Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Psychol Med ; : 1-8, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38587021

ABSTRACT

BACKGROUND: Auditory system plasticity is a promising target for neuromodulation, cognitive rehabilitation and therapeutic development in schizophrenia (SZ). Auditory-based targeted cognitive training (TCT) is a 'bottom up' intervention designed to enhance the speed and accuracy of auditory information processing, which has been shown to improve neurocognition in certain SZ patients. However, the dynamics of TCT learning as a function of training exercises and their impact on neurocognitive functioning and therapeutic outcomes are unknown. METHODS: Forty subjects (SZ, n = 21; healthy subjects (HS), n = 19) underwent comprehensive clinical, cognitive, and auditory assessments, including measurements of auditory processing speed (APS) at baseline and after 1-h of TCT. SZ patients additionally completed 30-hours of TCT and repeated assessments ~10-12 weeks later. RESULTS: SZ patients were deficient in APS at baseline (d = 0.96, p < 0.005) relative to HS. After 1-h of TCT, analyses revealed significant main effects of diagnosis (d = 1.75, p = 0.002) and time (d = 1.04, p < 0.001), and a diagnosis × time interaction (d = 0.85, p = 0.02) on APS. APS learning effects were robust after 1-h in SZ patients (d = 1.47, p < 0.001) and persisted throughout the 30-h of training. Baseline APS was associated with verbal learning gains after 30-h of TCT (r = 0.51, p = 0.02) in SZ. CONCLUSIONS: TCT learning metrics may have prognostic utility and aid in the prospective identification of individuals likely to benefit from TCT. Future experimental medicine studies may advance predictive algorithms that enhance TCT-related clinical, cognitive and functional outcomes.

2.
Psychiatry Res ; 327: 115215, 2023 09.
Article in English | MEDLINE | ID: mdl-37406367

ABSTRACT

Auditory-based targeted cognitive training (ATCT) programs are emerging pro-cognitive therapeutic interventions which aim to improve auditory processing to attenuate cognitive impairment in a "bottom up" manner. Biomarkers of early auditory information processing (EAIP) like mismatch negativity (MMN) and P3a have been used successfully to predict gains from a full 40 h course of ATCT in schizophrenia (SZ). Here we investigated the ability of EAIP biomarkers to predict ATCT performance in a group of subjects (n = 26) across SZ, MDD, PTSD and GAD diagnoses. Cognition was assessed via the MATRICS Consensus Cognitive Battery (MCCB) and MMN/P3a were collected prior to completing 1 h of "Sound Sweeps," a representative ATCT exercise. Baseline and final performance over the first two levels of cognitive training served as the primary dependent variables. Groups had similar MMN, but the SZ group had attenuated P3a. MMN and MCCB cognitive domain t-scores, but not P3a, were strongly correlated with most ATCT performance measures, and explained up to 61% of variance in ATCT performance. Diagnosis was not a significant predictor for ATCT performance. These data suggest that MMN can predict ATCT performance in heterogeneous neuropsychiatric populations and should be considered in ATCT studies across diagnostically diverse cohorts.


Subject(s)
Cognitive Dysfunction , Schizophrenia , Humans , Cognitive Training , Electroencephalography , Schizophrenia/therapy , Auditory Perception , Cognitive Dysfunction/diagnosis , Evoked Potentials, Auditory , Acoustic Stimulation
4.
J Alzheimers Dis ; 91(1): 355-362, 2023.
Article in English | MEDLINE | ID: mdl-36404550

ABSTRACT

BACKGROUND: The uncompetitive NMDA antagonist, memantine (MEM), enhances prepulse inhibition of startle (PPI) across species. MEM is used to treat Alzheimer's disease (AD); conceivably, its acute impact on PPI might be used to predict a patient's sensitivity to MEM's therapeutic effects. OBJECTIVE: To begin to test this possibility, we studied MEM effects on PPI and related measures in AD patients. METHODS: 18 carefully screened individuals with AD (mean age = 72.8 y; M:F=9 : 9) completed double-blind order-balanced testing with MEM (placebo versus 20 mg), assessing acoustic startle magnitude, habituation, PPI, and latency. RESULTS: Fifteen out of 18 participants exhibited reliable startle responses. MEM did not significantly impact startle magnitude or habituation. Compared to placebo responses, PPI was significantly increased after MEM (p < 0.04; d = 0.40); this comparison reached a large effect size for the 60 ms interval (d = 0.62), where maximal MEM effects on PPI were previously detected. Prepulses reduced peak startle latency ("latency facilitation") and this effect was amplified after MEM (p = 0.03; d = 0.41; for 60 ms intervals, d = 0.69). No effects of MEM were detected on cognition, nor were MEM effects on startle associated with cognitive or clinical measures. CONCLUSION: MEM enhances prepulse effects on startle magnitude and latency in AD; these changes in PPI and latency facilitation with MEM suggest that these measures can be used to detect an AD patient's neural sensitivity to acute MEM challenge. Studies in progress will determine whether such a "biomarker" measured at the outset on treatment can predict sensitivity to MEM's therapeutic effects.


Subject(s)
Alzheimer Disease , Memantine , Aged , Humans , Acoustic Stimulation , Alzheimer Disease/drug therapy , Cognition , Memantine/pharmacology , Memantine/therapeutic use , Reflex, Startle/physiology , Male , Female , Double-Blind Method
5.
Psychopharmacology (Berl) ; 239(3): 923-933, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35132440

ABSTRACT

The bench-to-bedside development of pro-cognitive therapeutics for psychiatric disorders has been mired by translational failures. This is, in part, due to the absence of pharmacologically sensitive cognitive biomarkers common to humans and rodents. Here, we describe a cross-species translational marker of reward processing that is sensitive to the aminergic agonist, d-amphetamine. Motivated by human electroencephalographic (EEG) findings, we recently reported that frontal midline delta-band power is an electrophysiological biomarker of reward surprise in humans and in mice. In the current series of experiments, we determined the impact of parametric doses of d-amphetamine on this reward-related EEG response from humans (n = 23) and mice (n = 28) performing a probabilistic learning task. In humans, d-amphetamine (placebo, 10 mg, 20 mg) boosted the Reward Positivity event-related potential (ERP) component as well as the spectral delta-band representations of this signal. In mice, d-amphetamine (placebo, 0.1 mg/kg, 0.3 mg/kg, 1.0 mg/kg) boosted both reward and punishment ERP features, yet there was no modulation of spectral activities. In sum, the present results confirm the role of dopamine in the generation of the Reward Positivity in humans, and pave the way toward a pharmacologically valid biomarker of reward sensitivity across species.


Subject(s)
Amphetamine , Reinforcement, Psychology , Amphetamine/pharmacology , Animals , Biomarkers , Electroencephalography , Humans , Mice , Reward
6.
Psychiatry Clin Neurosci ; 76(5): 179-186, 2022 May.
Article in English | MEDLINE | ID: mdl-35037330

ABSTRACT

AIM: Information processing is supported by the cortico-cortical transmission of neural oscillations across brain regions. Recent studies have demonstrated that the rhythmic firing of neural populations is not random but is governed by interactions with other frequency bands. Specifically, the amplitude of gamma-band oscillations is associated with the phase of lower frequency oscillations in support of short and long-range communications among networks. This cross-frequency relation is thought to reflect the temporal coordination of neural communication. While schizophrenia patients show abnormal oscillatory responses across multiple frequencies at rest, it is unclear whether the functional relationships among frequency bands are intact. This study aimed to characterize the lower frequency (delta/theta, 1-8 Hz) phase and the amplitude of gamma oscillations in healthy subjects and schizophrenia patients at rest. METHODS: Low frequency-phase (delta- and theta- band) angles and gamma-band amplitude relationships were assessed in 142 schizophrenia patients and 128 healthy subjects. RESULTS: Significant low-frequency phase alteration related to high-power gamma was detected across broadly distributed scalp regions in both healthy subjects and patients. In patients, delta phase synchronization related to high-power gamma was significantly decreased at the frontocentral, right middle temporal, and left temporoparietal electrodes but significantly increased at the left parietal electrode. CONCLUSIONS: High-power gamma-related delta phase alteration may reflect a core pathophysiologic abnormality in schizophrenia. Data-driven measures of functional relationships among frequency bands may prove useful in the development of novel therapeutics. Future studies are needed to determine whether these alterations are specific to schizophrenia or appear in other neuropsychiatric patient populations.


Subject(s)
Schizophrenia , Brain , Cognition , Electroencephalography , Humans
7.
Schizophr Res ; 243: 372-382, 2022 05.
Article in English | MEDLINE | ID: mdl-34187732

ABSTRACT

Deficits in early auditory information processing contribute to cognitive and psychosocial disability; this has prompted development of interventions that target low-level auditory processing, which may alleviate these disabilities. The frequency following response (FFR) is a constellation of event-related potential and frequency characteristics that reflect the processing of acoustic stimuli at the level of the brainstem and ascending portions of the auditory pathway. While FFR is a promising candidate biomarker of response to auditory-based cognitive training interventions, the psychometric properties of FFR in schizophrenia patients have not been studied. Here we assessed the psychometric reliability and magnitude of group differences across 18 different FFR parameters to determine which of these parameters demonstrate adequate internal consistency. Electroencephalography from 40 schizophrenia patients and 40 nonpsychiatric comparison subjects was recorded during rapid presentation of an auditory speech stimulus (6000 trials). Patients showed normal response amplitudes but longer latencies for most FFR peaks and lower signal-to-noise ratios (SNRs) than healthy subjects. Analysis of amplitude and latency estimates of peaks, however, indicated a need for a substantial increase in task length to obtain internal consistency estimates above 0.80. In contrast, excellent internal consistency (>0.95) was shown for FFR sustained responses. Only SNR scores reflecting the FFR sustained response yielded significant group differences and excellent internal consistency, suggesting that this measure is a viable candidate for use in clinical treatment studies. The present study highlights the use of internal consistency estimates to select FFR characteristics for use in future intervention studies interested in individual differences among patients.


Subject(s)
Schizophrenia , Speech Perception , Acoustic Stimulation , Biomarkers , Cognition , Electroencephalography , Humans , Reproducibility of Results , Schizophrenia/therapy , Speech Perception/physiology
8.
J Alzheimers Dis ; 84(4): 1431-1438, 2021.
Article in English | MEDLINE | ID: mdl-34690144

ABSTRACT

Memantine's benefits in Alzheimer's disease (AD) are modest and heterogeneous. We tested the feasibility of using sensitivity to acute memantine challenge to predict an individual's clinical response. Eight participants completed a double-blind challenge study of memantine (placebo versus 20 mg) effects on autonomic, subjective, cognitive, and neurophysiological measures, followed by a 24-week unblinded active-dose therapeutic trial (10 mg bid). Study participation was well tolerated. Subgroups based on memantine sensitivity on specific laboratory measures differed in their clinical response to memantine, some by large effect sizes. It appears feasible to use biomarkers to predict clinical sensitivity to memantine.


Subject(s)
Alzheimer Disease/drug therapy , Biomarkers , Cognition/drug effects , Excitatory Amino Acid Antagonists/administration & dosage , Memantine/administration & dosage , Aged , Double-Blind Method , Female , Humans , Male , Neuropsychological Tests
9.
Psychiatry Res ; 305: 114239, 2021 11.
Article in English | MEDLINE | ID: mdl-34673326

ABSTRACT

Neurophysiological biomarkers of auditory processing show promise predicting outcomes following auditory-based targeted cognitive training (TCT) in schizophrenia, but the viability of the frequency following response (FFR) as a biomarker has yet to be examined, despite its ecological and face validity for auditory-based interventions. FFR is an event-related potential (ERP) that reflects early auditory processing. We predicted that schizophrenia patients would show acute- and longer-term FFR malleability in the context of TCT. Patients were randomized to either TCT (n = 30) or treatment as usual (TAU; n = 22), and electroencephalography was recorded during rapid presentation of an auditory speech stimulus before treatment, after one hour of training, and after 30 h of training. Whereas patients in the TCT group did not show changes in FFR after training, amplitude reductions were observed in the TAU. FFR was positively associated with performance on a measure of single word-in-noise perception in the TCT group, and with a measure of sentence-in-noise perception in both groups. Psychometric reliability analyses of FFR scores indicated high internal consistency but low one-hour and 12-week test-rest reliability. These findings support the dissociation between measures of speech discriminability along the hierarchy of cortical and subcortical early auditory information processing in schizophrenia.


Subject(s)
Cognition Disorders , Schizophrenia , Speech Perception , Acoustic Stimulation , Auditory Perception/physiology , Biomarkers , Cognition , Cognition Disorders/complications , Electroencephalography , Humans , Reproducibility of Results , Schizophrenia/complications , Schizophrenia/therapy , Speech Perception/physiology
10.
Schizophr Res ; 236: 135-141, 2021 10.
Article in English | MEDLINE | ID: mdl-34500174

ABSTRACT

BACKGROUND: Sensory processing abnormalities are common in schizophrenia (SZ) and impact everyday functions, such as speech perception in noisy environments. Auditory-based targeted cognitive training (TCT) is a "bottom up" cognitive remediation intervention designed to enhance the speed and accuracy of low-level auditory information processing. However, the effects of TCT on behavioral measures of central auditory processing (CAP) and the role of CAP function on verbal learning outcomes in SZ are unknown. METHODS: SZ (n = 42) and healthy subjects (CTL; n = 18) underwent comprehensive clinical, neurocognitive, and auditory assessments, including tests of hearing sensitivity and speech recognition (Words-in-Noise (WIN), Quick Speech-in-Noise (SIN)). SZ patients were randomized to receive either treatment-as-usual (TAU); or 30-h of TCT + TAU using a stratified, parallel design. SZ patients repeated assessments ~10-12 weeks later. RESULTS: Patients exhibited deficits in both WIN (p < 0.05, d = 0.50) and SIN (p < 0.01, d = 0.63). A treatment × time interaction on WIN (p < 0.05, d = 0.74), but not SIN discriminability, was seen in the TCT group relative to TAU. Specific enhancements in the 4-dB over background range drove gains in WIN performance. Moreover, SZ patients with greater CAP deficits experienced robust gains in verbal learning after 30-h of TCT relative to SZ patients without CAP impairment (p < 0.01, d = 1.28). CONCLUSION: Findings demonstrate that intensive auditory training enhances the fidelity of auditory processing and perception, such that specific CAP deficits were 'normalized' and were predictive of gains in verbal learning after TCT. It is conceivable that patients with deficiencies in CAP measures may benefit most from TCT and other interventions targeting auditory dysfunction in SZ.


Subject(s)
Cognition Disorders , Schizophrenia , Auditory Perception , Cognition , Humans , Schizophrenia/complications , Schizophrenia/therapy , Verbal Learning
11.
Am J Psychiatry ; 178(9): 838-847, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33985348

ABSTRACT

OBJECTIVE: Many psychotropic medications used to treat schizophrenia have significant anticholinergic properties, which are linked to cognitive impairment and dementia risk in healthy subjects. Clarifying the impact of cognitive impairment attributable to anticholinergic medication burden may help optimize cognitive outcomes in schizophrenia. The aim of this study was to comprehensively characterize how this burden affects functioning across multiple cognitive domains in schizophrenia outpatients. METHODS: Cross-sectional data were analyzed using inferential statistics and exploratory structural equation modeling to determine the relationship between anticholinergic medication burden and cognition. Patients with a diagnosis of schizophrenia or schizoaffective disorder (N=1,120) were recruited from the community at five U.S. universities as part of the Consortium on the Genetics of Schizophrenia-2. For each participant, prescribed medications were rated and summed according to a modified Anticholinergic Cognitive Burden (ACB) scale. Cognitive functioning was assessed by performance on domains of the Penn Computerized Neurocognitive Battery (PCNB). RESULTS: ACB score was significantly associated with cognitive performance, with higher ACB groups scoring worse than lower ACB groups on all domains tested on the PCNB. Similar effects were seen on other cognitive tests. Effects remained significant after controlling for demographic characteristics and potential proxies of illness severity, including clinical symptoms and chlorpromazine-equivalent antipsychotic dosage. CONCLUSIONS: Anticholinergic medication burden in schizophrenia is substantial, common, conferred by multiple medication classes, and associated with cognitive impairments across all cognitive domains. Anticholinergic medication burden from all medication classes-including psychotropics used in usual care-should be considered in treatment decisions and accounted for in studies of cognitive functioning in schizophrenia.


Subject(s)
Cholinergic Antagonists/adverse effects , Cognitive Dysfunction/chemically induced , Schizophrenia/drug therapy , Adolescent , Adult , Aged , Cholinergic Antagonists/therapeutic use , Cognition/drug effects , Cohort Studies , Cross-Sectional Studies , Humans , Middle Aged , Neuropsychological Tests , Schizophrenia/complications , Young Adult
12.
Schizophr Res ; 231: 73-81, 2021 05.
Article in English | MEDLINE | ID: mdl-33780847

ABSTRACT

BACKGROUND: Schizophrenia patients have abnormal electroencephalographic (EEG) power over multiple frequency bands, even at rest, though the primary neural generators and spatiotemporal dynamics of these abnormalities are largely unknown. Disturbances in the precise synchronization of oscillations within and across cortical sources may underlie abnormal resting-state EEG activity in schizophrenia patients. METHODS: A novel assessment method was applied to identify the independent contributing sources of resting-state EEG and assess the phase discontinuity in schizophrenia patients (N = 148) and healthy subjects (N = 143). RESULTS: A network of 11 primary contributing sources of scalp EEG was identified in both groups. Schizophrenia patients showed abnormal elevations of EEG power in the temporal region in the theta, beta, and gamma-bands, as well as the posterior cingulate gyrus in the delta, theta, alpha, and beta-bands. The higher theta-band power in the middle temporal gyrus was significantly correlated with verbal memory impairment in patients. The peak frequency of alpha was lower in patients in the cingulate and temporal regions. Furthermore, patients showed a higher rate of alpha phase discontinuity in the temporal region as well as a lower rate of theta phase discontinuity in the temporal and posterior cingulate regions. CONCLUSIONS: Abnormal rates of phase discontinuity of alpha- and theta-band, abnormal elevations of EEG power in multiple bands, and a lower peak frequency of alpha were identified in schizophrenia patients at rest. Clarification of the mechanistic substrates of abnormal phase discontinuity may clarify core pathophysiologic abnormalities of schizophrenia and contribute to the development of novel biomarkers for therapeutic interventions.


Subject(s)
Schizophrenia , Brain , Electroencephalography , Humans , Memory , Temporal Lobe
13.
Psychiatry Clin Neurosci ; 75(5): 172-179, 2021 May.
Article in English | MEDLINE | ID: mdl-33470494

ABSTRACT

AIM: Gamma-band auditory steady-state response (ASSR) is a neurophysiologic index that is increasingly used as a translational biomarker in the development of treatments of neuropsychiatric disorders. While gamma-band ASSR is generated by distributed networks of highly interactive temporal and frontal cortical sources, the majority of human gamma-band ASSR studies using electroencephalography (EEG) highlight activity from only a single frontocentral scalp site, Fz, where responses tend to be largest and reductions in schizophrenia patients are most evident. However, no previous study has characterized the relative source contributions to Fz, which is a necessary step to improve the concordance of preclinical and clinical EEG studies. METHODS: A novel method to back-project the contributions of independent cortical source components was applied to assess the independent sources and their proportional contributions to Fz as well as source-resolved responses in 432 schizophrenia patients and 294 healthy subjects. RESULTS: Independent contributions of gamma-band ASSR to Fz were detected from orbitofrontal, bilateral superior/middle/inferior temporal, bilateral middle frontal, and posterior cingulate gyri in both groups. In contrast to expectations, the groups showed comparable source contribution weight to gamma-band ASSR at Fz. While gamma-band ASSR reductions at Fz were present in schizophrenia patients consistent with previous studies, no group differences in individual source-level responses to Fz were detected. CONCLUSION: Small differences in multiple independent sources summate to produce scalp-level differences at Fz. The identification of independent source contributions to a single scalp sensor represents a promising methodology for measuring dissociable and homologous biomarker targets in future translational studies.


Subject(s)
Electroencephalography/methods , Evoked Potentials, Auditory/physiology , Frontal Lobe/physiology , Gamma Rhythm/physiology , Schizophrenia/physiopathology , Adult , Biomarkers , Female , Humans , Male , Middle Aged
14.
Schizophr Res ; 228: 280-287, 2021 02.
Article in English | MEDLINE | ID: mdl-33493776

ABSTRACT

BACKGROUND: Schizophrenia patients show widespread deficits in neurocognitive, clinical, and psychosocial functioning. Mismatch negativity (MMN) and gamma-band auditory steady-state response (ASSR) are robust translational biomarkers associated with schizophrenia and associated with cognitive dysfunction, negative symptom severity, and psychosocial disability. Although these biomarkers are conceptually linked as measures of early auditory information processing, it is unclear whether MMN and gamma-band ASSR account for shared vs. non-shared variance in cognitive, clinical, and psychosocial functioning. METHODS: Multiple regression analyses with MMN, gamma-band ASSR, and clinical measures were performed in large cohorts of schizophrenia outpatients (N = 428) and healthy comparison subjects (N = 283). RESULTS: Reduced MMN (d = 0.67), gamma-band ASSR (d = -0.40), and lower cognitive function were confirmed in schizophrenia patients. Regression analyses revealed that reduced MMN amplitude showed unique associations with lower verbal learning and negative symptoms, reduced gamma-band ASSR showed a unique association with working memory deficits, and both reduced MMN amplitude and reduced gamma-band ASSR showed an association with daily functioning impairment in schizophrenia patients. CONCLUSION: MMN and ASSR measures are non-redundant and complementary measures of early auditory information processing that are associated with important domains of functioning. Studies are needed to clarify the neural substrates of MMN and gamma-band ASSR to improve our understanding of the pathophysiology of schizophrenia and accelerate their use in the development of novel therapeutic interventions.


Subject(s)
Schizophrenia , Acoustic Stimulation , Auditory Perception , Cognition , Electroencephalography , Evoked Potentials, Auditory , Humans , Memory, Short-Term , Schizophrenia/complications
15.
Int J Psychophysiol ; 161: 76-85, 2021 03.
Article in English | MEDLINE | ID: mdl-33453303

ABSTRACT

BACKGROUND: Mismatch negativity (MMN) and P3a are event-related potential measures of early auditory information processing that are increasingly used as translational biomarkers in the development of treatments for neuropsychiatric disorders. These responses are reduced in schizophrenia patients over the frontocentral scalp electrodes and are associated with important domains of cognitive and psychosocial functioning. While MMN and P3a responses are generated by a dynamic network of cortical sources distributed across the temporal and frontal brain regions, it is not clear how these sources independently contribute to MMN and P3a at the primary frontocentral scalp electrode or to abnormalities observed in schizophrenia. This study aimed to determine the independent source contributions and characterize the magnitude of impairment in source-level MMN and P3a responses in schizophrenia patients. METHODS: A novel method was applied to back-project the contributions of 11 independent cortical source components to Fz, the primary scalp sensor that is used in clinical studies, in n = 589 schizophrenia patients and n = 449 healthy comparison subjects. RESULTS: The groups showed comparable individual source contributions underlying both MMN and P3a responses at Fz. Source-level responses revealed an increasing magnitude of impairment in schizophrenia patients from the temporal to more frontal sources. CONCLUSIONS: Schizophrenia patients have a normal architecture of source contributions that are accompanied by widespread abnormalities in source resolved mismatch and P3a responses, with more prominent deficits detected from the frontal sources. Quantification of source contributions and source-level responses accelerates clarification of the neural networks underlying MMN reduction at Fz in schizophrenia patients.


Subject(s)
Schizophrenia , Brain , Electroencephalography , Evoked Potentials , Evoked Potentials, Auditory , Frontal Lobe , Humans
16.
Schizophr Bull ; 47(2): 373-385, 2021 03 16.
Article in English | MEDLINE | ID: mdl-32856089

ABSTRACT

Cognitive impairment is a hallmark of schizophrenia and a robust predictor of functional outcomes. Impairments are found in all phases of the illness and are only moderately attenuated by currently approved therapeutics. Neurophysiological indices of sensory discrimination (ie, mismatch negativity (MMN) and P3a amplitudes) and gamma-band auditory steady-state response (ASSR; power and phase locking) are translational biomarkers widely used in the development of novel therapeutics for neuropsychiatric disorders. It is unclear whether laboratory-based EEG measures add explanatory power to well-established models that use only cognitive, clinical, and functional outcome measures. Moreover, it is unclear if measures of sensory discrimination and gamma-band ASSR uniquely contribute to putative causal pathways linking sensory discrimination, neurocognition, negative symptoms, and functional outcomes in schizophrenia. To answer these questions, hierarchical associations among sensory processing, neurocognition, clinical symptoms, and functional outcomes were assessed via structural equation modeling in a large sample of schizophrenia patients (n = 695) and healthy comparison subjects (n = 503). The results showed that the neurophysiologic indices of sensory discrimination and gamma-band ASSR both significantly contribute to and yield unique hierarchical, "bottom-up" effects on neurocognition, symptoms, and functioning. Measures of sensory discrimination showed direct effects on neurocognition and negative symptoms, while gamma-band ASSR had a direct effect on neurocognition in patients. Continued investigation of the neural mechanisms underlying abnormal networks of MMN/P3a and gamma-band ASSR is needed to clarify the pathophysiology of schizophrenia and the development of novel therapeutic interventions.


Subject(s)
Cognitive Dysfunction/physiopathology , Discrimination, Psychological/physiology , Evoked Potentials, Auditory/physiology , Gamma Rhythm/physiology , Schizophrenia/physiopathology , Adult , Attention , Auditory Perception , Cognitive Dysfunction/etiology , Event-Related Potentials, P300/physiology , Humans , Schizophrenia/complications
17.
Psychol Serv ; 18(4): 554-565, 2021 Nov.
Article in English | MEDLINE | ID: mdl-32658511

ABSTRACT

In clinical trials, standardized assessment conducted by research staff facilitates identification of treatment benefit. Narrative notes completed by clinicians offer a novel source to characterize and contextualize outcomes. In this study, we examine qualitative analysis of clinical notes as a method to augment quantitative outcome measures and supply meaningful context in clinical trials. Two hundred eighty-four clinical progress notes from 19 participants with schizophrenia or schizoaffective disorder assigned to receive either auditory-targeted cognitive training or treatment as usual were included. Qualitative analysis of weekly progress notes written by clinicians involved in ongoing care of the participants was used to identify overall outcome trajectories and specific changes in program participation, social functioning, and symptom severity. Trajectories were compared with the parent study's 2 primary outcome measures. Qualitative analysis identified personalized and complex trajectories for individual participants. Approximately half the participants improved overall. Most participants displayed improved program participation and social functioning, whereas most participants experienced symptom deterioration. Engagement in targeted cognitive training did not impact change in trajectories. Qualitative trajectories were congruent (e.g., both indicated improvement) with the 2 primary outcome measures for 26-36% of the participants depending on the comparison. Including qualitative analysis of clinician progress notes provides useful context and identifies underlying processes not captured in quantitative data. However, they cannot replace quantitative outcome measurement. Better alignment with clinician- and patient-targeted outcomes may strengthen clinical trials. Qualitative analysis of routinely collected data can benefit research and programmatic decision making in usual care settings. (PsycInfo Database Record (c) 2021 APA, all rights reserved).


Subject(s)
Cognition Disorders , Psychotic Disorders , Schizophrenia , Humans , Outcome Assessment, Health Care , Psychotic Disorders/therapy , Schizophrenia/therapy
18.
Front Psychiatry ; 11: 608154, 2020.
Article in English | MEDLINE | ID: mdl-33329160

ABSTRACT

Background: Patients with schizophrenia show abnormal spontaneous oscillatory activity in scalp-level electroencephalographic (EEG) responses across multiple frequency bands. While oscillations play an essential role in the transmission of information across neural networks, few studies have assessed the frequency-specific dynamics across cortical source networks at rest. Identification of the neural sources and their dynamic interactions may improve our understanding of core pathophysiologic abnormalities associated with the neuropsychiatric disorders. Methods: A novel multivector autoregressive modeling approach for assessing effective connectivity among cortical sources was developed and applied to resting-state EEG recordings obtained from n = 139 schizophrenia patients and n = 126 healthy comparison subjects. Results: Two primary abnormalities in resting-state networks were detected in schizophrenia patients. The first network involved the middle frontal and fusiform gyri and a region near the calcarine sulcus. The second network involved the cingulate gyrus and the Rolandic operculum (a region that includes the auditory cortex). Conclusions: Schizophrenia patients show widespread patterns of hyper-connectivity across a distributed network of the frontal, temporal, and occipital brain regions. Results highlight a novel approach for characterizing alterations in connectivity in the neuropsychiatric patient populations. Further mechanistic characterization of network functioning is needed to clarify the pathophysiology of neuropsychiatric and neurological diseases.

19.
Schizophr Res ; 224: 33-39, 2020 10.
Article in English | MEDLINE | ID: mdl-33189519

ABSTRACT

BACKGROUND: Latency of the acoustic startle reflex is the time from presentation of the startling stimulus until the response, and provides an index of neural processing speed. Schizophrenia subjects exhibit slowed latency compared to healthy controls. One prior publication reported significant heritability of latency. The current study was undertaken to replicate and extend this solitary finding in a larger cohort. METHODS: Schizophrenia probands, their relatives, and control subjects from the Consortium on the Genetics of Schizophrenia (COGS-1) were tested in a paradigm to ascertain magnitude, latency, and prepulse inhibition of startle. Trial types in the paradigm were: pulse-alone, and trials with 30, 60, or 120 ms between the prepulse and pulse. Comparisons of subject groups were conducted with ANCOVAs to assess startle latency and magnitude. Heritability of startle magnitude and latency was analyzed with a variance component method implemented in SOLAR v.4.3.1. RESULTS: 980 subjects had analyzable startle results: 199 schizophrenia probands, 456 of their relatives, and 325 controls. A mixed-design ANCOVA on startle latency in the four trial types was significant for subject group (F(2,973) = 4.45, p = 0.012) such that probands were slowest, relatives were intermediate and controls were fastest. Magnitude to pulse-alone trials differed significantly between groups by ANCOVA (F(2,974) = 3.92, p = 0.020) such that controls were lowest, probands highest, and relatives intermediate. Heritability was significant (p < 0.0001), with heritability of 34-41% for latency and 45-59% for magnitude. CONCLUSION: Both startle latency and magnitude are significantly heritable in the COGS-1 cohort. Startle latency is a strong candidate for being an endophenotype in schizophrenia.


Subject(s)
Schizophrenia , Acoustic Stimulation , Acoustics , Humans , Prepulse Inhibition , Reflex, Startle/genetics , Schizophrenia/genetics
20.
Transl Psychiatry ; 10(1): 405, 2020 11 23.
Article in English | MEDLINE | ID: mdl-33230190

ABSTRACT

Cognitive impairments are pervasive and disabling features of schizophrenia. Targeted cognitive training (TCT) is a "bottom-up" cognitive remediation intervention with efficacy for neurocognitive outcomes in schizophrenia, yet individual responses are variable. Gamma oscillatory measures are leading candidate biomarkers in the development of biologically informed pro-cognitive therapeutics. Forty-two schizophrenia patients were recruited from a long-term residential treatment facility. Participants were randomized to receive either 1 h of cognitive training (TCT, n = 21) or computer games (TAU, n = 21). All participants received standard-of-care treatment; the TCT group additionally completed 30 h of cognitive training. The auditory steady-state response paradigm was used to elicit gamma oscillatory power and synchrony during electroencephalogram recordings. Detailed clinical and cognitive assessments were collected at baseline and after completion of the study. Baseline gamma power predicted cognitive gains after a full course of TCT (MCCB, R2 = 0.31). A change in gamma power after 1-h TCT exposure predicted improvement in both positive (SAPS, R2 = 0.40) and negative (SANS, R2 = 0.30) symptoms. These relationships were not observed in the TAU group (MCCB, SAPS, and SANS, all R2 < 0.06). The results indicate that the capacity to support gamma oscillations, as well as the plasticity of the underlying ASSR circuitry after acute exposure to 1 h of TCT, reflect neural mechanisms underlying the efficacy of TCT, and may be used to predict individualized treatment outcomes. These findings suggest that gamma oscillatory biomarkers applied within the context of experimental medicine designs can be used to personalize individual treatment options for pro-cognitive interventions in patients with schizophrenia.


Subject(s)
Cognition Disorders , Cognitive Dysfunction , Cognitive Remediation , Schizophrenia , Cognition , Cognitive Dysfunction/therapy , Humans , Schizophrenia/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...