteins , Female , Humans , Male , Neural Cell Adhesion Molecules , Schizophrenia/drug therapy , Tardive Dyskinesia/chemically induced , White People/genetics
15.
Neuroimage ; 124(Pt B): 1155-1167, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26142271

ABSTRACT

SchizConnect (www.schizconnect.org) is built to address the issues of multiple data repositories in schizophrenia neuroimaging studies. It includes a level of mediation--translating across data sources--so that the user can place one query, e.g. for diffusion images from male individuals with schizophrenia, and find out from across participating data sources how many datasets there are, as well as downloading the imaging and related data. The current version handles the Data Usage Agreements across different studies, as well as interpreting database-specific terminologies into a common framework. New data repositories can also be mediated to bring immediate access to existing datasets. Compared with centralized, upload data sharing models, SchizConnect is a unique, virtual database with a focus on schizophrenia and related disorders that can mediate live data as information is being updated at each data source. It is our hope that SchizConnect can facilitate testing new hypotheses through aggregated datasets, promoting discovery related to the mechanisms underlying schizophrenic dysfunction.


Subject(s)
Databases, Factual , Datasets as Topic , Information Dissemination/methods , Neuroimaging , Schizophrenia/pathology , Adolescent , Adult , Aged , Child , Database Management Systems , Female , Humans , Internet , Male , Middle Aged , Terminology as Topic , User-Computer Interface , Young Adult
16.
Neuroimage ; 124(Pt B): 1074-1079, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26364863

ABSTRACT

The Function Biomedical Informatics Research Network (FBIRN) developed methods and tools for conducting multi-scanner functional magnetic resonance imaging (fMRI) studies. Method and tool development were based on two major goals: 1) to assess the major sources of variation in fMRI studies conducted across scanners, including instrumentation, acquisition protocols, challenge tasks, and analysis methods, and 2) to provide a distributed network infrastructure and an associated federated database to host and query large, multi-site, fMRI and clinical data sets. In the process of achieving these goals the FBIRN test bed generated several multi-scanner brain imaging data sets to be shared with the wider scientific community via the BIRN Data Repository (BDR). The FBIRN Phase 1 data set consists of a traveling subject study of 5 healthy subjects, each scanned on 10 different 1.5 to 4 T scanners. The FBIRN Phase 2 and Phase 3 data sets consist of subjects with schizophrenia or schizoaffective disorder along with healthy comparison subjects scanned at multiple sites. In this paper, we provide concise descriptions of FBIRN's multi-scanner brain imaging data sets and details about the BIRN Data Repository instance of the Human Imaging Database (HID) used to publicly share the data.


Subject(s)
Databases, Factual , Medical Informatics , Adolescent , Adult , Aged , Biomedical Research , Female , Healthy Volunteers , Humans , Information Dissemination , Magnetic Resonance Imaging , Male , Middle Aged , Neuroimaging , Psychotic Disorders/pathology , Reference Values , Research , Schizophrenia/pathology , Young Adult
17.
Article in English | MEDLINE | ID: mdl-26745992

ABSTRACT

BACKGROUND: The catechol-O-methyltransferase (COMT) enzyme plays a crucial role in dopamine degradation, and the COMT Val158Met polymorphism (rs4680) is associated with significant differences in enzymatic activity and consequently dopamine concentrations in the prefrontal cortex. Multiple studies have analyzed the COMT Val158Met variant in relation to antipsychotic response. Here, we conducted a meta-analysis examining the relationship between COMT Val158Met and antipsychotic response. METHODS: Searches using PubMed, Web of Science, and PsycInfo databases (03/01/2015) yielded 23 studies investigating COMT Val158Met variation and antipsychotic response in schizophrenia and schizo-affective disorder. Responders/nonresponders were defined using each study's original criteria. If no binary response definition was used, authors were asked to define response according to at least 30% Positive and Negative Syndrome Scale score reduction (or equivalent in other scales). Analysis was conducted under a fixed-effects model. RESULTS: Ten studies met inclusion criteria for the meta-analysis. Five additional antipsychotic-treated samples were analyzed for Val158Met and response and included in the meta-analysis (ntotal=1416). Met/Met individuals were significantly more likely to respond than Val-carriers (P=.039, ORMet/Met=1.37, 95% CI: 1.02-1.85). Met/Met patients also experienced significantly greater improvement in positive symptoms relative to Val-carriers (P=.030, SMD=0.24, 95% CI: 0.024-0.46). Posthoc analyses on patients treated with atypical antipsychotics (n=1207) showed that Met/Met patients were significantly more likely to respond relative to Val-carriers (P=.0098, ORMet/Met=1.54, 95% CI: 1.11-2.14), while no difference was observed for typical-antipsychotic-treated patients (n=155) (P=.65). CONCLUSIONS: Our findings suggest that the COMT Val158Met polymorphism is associated with response to antipsychotics in schizophrenia and schizo-affective disorder patients. This effect may be more pronounced for atypical antipsychotics.


Subject(s)
Antipsychotic Agents/therapeutic use , Catechol O-Methyltransferase/genetics , Pharmacogenomic Variants , Polymorphism, Genetic , Psychotic Disorders/drug therapy , Schizophrenia/drug therapy , Antipsychotic Agents/adverse effects , Humans , Odds Ratio , Pharmacogenetics , Pharmacogenomic Testing , Psychotic Disorders/enzymology , Psychotic Disorders/genetics , Psychotic Disorders/psychology , Remission Induction , Risk Factors , Schizophrenia/enzymology , Schizophrenia/genetics , Schizophrenic Psychology , Treatment Outcome
18.
CNS Spectr ; 21(2): 199-206, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25269787

ABSTRACT

OBJECTIVE/INTRODUCTION: Unemployment can negatively impact quality of life among patients with schizophrenia. Employment status depends on ability, opportunity, education, and cultural influences. A clinician-rated scale of work readiness, independent of current work status, can be a valuable assessment tool. A series of studies were conducted to create and validate a Work Readiness Questionnaire (WoRQ) for clinicians to assess patient ability to engage in socially useful activity, independent of work availability. METHODS: Content validity, test-retest and inter-rater reliability, and construct validity were evaluated in three separate studies. RESULTS: Content validity was supported. Cronbach's α was 0.91, in the excellent range. Clinicians endorsed WoRQ concepts, including treatment adherence, physical appearance, social competence, and symptom control. The final readiness decision showed good test-retest reliability and moderate inter-rater reliability. Work readiness was associated with higher function and lower levels of negative symptoms. Low positive and high negative predictive values confirmed the concept validity. DISCUSSION: The WoRQ has suitable psychometric properties for use in a clinical trial for patients with a broad range of symptom severity. The scale may be applicable to assess therapeutic interventions. It is not intended to assess eligibility for supported work interventions. CONCLUSIONS: The WoRQ is suitable for use in schizophrenia clinical trials to assess patient work functional potential.


Subject(s)
Employment/psychology , Patient Compliance , Physical Appearance, Body , Schizophrenia , Schizophrenic Psychology , Social Skills , Adult , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Psychometrics , Reproducibility of Results , Surveys and Questionnaires , Work/psychology
19.
Hum Psychopharmacol ; 31(2): 121-34, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26876050

ABSTRACT

OBJECTIVE: Approximately 30% of patients with schizophrenia fail to respond to antipsychotic therapy and are classified as having treatment-resistant schizophrenia. Clozapine is the most efficacious drug for treatment-resistant schizophrenia and may deliver superior therapeutic effects partly by modulating glutamate neurotransmission. Response to clozapine is highly variable and may depend on genetic factors as indicated by twin studies. We investigated eight polymorphisms in the N-methyl-D-aspartate glutamate receptor subunit gene GRIN2B with response to clozapine. METHODS: GRIN2B variants were genotyped using standard TaqMan procedures in 175 European patients with schizophrenia deemed resistant or intolerant to treatment. Response was assessed using change in Brief Psychiatric Rating Scale scores following six months of clozapine therapy. Categorical and continuous response was assessed using chi-squared test and analysis of covariance, respectively. RESULTS: No associations were observed between the variants and response to clozapine. A-allele carriers of rs1072388 responded marginally better to clozapine therapy than GG-homozygotes; however, the difference was not statistically significant (p = 0.067, uncorrected). CONCLUSIONS: Our findings do not support a role for these GRIN2B variants in altering response to clozapine in our sample. Investigation of additional glutamate variants in clozapine response is warranted.


Subject(s)
Antipsychotic Agents/therapeutic use , Clozapine/therapeutic use , Polymorphism, Single Nucleotide , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/drug therapy , Schizophrenia/genetics , Adult , Drug Resistance/genetics , Female , Genetic Association Studies , Haplotypes , Humans , Linkage Disequilibrium , Male , Psychiatric Status Rating Scales , Treatment Outcome , White People/genetics
20.
Proc Natl Acad Sci U S A ; 110(8): 3077-82, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23365139

ABSTRACT

Aggregation of misfolded proteins is characteristic of a number of neurodegenerative diseases, including Huntington disease (HD). The CCT/TRiC (chaperonin containing TCP-1/TCP-1 ring) chaperonin complex can inhibit aggregation and cellular toxicity induced by expanded repeat Huntingtin (mHtt) fragments. The substrate-binding apical domain of CCT/TRiC subunit CCT1, ApiCCT1, is sufficient to inhibit aggregation of expanded repeat mHtt fragments in vitro, providing therapeutic promise for HD. However, a key hurdle in considering ApiCCT1 as a potential treatment is in delivery. Because ApiCCT1 has a region of similarity to the HIV Tat protein cell-transduction domain, we tested whether recombinant ApiCCT1 (ApiCCT1(r)) protein could enter cells following exogenous delivery and modulate an established panel of mHtt-mediated cell-based phenotypes. Cell fractionation studies demonstrate that exogenous ApiCCT1(r) can penetrate cell membranes and can localize to the nucleus, consistent with a strategy that can target both cytosolic and nuclear pathogenic events in HD. ApiCCT1(r) application does indeed modulate HD cellular phenotypes by decreasing formation of visible inclusions, fibrillar oligomers, and insoluble mHtt derived from expression of a truncated mHtt exon 1 fragment. ApiCCT1(r) also delays the onset of inclusion body formation as visualized via live imaging. ApiCCT1(r) reduces mHtt-mediated toxicity in immortalized striatal cells derived from full-length knock-in HD mice, suggesting that therapeutic benefit may extend beyond effects on aggregation. These studies provide the basis for a potentially robust and unique therapeutic strategy to target mHtt-mediated protein pathogenesis.


Subject(s)
Chaperonins/administration & dosage , Mutation , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Cell Survival , Chaperonins/chemistry , Electrophoresis, Polyacrylamide Gel , Huntingtin Protein , Mice , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , PC12 Cells , Phenotype , Rats
SELECTION OF CITATIONS
SEARCH DETAIL