1
|
Chen Y, Wang S, Zhang X, Yang Q, Hua M, Li Y, Qin W, Liu F, Liang M. Functional Connectivity-Based Searchlight Multivariate Pattern Analysis for Discriminating Schizophrenia Patients and Predicting Clinical Variables. Schizophr Bull 2024; 51:108-119. [PMID: 38819252 DOI: 10.1093/schbul/sbae084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
BACKGROUND Schizophrenia, a multifaceted psychiatric disorder characterized by functional dysconnectivity, poses significant challenges in clinical practice. This study explores the potential of functional connectivity (FC)-based searchlight multivariate pattern analysis (CBS-MVPA) to discriminate between schizophrenia patients and healthy controls while also predicting clinical variables. STUDY DESIGN We enrolled 112 schizophrenia patients and 119 demographically matched healthy controls. Resting-state functional magnetic resonance imaging data were collected, and whole-brain FC subnetworks were constructed. Additionally, clinical assessments and cognitive evaluations yielded a dataset comprising 36 clinical variables. Finally, CBS-MVPA was utilized to identify subnetworks capable of effectively distinguishing between the patient and control groups and predicting clinical scores. STUDY RESULTS The CBS-MVPA approach identified 63 brain subnetworks exhibiting significantly high classification accuracies, ranging from 62.2% to 75.6%, in distinguishing individuals with schizophrenia from healthy controls. Among them, 5 specific subnetworks centered on the dorsolateral superior frontal gyrus, orbital part of inferior frontal gyrus, superior occipital gyrus, hippocampus, and parahippocampal gyrus showed predictive capabilities for clinical variables within the schizophrenia cohort. CONCLUSION This study highlights the potential of CBS-MVPA as a valuable tool for localizing the information related to schizophrenia in terms of brain network abnormalities and capturing the relationship between these abnormalities and clinical variables, and thus, deepens our understanding of the neurological mechanisms of schizophrenia.
Collapse
Affiliation(s)
- Yayuan Chen
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging and The Province and Ministry Cosponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Sijia Wang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xi Zhang
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging and The Province and Ministry Cosponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Qingqing Yang
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minghui Hua
- Department of Radiology, Chest Hospital, Tianjin University, Tianjin, China
| | - Yifan Li
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging and The Province and Ministry Cosponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Wen Qin
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Feng Liu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Meng Liang
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging and The Province and Ministry Cosponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| |
Collapse
|
2
|
Kupjetz M, Wences Chirino TY, Joisten N, Zimmer P. Kynurenine pathway dysregulation as a mechanistic link between cognitive impairment and brain damage: Implications for multiple sclerosis. Brain Res 2024:149415. [PMID: 39710050 DOI: 10.1016/j.brainres.2024.149415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/29/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Cognitive impairment is a core symptom of multiple sclerosis (MS), resulting from inflammation-related brain damage and brain network dysfunction. Inflammation also causes dysregulation of the kynurenine pathway which is the primary route of tryptophan catabolism. Kynurenine pathway dysregulation is characterised by a shift in concentrations of tryptophan catabolites, also referred to as kynurenines. Some kynurenines have neurotoxic effects that partly resemble the molecular mechanisms of MS pathophysiology underpinning brain damage and network dysfunction. The kynurenine pathway may therefore qualify as a mechanistic link between systemic inflammation, brain damage, and cognitive impairment in MS. This perspective article (1) provides an overview of inflammation-related KP dysregulation and MS-relevant neuroimmune properties of kynurenines and (2) summarises the current evidence on associations between systemic kynurenines, imaging metrics of brain structure or related markers, and cognitive performance in populations that present with kynurenine pathway dysregulation and are prone to cognitive impairment. These findings (3) are used to set a research agenda for future studies aimed at clarifying the role of the kynurenine pathway in brain damage and cognitive impairment in MS.
Collapse
Affiliation(s)
- Marie Kupjetz
- Research Group 'Sports Medicine', Institute for Sport and Sport Science, TU Dortmund University, Otto-Hahn-Str. 3, Dortmund 44227, Germany.
| | - Tiffany Y Wences Chirino
- Research Group 'Sports Medicine', Institute for Sport and Sport Science, TU Dortmund University, Otto-Hahn-Str. 3, Dortmund 44227, Germany.
| | - Niklas Joisten
- Research Group 'Sports Medicine', Institute for Sport and Sport Science, TU Dortmund University, Otto-Hahn-Str. 3, Dortmund 44227, Germany; Division of Exercise and Movement Science, Institute for Sport Science, University of Göttingen, Sprangerweg 2, Göttingen, Lower Saxony 37075, Germany.
| | - Philipp Zimmer
- Research Group 'Sports Medicine', Institute for Sport and Sport Science, TU Dortmund University, Otto-Hahn-Str. 3, Dortmund 44227, Germany.
| |
Collapse
|
3
|
Jenkins AK, Ketchesin KD, Becker-Krail DD, McClung CA. Molecular Rhythmicity in Glia: Importance for Brain Health and Relevance to Psychiatric Disease. Biol Psychiatry 2024; 96:909-918. [PMID: 38735357 PMCID: PMC11550267 DOI: 10.1016/j.biopsych.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/05/2024] [Accepted: 05/03/2024] [Indexed: 05/14/2024]
Abstract
Circadian rhythms are approximate 24-hour rhythms present in nearly all aspects of human physiology, including proper brain function. These rhythms are produced at the cellular level through a transcriptional-translational feedback loop known as the molecular clock. Diurnal variation in gene expression has been demonstrated in brain tissue from multiple species, including humans, in both cortical and subcortical regions. Interestingly, these rhythms in gene expression have been shown to be disrupted across psychiatric disorders and may be implicated in their underlying pathophysiology. However, little is known regarding molecular rhythms in specific cell types in the brain and how they might be involved in psychiatric disease. Although glial cells (e.g., astrocytes, microglia, and oligodendrocytes) have been historically understudied compared to neurons, evidence of the molecular clock is found within each of these cell subtypes. Here, we review the current literature, which suggests that molecular rhythmicity is essential to functional physiologic outputs from each glial subtype. Furthermore, disrupted molecular rhythms within these cells and the resultant functional deficits may be relevant to specific phenotypes across psychiatric illnesses. Given that circadian rhythm disruptions have been so integrally tied to psychiatric disease, the molecular mechanisms governing these associations could represent exciting new avenues for future research and potential novel pharmacologic targets for treatment.
Collapse
Affiliation(s)
- Aaron K Jenkins
- Translational Neuroscience Program, Department of Psychiatry, and Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kyle D Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, and Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Darius D Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, and Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Colleen A McClung
- Translational Neuroscience Program, Department of Psychiatry, and Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
4
|
Huang AS, Wimmer RD, Lam NH, Wang BA, Suresh S, Roeske MJ, Pleger B, Halassa MM, Woodward ND. A prefrontal thalamocortical readout for conflict-related executive dysfunction in schizophrenia. Cell Rep Med 2024; 5:101802. [PMID: 39515319 PMCID: PMC11604477 DOI: 10.1016/j.xcrm.2024.101802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/27/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
Executive dysfunction is a prominent feature of schizophrenia and may drive core symptoms. Dorsolateral prefrontal cortex (dlPFC) deficits have been linked to schizophrenia executive dysfunction, but mechanistic details critical for treatment development remain unclear. Here, capitalizing on recent animal circuit studies, we develop a task predicted to engage human dlPFC and its interactions with the mediodorsal thalamus (MD). We find that individuals with schizophrenia exhibit selective performance deficits when attention is guided by conflicting cues. Task performance correlates with lateralized MD-dlPFC functional connectivity, identifying a neural readout that predicts susceptibility to conflict during working memory in a larger independent schizophrenia cohort. In healthy subjects performing a probabilistic reversal task, this MD-dlPFC network predicts switching behavior. Overall, our three independent experiments introduce putative biomarkers for executive function in schizophrenia and highlight animal circuit studies as inspiration for the development of clinically relevant readouts.
Collapse
Affiliation(s)
- Anna S Huang
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ralf D Wimmer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Norman H Lam
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Bin A Wang
- Department of Neurology, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Collaborative Research Centre 874 "Integration and Representation of Sensory Processes", Ruhr-University Bochum, Bochum, Germany
| | - Sahil Suresh
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Maxwell J Roeske
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Burkhard Pleger
- Department of Neurology, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Collaborative Research Centre 874 "Integration and Representation of Sensory Processes", Ruhr-University Bochum, Bochum, Germany
| | - Michael M Halassa
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA; Department of Psychiatry, Tufts University School of Medicine, Boston, MA, USA.
| | - Neil D Woodward
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
5
|
Okuda T, Kimoto S, Kawabata R, Bian Y, Tsubomoto M, Okamura K, Enwright JF, Kikuchi M, Lewis DA, Hashimoto T. Alterations in inhibitory neuron subtype-selective transcripts in the prefrontal cortex: comparisons across schizophrenia and mood disorders. Psychol Med 2024; 54:1-10. [PMID: 39478366 PMCID: PMC11578916 DOI: 10.1017/s0033291724002344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/19/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND In schizophrenia (SZ), impairments in cognitive functions, such as working memory, have been associated with alterations in certain types of inhibitory neurons that utilize the neurotransmitter γ-aminobutyric acid (GABA) in the dorsolateral prefrontal cortex (DLPFC). For example, GABA neurons that express parvalbumin (PV) or somatostatin (SST) have more prominent gene expression alterations than those that express vasoactive intestinal peptide (VIP). In bipolar disorder (BD) and major depression (MD), which exhibit similar, but less severe, cognitive impairments than SZ, alterations of transcript levels in GABA neurons have also been reported. However, the extent to which GABA neuron subtype-selective transcripts in the DLPFC are affected, and the relative magnitudes of the diagnosis-associated effects, have not been directly compared across SZ, BD, and MD in the same study. METHODS We used quantitative polymerase chain reaction to examine levels of GABA neuron subtype-selective transcripts (PV, potassium voltage-gated channel modifier subfamily-S member-3, SST, VIP, and calretinin mRNAs), as well as the pan-GABA neuron marker 67 kDa glutamate decarboxylase mRNA, in DLPFC total gray matter of 160 individuals, including those with SZ, BD, or MD and unaffected comparison (UC) individuals. RESULTS Relative to UC individuals, individuals with SZ exhibited large deficits in levels of all transcripts except for calretinin mRNA, whereas individuals with BD or MD showed a marked deficit only for PV or SST mRNAs, respectively. CONCLUSIONS These findings suggest that broader and more severe alterations in DLPFC GABA neurons might contribute to the greater cognitive impairments in SZ relative to BD and MD.
Collapse
Affiliation(s)
- Takeshi Okuda
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Sohei Kimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, 634-8521, Japan
- Department of Neuropsychiatry, Wakayama Medical University School of Medicine, Wakayama, 641-8509, Japan
| | - Rika Kawabata
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Yufan Bian
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Makoto Tsubomoto
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Kazuya Okamura
- Department of Neuropsychiatry, Wakayama Medical University School of Medicine, Wakayama, 641-8509, Japan
| | - John F. Enwright
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Mitsuru Kikuchi
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
- Research Center for Child Development, Kanazawa University, Kanazawa 920-8640, Japan
| | - David A. Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Takanori Hashimoto
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Psychiatry, National Hospital Organization Hokuriku Hospital, Nanto, 939-1893, Japan
| |
Collapse
|
6
|
Haker R, Helft C, Natali Shamir E, Shahar M, Solomon H, Omer N, Blumenfeld-Katzir T, Zlotzover S, Piontkewitz Y, Weiner I, Ben-Eliezer N. Characterization of Brain Abnormalities in Lactational Neurodevelopmental Poly I:C Rat Model of Schizophrenia and Depression Using Machine-Learning and Quantitative MRI. J Magn Reson Imaging 2024. [PMID: 39466009 DOI: 10.1002/jmri.29634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND A recent neurodevelopmental rat model, utilizing lactational exposure to polyriboinosinic-polyribocytidilic acid (Poly I:C) leads to mimics of behavioral phenotypes resembling schizophrenia-like symptoms in male offspring and depression-like symptoms in female offspring. PURPOSE To identify mechanisms of neuronal abnormalities in lactational Poly I:C offspring using quantitative MRI (qMRI) tools. STUDY TYPE Prospective. ANIMAL MODEL Twenty Poly I:C rats and 20 healthy control rats, age 130 postnatal day. FIELD STRENGTH/SEQUENCE 7 T. Multiflip-angle FLASH protocol for T1 mapping; multi-echo spin-echo T2-mapping protocol; echo planar imaging protocol for diffusion tensor imaging. ASSESSMENT Nursing dams were injected with the viral mimic Poly I:C or saline (control group). In adulthood, quantitative maps of T1, T2, proton density, and five diffusion metrics were generated for the offsprings. Seven regions of interest (ROIs) were segmented, followed by extracting 10 quantitative features for each ROI. STATISTICAL TESTS Random forest machine learning (ML) tool was employed to identify MRI markers of disease and classify Poly I:C rats from healthy controls based on quantitative features. RESULTS Poly I:C rats were identified from controls with an accuracy of 82.5 ± 25.9% for females and 85.0 ± 24.0% for males. Poly I:C females exhibited differences mainly in diffusion-derived parameters in the thalamus and the medial prefrontal cortex (MPFC), while males displayed changes primarily in diffusion-derived parameters in the corpus callosum and MPFC. DATA CONCLUSION qMRI shows potential for identifying sex-specific brain abnormalities in the Poly I:C model of neurodevelopmental disorders. LEVEL OF EVIDENCE NA TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Rona Haker
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Coral Helft
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | | | - Moni Shahar
- The AI and Data Science Center, Tel Aviv University, Tel Aviv, Israel
| | - Hadas Solomon
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Noam Omer
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | | | - Sharon Zlotzover
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Yael Piontkewitz
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ina Weiner
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Noam Ben-Eliezer
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- Center for Advanced Imaging Innovation and Research (CAI2R), New York University School of Medicine, New York, New York, USA
| |
Collapse
|
7
|
Zhu J, Zhou L, Zhou Y, Lin Y, Cai Y, Wu J, Shi C. Diagnosis of schizophrenia by integrated saccade scores and associations with psychiatric symptoms, and functioning. Medicine (Baltimore) 2024; 103:e39935. [PMID: 39465854 PMCID: PMC11479490 DOI: 10.1097/md.0000000000039935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 09/13/2024] [Indexed: 10/29/2024] Open
Abstract
Eye movement as a neurobiological biomarker of schizophrenia. We aim to estimate diagnostic accuracy of integrated pro/antisaccade eye movement measurements to discriminate between healthy individuals and schizophrenic patients. We compared the eye movement performance of 85 healthy individuals and 116 schizophrenia-stable patients during prosaccade and antisaccade tasks. The difference eye movement measurements were accumulated by stepwise discriminant analysis to produce an integrated score. Finally, the diagnostic value of the integrated score was calculated by the receiver operating characteristic (ROC) area under the curve (AUC), and the best sensitivity and specificity were calculated based on the given cutoff values. Using discriminant analysis, an integrated score included the residual gain and latency (step) during the prosaccade test, the error rate, and the corrected error rate during the antisaccade test. We found that the integrated score could well classify schizophrenia patients and healthy individuals with an accuracy of 80.6%. In the ROC, Youden's index was 0.634 (sensitivity = 81.0%, specificity = 82.4%) and AUC was 0.871. There were significant difference patterns of correlation between the severity of psychiatric symptoms and daily functioning and diagnostic eye movement measurements. Using only 2 saccade tasks to discriminate well between schizophrenia patients and healthy controls, suggesting that abnormalities in saccade behavior is a potential biomarker and efficient diagnostic tool for identifying schizophrenia. The underlying neuropathologic mechanisms associated with abnormal saccades may provide insights into the intervention and diagnosis of schizophrenia.
Collapse
Affiliation(s)
- Jiahui Zhu
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Peking University, Beijing, China
| | - Li Zhou
- School of Education, Xinjiang Normal University, Xinjiang, China
| | - Yuanyuan Zhou
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Peking University, Beijing, China
| | - Yunhan Lin
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Peking University, Beijing, China
| | - Yumei Cai
- Peking University Institute of Population Research, Beijing, China
| | - Jiayuan Wu
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chuan Shi
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Peking University, Beijing, China
| |
Collapse
|
8
|
Mahapatra S, Mangot A, Tamboli A. A Comparative Study of Frontal and Cerebellar Lobe Volumes Between Patients With First-Episode Schizophrenia and Healthy Controls and Its Association With Psychopathology and Neurological Soft Signs in Patients. Cureus 2024; 16:e71389. [PMID: 39539907 PMCID: PMC11557444 DOI: 10.7759/cureus.71389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024] Open
Abstract
Objectives Our study aimed to investigate the fronto-cerebellar volumes in both patients and controls, as well as explore their relationship with symptomatology. Our primary objectives were to compare the frontal and cerebellar lobe volumetric measurements between patients with first-episode schizophrenia (FES) and healthy controls and to assess the relationship of these volumes with psychopathology, cognition, and neurological soft signs in FES patients. The secondary objective was to explore the association of fronto-cerebellar lobe volumes with socio-demographic factors among patients and controls, as well as the duration of untreated illness (DUI) among patients. Materials and methods This was a cross-sectional, case-control study involving 60 participants, including 30 antipsychotic-naïve FES patients and 30 healthy controls. Participants underwent MRI scanning to measure frontal and cerebellar lobe volumes using the volBrain platform. Additionally, FES patients were assessed using the Positive and Negative Syndrome Scale (PANSS), Montreal Cognitive Assessment-Basic (MoCA-B), and Brief Motor Scale (BMS). Pearson's correlation, independent sample t-tests, multivariate linear regression, and binomial logistic regression were used to analyze the relationships between brain volumes, clinical assessments, and socio-demographic factors. Results No significant differences in frontal volumes were found between the two groups, while cerebellar volumes were significantly smaller in FES patients (p=0.004), particularly in younger males (p=0.026). Frontal volumes were negatively correlated with age in both groups (p=0.012), which remained robust in patients even after controlling for their gender, education, and DUI (p=0.012, aR2=0.221). Cerebellar volume reduction was associated with a higher likelihood of being classified as a patient (p=0.029). BMS was significantly correlated with frontal lobe volumes, especially in motor sequencing (MoSe), after adjusting for age, gender, and education (p=0.009). BMS MoSe scores were also significantly positively correlated with the DUI (r=0.415, p=0.023) and PANSS-General Psychopathology (GP) (r=0.494, p=0.005). MoCA-B scores were significantly lower in females than males (p=0.016), while PANSS-GP was significantly negatively correlated with age (r=-0.432, p=0.017). Conclusion Frontal and cerebellar volumes were differentially impacted in FES, with cerebellar atrophy being a significant distinguishing feature of the disorder. Frontal atrophy was associated with motor dysfunction but did not appear to influence psychopathology or cognition significantly in the early stages. The independent effects of frontal and cerebellar volumes, as shown by the lack of correlation between them, may suggest that these brain regions undergo separate pathological processes in schizophrenia, with frontal functional changes impacting motor function and cerebellar structural changes contributing to broader psychiatric symptoms, thereby warranting further exploration involving larger sample size.
Collapse
Affiliation(s)
- Siddhant Mahapatra
- Psychiatry, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Ajish Mangot
- Psychiatry, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Asif Tamboli
- Radiodiagnosis, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| |
Collapse
|
9
|
Uliana DL, Lisboa JRF, Gomes FV, Grace AA. The excitatory-inhibitory balance as a target for the development of novel drugs to treat schizophrenia. Biochem Pharmacol 2024; 228:116298. [PMID: 38782077 PMCID: PMC11410545 DOI: 10.1016/j.bcp.2024.116298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
The intricate balance between excitation and inhibition (E/I) in the brain plays a crucial role in normative information processing. Dysfunctions in the E/I balance have been implicated in various psychiatric disorders, including schizophrenia (SCZ). In particular, abnormalities in GABAergic signaling, specifically in parvalbumin (PV)-containing interneurons, have been consistently observed in SCZ pathophysiology. PV interneuron function is vital for maintaining an ideal E/I balance, and alterations in PV interneuron-mediated inhibition contribute to circuit deficits observed in SCZ, including hippocampus hyperactivity and midbrain dopamine system overdrive. While current antipsychotic medications primarily target D2 dopamine receptors and are effective primarily in treating positive symptoms, novel therapeutic strategies aiming to restore the E/I balance could potentially mitigate not only positive symptoms but also negative symptoms and cognitive deficits. This could involve, for instance, increasing the inhibitory drive onto excitatory neurons or decreasing the putative enhanced pyramidal neuron activity due to functional loss of PV interneurons. Compounds targeting the glycine site at glutamate NMDA receptors and muscarinic acetylcholine receptors on PV interneurons that can increase PV interneuron drive, as well as drugs that increase the postsynaptic action of GABA, such as positive allosteric modulators of α5-GABA-A receptors, and decrease glutamatergic output, such as mGluR2/3 agonists, represent promising approaches. Preventive strategies aiming at E/I balance also represent a path to reduce the risk of transitioning to SCZ in high-risk individuals. Therefore, compounds with novel mechanisms targeting E/I balance provide optimism for more effective and tailored interventions in the management of SCZ.
Collapse
Affiliation(s)
- Daniela L Uliana
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joao Roberto F Lisboa
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
Tan XW, Gulwant Singh HK, Koh JZJ, Tan RSY, Tor PC. Personalised transcranial magnetic stimulation for treatment-resistant depression, depression with comorbid anxiety and negative symptoms of schizophrenia: a narrative review. Singapore Med J 2024; 65:544-551. [PMID: 39379030 PMCID: PMC11575723 DOI: 10.4103/singaporemedj.smj-2024-133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/27/2024] [Indexed: 10/10/2024]
Abstract
ABSTRACT Transcranial magnetic stimulation (TMS) is a promising intervention for treatment-resistant psychiatric disorders. However, conventional TMS typically utilises a one-size-fits-all approach when determining stimulation targets. Recent retrospective brain circuit-based analyses using lesion network mapping have suggested that a left dorsal lateral prefrontal cortex target has a higher efficacy for alleviating depression symptoms, a dorsomedial prefrontal cortex target is more effective for anxiety symptoms, and a rostromedial prefrontal cortex target is effective for schizophrenia-associated psychiatric symptoms. Nonetheless, symptom-specific brain circuit targeting has not been tested prospectively. We conducted a narrative review of selected literature to investigate individualised targeting for TMS and discuss potential future directions to elucidate the efficacy of this approach.
Collapse
Affiliation(s)
- Xiao Wei Tan
- Department of Mood and Anxiety, Institute of Mental Health, Singapore
| | | | | | | | | |
Collapse
|
11
|
Zheng WL, Wu Z, Hummos A, Yang GR, Halassa MM. Rapid context inference in a thalamocortical model using recurrent neural networks. Nat Commun 2024; 15:8275. [PMID: 39333467 PMCID: PMC11436643 DOI: 10.1038/s41467-024-52289-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 08/29/2024] [Indexed: 09/29/2024] Open
Abstract
Cognitive flexibility is a fundamental ability that enables humans and animals to exhibit appropriate behaviors in various contexts. The thalamocortical interactions between the prefrontal cortex (PFC) and the mediodorsal thalamus (MD) have been identified as crucial for inferring temporal context, a critical component of cognitive flexibility. However, the neural mechanism responsible for context inference remains unknown. To address this issue, we propose a PFC-MD neural circuit model that utilizes a Hebbian plasticity rule to support rapid, online context inference. Specifically, the model MD thalamus can infer temporal contexts from prefrontal inputs within a few trials. This is achieved through the use of PFC-to-MD synaptic plasticity with pre-synaptic traces and adaptive thresholding, along with winner-take-all normalization in the MD. Furthermore, our model thalamus gates context-irrelevant neurons in the PFC, thus facilitating continual learning. We evaluate our model performance by having it sequentially learn various cognitive tasks. Incorporating an MD-like component alleviates catastrophic forgetting of previously learned contexts and demonstrates the transfer of knowledge to future contexts. Our work provides insight into how biological properties of thalamocortical circuits can be leveraged to achieve rapid context inference and continual learning.
Collapse
Affiliation(s)
- Wei-Long Zheng
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, Shanghai, China.
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China.
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Zhongxuan Wu
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | - Ali Hummos
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guangyu Robert Yang
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Altera.AL, Inc., Menlo Park, CA, USA
| | - Michael M Halassa
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA.
- Department of Psychiatry, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
12
|
Guo J, He C, Song H, Gao H, Yao S, Dong SS, Yang TL. Unveiling Promising Neuroimaging Biomarkers for Schizophrenia Through Clinical and Genetic Perspectives. Neurosci Bull 2024; 40:1333-1352. [PMID: 38703276 PMCID: PMC11365900 DOI: 10.1007/s12264-024-01214-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 01/08/2024] [Indexed: 05/06/2024] Open
Abstract
Schizophrenia is a complex and serious brain disorder. Neuroscientists have become increasingly interested in using magnetic resonance-based brain imaging-derived phenotypes (IDPs) to investigate the etiology of psychiatric disorders. IDPs capture valuable clinical advantages and hold biological significance in identifying brain abnormalities. In this review, we aim to discuss current and prospective approaches to identify potential biomarkers for schizophrenia using clinical multimodal neuroimaging and imaging genetics. We first described IDPs through their phenotypic classification and neuroimaging genomics. Secondly, we discussed the applications of multimodal neuroimaging by clinical evidence in observational studies and randomized controlled trials. Thirdly, considering the genetic evidence of IDPs, we discussed how can utilize neuroimaging data as an intermediate phenotype to make association inferences by polygenic risk scores and Mendelian randomization. Finally, we discussed machine learning as an optimum approach for validating biomarkers. Together, future research efforts focused on neuroimaging biomarkers aim to enhance our understanding of schizophrenia.
Collapse
Affiliation(s)
- Jing Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Changyi He
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Huimiao Song
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Huiwu Gao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Shi Yao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Shan-Shan Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Tie-Lin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
13
|
Yoshikawa A, Obata Y, Kakiuchi C, Nakanishi A, Kimura S, Aoki S, Kato T. Case series of patients with early psychosis presenting hypoperfusion in angular gyrus and self-disturbance: Implication for the sense of agency and schizophrenia. Neuropsychopharmacol Rep 2024. [PMID: 39212170 DOI: 10.1002/npr2.12476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Self-disturbance has been considered as a core symptomatology of schizophrenia and its emergence from the prodromal phase makes it a crucial target for early detection and intervention in schizophrenia. Currently, the clinical assessment of self-disturbance relies on the self-report of patients, and clinicians have no diagnostic tools in clinical practice. Identifying the neural substrate of self-disturbance would be of great clinical value by shedding light on the core dimension of schizophrenia. CASE PRESENTATION We first introduce an adolescent patient who initially presented self-disturbance, and clinically detectable hypoperfusion in angular gyrus (AG) was observed when early psychosis was suspected. Interestingly, the hypoperfusion in AG may correspond to improvement and exacerbation of self-disturbance. This clinical observation led us to pursue the relationship between the decreased blood flow in the AG and self-disturbance. Among 15 cases with suspected early psychosis in which single photon emission computed tomography was performed to exclude organic factors, we found additional 5 cases, including one prodromal patient, showing hypoperfusion in the AG and self-disturbance with significant correlation (r = 0.79, p = 0.00025). DISCUSSION The self-disturbance has been interpreted as a reflection of disturbance of the "Sense of Agency", the ability to attribute their action and/or thoughts to themselves. AG has been shown to play a pivotal role in the sense of agency. These cases suggest that the hypoperfusion in AG associated with the disruption in the sense of agency would be an early clinical sign of schizophrenia. Further longitudinal studies are needed to test this hypothesis.
Collapse
Affiliation(s)
- Akane Yoshikawa
- Department of Psychiatry, Juntendo University School of Medicine, Bunkyō, Japan
| | - Youhei Obata
- Department of Psychiatry, Juntendo University School of Medicine, Bunkyō, Japan
| | - Chihiro Kakiuchi
- Department of Psychiatry, Juntendo University School of Medicine, Bunkyō, Japan
| | - Atsushi Nakanishi
- Department of Radiology, Juntendo University School of Medicine, Bunkyō, Japan
| | - Satoshi Kimura
- Department of Radiological Technology, Juntendo University Hospital, Bunkyō, Japan
| | - Shigeki Aoki
- Department of Radiology, Juntendo University School of Medicine, Bunkyō, Japan
| | - Tadafumi Kato
- Department of Psychiatry, Juntendo University School of Medicine, Bunkyō, Japan
| |
Collapse
|
14
|
O’Brien JT, Jalilvand SP, Suji NA, Jupelly RK, Phensy A, Mwirigi JM, Elahi H, Price TJ, Kroener S. Elevations in the Mitochondrial Matrix Protein Cyclophilin D Correlate With Reduced Parvalbumin Expression in the Prefrontal Cortex of Patients With Schizophrenia. Schizophr Bull 2024; 50:1197-1207. [PMID: 38412332 PMCID: PMC11349014 DOI: 10.1093/schbul/sbae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
BACKGROUND AND HYPOTHESIS Cognitive deficits in schizophrenia are linked to dysfunctions of the dorsolateral prefrontal cortex (DLPFC), including alterations in parvalbumin (PV)-expressing interneurons (PVIs). Redox dysregulation and oxidative stress may represent convergence points in the pathology of schizophrenia, causing dysfunction of GABAergic interneurons and loss of PV. Here, we show that the mitochondrial matrix protein cyclophilin D (CypD), a critical initiator of the mitochondrial permeability transition pore (mPTP) and modulator of the intracellular redox state, is altered in PVIs in schizophrenia. STUDY DESIGN Western blotting was used to measure CypD protein levels in postmortem DLPFC specimens of schizophrenic patients (n = 27) and matched comparison subjects with no known history of psychiatric or neurological disorders (n = 26). In a subset of this cohort, multilabel immunofluorescent confocal microscopy with unbiased stereological sampling methods were used to quantify (1) numbers of PVI across the cortical mantle (20 unaffected comparison, 14 schizophrenia) and (2) PV and CypD protein levels from PVIs in the cortical layers 2-4 (23 unaffected comparison, 18 schizophrenia). STUDY RESULTS In schizophrenic patients, the overall number of PVIs in the DLPFC was not significantly altered, but in individual PVIs of layers 2-4 PV protein levels decreased along a superficial-to-deep gradient when compared to unaffected comparison subjects. These laminar-specific PVI alterations were reciprocally linked to significant CypD elevations both in PVIs and total DLPFC gray matter. CONCLUSIONS Our findings support previously reported PVI anomalies in schizophrenia and suggest that CypD-mediated mPTP formation could be a potential contributor to PVI dysfunction in schizophrenia.
Collapse
Affiliation(s)
- John T O’Brien
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Sophia P Jalilvand
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Neha A Suji
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Rohan K Jupelly
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Aarron Phensy
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Juliet M Mwirigi
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Hajira Elahi
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Sven Kroener
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
15
|
Burns JN, Jenkins AK, Yin R, Zong W, Vadnie CA, DePoy LM, Petersen KA, Tsyglakova M, Scott MR, Tseng GC, Huang YH, McClung CA. Molecular and cellular rhythms in excitatory and inhibitory neurons in the mouse prefrontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.05.601880. [PMID: 39005410 PMCID: PMC11245095 DOI: 10.1101/2024.07.05.601880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Previous studies have shown that there are rhythms in gene expression in the mouse prefrontal cortex (PFC); however, the contribution of different cell types and potential variation by sex has not yet been determined. Of particular interest are excitatory pyramidal cells and inhibitory parvalbumin (PV) interneurons, as interactions between these cell types are essential for regulating the excitation/inhibition balance and controlling many of the cognitive functions regulated by the PFC. In this study, we identify cell-type specific rhythms in the translatome of PV and pyramidal cells in the mouse PFC and assess diurnal rhythms in PV cell electrophysiological properties. We find that while core molecular clock genes are conserved and synchronized between cell types, pyramidal cells have nearly twice as many rhythmic transcripts as PV cells (35% vs. 18%). Rhythmic transcripts in pyramidal cells also show a high degree of overlap between sexes, both in terms of which transcripts are rhythmic and in the biological processes associated with them. Conversely, in PV cells, rhythmic transcripts from males and females are largely distinct. Moreover, we find sex-specific effects of phase on action potential properties in PV cells that are eliminated by environmental circadian disruption. Together, this study demonstrates that rhythms in gene expression and electrophysiological properties in the mouse PFC vary by both cell type and sex. Moreover, the biological processes associated with these rhythmic transcripts may provide insight into the unique functions of rhythms in these cells, as well as their selective vulnerabilities to circadian disruption.
Collapse
Affiliation(s)
- Jennifer N. Burns
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Aaron K. Jenkins
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - RuoFei Yin
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261
| | - Wei Zong
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261
| | - Chelsea A. Vadnie
- David O. Robbins Neuroscience Program, Department of Psychology, Ohio Wesleyan University, Delaware, OH 43015
| | - Lauren M. DePoy
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Kaitlyn A Petersen
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Mariya Tsyglakova
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Madeline R. Scott
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - George C. Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261
| | - Yanhua H. Huang
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Colleen A. McClung
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| |
Collapse
|
16
|
Skiba RM, Chinchani AM, Menon M, Lepage M, Lavigne KM, Malla A, Joober R, Goldberg JO, Heinrichs RW, Castle DJ, Burns A, Best MW, Rossell SL, Walther S, Woodward TS. Overlap between individual differences in cognition and symptoms of schizophrenia. Schizophr Res 2024; 270:220-228. [PMID: 38924940 DOI: 10.1016/j.schres.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Neurocognitive impairment is a core feature of schizophrenia spectrum disorders (SSDs), and the relationship between cognition and symptoms in SSDs has been widely researched. Negative symptoms are related to a wide range of cognitive impairments; however, the aspects of negative symptoms that underpin this relationship have yet to be specified. STUDY DESIGN We used iterative Constrained Principal Component Analysis (iCPCA) to explore the relationship between 18 cognitive measures (including processing speed, attention, working, spatial and verbal memory and executive functions) and 46 symptoms in schizophrenia at the individual item level while minimizing the risk of Type I errors. ICPCA was conducted on a sample of SSD patients in the early stages of psychiatric treatment (n = 121) to determine the components of cognition overlapping with symptoms measured by the Scale for the Assessment of Negative Symptoms (SANS) and the Scale for the Assessment of Positive Symptoms (SAPS). RESULTS We found that a verbal memory component was associated with items from SANS and SAPS related to impoverished and disorganized emotional communication, language, and thought. In contrast, a working memory component was associated with SANS items related to motor system impoverishment. CONCLUSIONS The iCPCA allowed us to explore the associations between individual items, optimized to understand the overlap between symptoms and cognition. The specific symptoms linked to verbal and working memory impairments imply distinct brain networks, which further investigation may lead to our deeper understanding of the illness and the development of treatment methods.
Collapse
Affiliation(s)
- Rafal M Skiba
- BC Mental Health and Addictions Research Institute, Vancouver, BC, Canada; Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Abhijit M Chinchani
- BC Mental Health and Addictions Research Institute, Vancouver, BC, Canada; Department of Bioinformatics, University of British Columbia, Vancouver, Canada
| | - Mahesh Menon
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Martin Lepage
- Douglas Mental Health University Institute, McGill University, Montreal, Qc, Canada; Department of Psychiatry, McGill University, Montreal, Qc, Canada
| | - Katie M Lavigne
- Douglas Mental Health University Institute, McGill University, Montreal, Qc, Canada; Department of Psychiatry, McGill University, Montreal, Qc, Canada
| | - Ashok Malla
- Douglas Mental Health University Institute, McGill University, Montreal, Qc, Canada; Department of Psychiatry, McGill University, Montreal, Qc, Canada
| | - Ridha Joober
- Douglas Mental Health University Institute, McGill University, Montreal, Qc, Canada; Department of Psychiatry, McGill University, Montreal, Qc, Canada
| | - Joel O Goldberg
- Department of Psychology, York University, Toronto, ON, Canada
| | | | - David J Castle
- Department of Psychiatry, University of Tasmania, Hobart, TAS, Australia
| | - Amy Burns
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Michael W Best
- Department of Psychological Clinical Science, University of Toronto, Toronto, ON, Canada
| | - Susan L Rossell
- Centre for Mental Health, Swinburne University of Technology, Melbourne, VIC, Australia; Department of Mental Health, St Vincent's Hospital, Melbourne, VIC, Australia
| | - Sebastian Walther
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Switzerland
| | - Todd S Woodward
- BC Mental Health and Addictions Research Institute, Vancouver, BC, Canada; Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
17
|
Donati FL, Mayeli A, Nascimento Couto BA, Sharma K, Janssen S, Krafty RJ, Casali AG, Ferrarelli F. Prefrontal Oscillatory Slowing in Early-Course Schizophrenia Is Associated With Worse Cognitive Performance and Negative Symptoms: A Transcranial Magnetic Stimulation-Electroencephalography Study. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024:S2451-9022(24)00201-5. [PMID: 39059465 DOI: 10.1016/j.bpsc.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/02/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Abnormalities in dorsolateral prefrontal cortex (DLPFC) oscillations are neurophysiological signatures of schizophrenia thought to underlie its cognitive deficits. Transcranial magnetic stimulation with electroencephalography (TMS-EEG) provides a measure of cortical oscillations unaffected by sensory relay functionality and/or patients' level of engagement, which are important confounding factors in schizophrenia. Previous TMS-EEG work showed reduced fast, gamma-range oscillations and a slowing of the main DLPFC oscillatory frequency, or natural frequency, in chronic schizophrenia. However, it is unclear whether this DLPFC natural frequency slowing is present in early-course schizophrenia (EC-SCZ) and is associated with symptom severity and cognitive dysfunction. METHODS We applied TMS-EEG to the left DLPFC in 30 individuals with EC-SCZ and 28 healthy control participants. Goal-directed working memory performance was assessed using the AX-Continuous Performance Task. The EEG frequency with the highest cumulative power at the stimulation site, or natural frequency, was extracted. We also calculated the local relative spectral power as the average power in each frequency band divided by the broadband power. RESULTS Compared with the healthy control group, the EC-SCZ group had reduced DLPFC natural frequency (p = .0000002, Cohen's d = -2.32) and higher DLPFC beta-range relative spectral power (p = .0003, Cohen's d = 0.77). In the EC-SCZ group, the DLPFC natural frequency was inversely associated with negative symptoms. Across all participants, the beta band relative spectral power negatively correlated with AX-Continuous Performance Task performance. CONCLUSIONS DLPFC oscillatory slowing is an early pathophysiological biomarker of schizophrenia that is associated with its symptom severity and cognitive impairments. Future work should assess whether noninvasive neurostimulation, including repetitive TMS, can ameliorate prefrontal oscillatory deficits and related clinical functions in patients with EC-SCZ.
Collapse
Affiliation(s)
- Francesco L Donati
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Health Science, University of Milan, Milan, Italy
| | - Ahmad Mayeli
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Kamakashi Sharma
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sabine Janssen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert J Krafty
- Department of Biostatistics & Bioinformatics, Emory University, Atlanta, Georgia
| | - Adenauer G Casali
- Institute of Science and Technology, Federal University of São Paulo, São José dos Campos, Brazil
| | - Fabio Ferrarelli
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
18
|
Kuban W, Haduch A, Bromek E, Basińska-Ziobroń A, Gawlińska K, Gawliński D, Filip M, Daniel WA. The Effect of Maternal High-Fat or High-Carbohydrate Diet during Pregnancy and Lactation on Cytochrome P450 2D (CYP2D) in the Liver and Brain of Rat Offspring. Int J Mol Sci 2024; 25:7904. [PMID: 39063146 PMCID: PMC11276948 DOI: 10.3390/ijms25147904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/10/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Cytochrome P450 2D (CYP2D) is important in psychopharmacology as it is engaged in the metabolism of drugs, neurosteroids and neurotransmitters. An unbalanced maternal diet during pregnancy and lactation can cause neurodevelopmental abnormalities and increases the offspring's predisposition to neuropsychiatric diseases. The aim of the present study was to evaluate the effect of maternal modified types of diet: a high-fat diet (HFD) and high-carbohydrate diet (HCD) during pregnancy and lactation on CYP2D in the liver and brain of male offspring at 28 (adolescent) or 63 postnatal days (young adult). The CYP2D activity and protein level were measured in the liver microsomes and the levels of mRNAs of CYP2D1, 2D2 and 2D4 were investigated both in the liver and brain. In the liver, both HFD and HCD increased the mRNA levels of all the three investigated CYP2D genes in adolescents, but an opposite effect was observed in young adults. The CYP2D protein level increased in adolescents but not in young adults. In contrast, young adults showed significantly decreased CYP2D activity. Similar effect of HFD on the CYP2D mRNAs was observed in the prefrontal cortex, while the effect of HCD was largely different than in the liver (the CYP2D2 expression was not affected, the CYP2D4 expression was decreased in young adults). In conclusion, modified maternal diets influence the expression of individual CYP2D1, CYP2D2 and CYP2D4 genes in the liver and brain of male offspring, which may affect the metabolism of CYP2D endogenous substrates and drugs and alter susceptibility to brain diseases and pharmacotherapy outcome.
Collapse
Affiliation(s)
- Wojciech Kuban
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland; (W.K.); (A.H.); (E.B.); (A.B.-Z.)
| | - Anna Haduch
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland; (W.K.); (A.H.); (E.B.); (A.B.-Z.)
| | - Ewa Bromek
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland; (W.K.); (A.H.); (E.B.); (A.B.-Z.)
| | - Agnieszka Basińska-Ziobroń
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland; (W.K.); (A.H.); (E.B.); (A.B.-Z.)
| | - Kinga Gawlińska
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland; (K.G.); (D.G.); (M.F.)
| | - Dawid Gawliński
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland; (K.G.); (D.G.); (M.F.)
| | - Małgorzata Filip
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland; (K.G.); (D.G.); (M.F.)
| | - Władysława A. Daniel
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland; (W.K.); (A.H.); (E.B.); (A.B.-Z.)
| |
Collapse
|
19
|
Bjornson KJ, Vanderplow AM, Bhasker AI, Cahill ME. Increased regional activity of a pro-autophagy pathway in schizophrenia as a contributor to sex differences in the disease pathology. Cell Rep Med 2024; 5:101652. [PMID: 39019008 PMCID: PMC11293356 DOI: 10.1016/j.xcrm.2024.101652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/14/2024] [Accepted: 06/19/2024] [Indexed: 07/19/2024]
Abstract
Based on recent genome-wide association studies, it is theorized that altered regulation of autophagy contributes to the pathophysiology of schizophrenia and bipolar disorder. As activity of autophagy-regulatory pathways is controlled by discrete phosphorylation sites on the relevant proteins, phospho-protein profiling is one of the few approaches available for enabling a quantitative assessment of autophagic activity in the brain. Despite this, a comprehensive phospho-protein assessment in the brains of schizophrenia and bipolar disorder subjects is currently lacking. Using this direction, our broad screening identifies an increase in AMP-activated protein kinase (AMPK)-mediated phospho-activation of the pro-autophagy protein beclin-1 solely in the prefrontal cortex of female, but not male, schizophrenia subjects. Using a reverse translational approach, we surprisingly find that this increase in beclin-1 activity facilitates synapse formation and enhances cognition. These findings are interpreted in the context of human studies demonstrating that female schizophrenia subjects have a lower susceptibility to cognitive dysfunction than males.
Collapse
Affiliation(s)
- Kathryn J Bjornson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Amanda M Vanderplow
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Aishwarya I Bhasker
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michael E Cahill
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
20
|
Hou W, Zhou F, Wang Q, Li H, Qin X, Ding Y, Dong F, Bo Q, Li A, Zhang L, Chen Z, Wang Z, Li X, Lee J, Wang C. Effect of transcranial direct current stimulation with concurrent cognitive performance targeting posterior parietal cortex vs prefrontal cortex on working memory in schizophrenia: a randomized clinical trial. Transl Psychiatry 2024; 14:279. [PMID: 38977683 PMCID: PMC11231223 DOI: 10.1038/s41398-024-02994-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024] Open
Abstract
Working memory deficits are linked to irregularities in the dorsolateral prefrontal cortex (DLPFC) and the posterior parietal cortex (PPC) in schizophrenia, effective intervention strategies are lacking. We evaluated the differential efficacy and underlying neuromechanisms of targeting transcranial direct current stimulation (tDCS) at the DLPFC and the PPC with concurrent cognitive performance for working memory in schizophrenia. In a randomized and double-blind clinical trial, sixty clinically stable schizophrenic patients with below-average working memory were randomly assigned to active DLPFC, active PPC, and sham tDCS groups. Two sessions of tDCS during N-back task were delivered daily for five days. The primary outcome was changes in spatial span test scores from baseline to week 1. The secondary outcomes included changes in scores of color delay-estimation task, other cognitive tasks, and mismatch negativity (biomarker of N-methyl-d-aspartate receptor functioning). Compared with the active DLPFC group, the active PPC group demonstrated significantly greater improvement in spatial span test scores (p = 0.008, d = 0.94) and an augmentation in color delay-estimation task capacity at week 1; the latter sustained to week 2. Compared with the sham tDCS group, the active PPC group did not show a significant improvement in spatial span test scores at week 1 and 2; however, significant enhancement was observed in their color delay-estimation task capacity at week 2. Additionally, mismatch negativity amplitude was enhanced, and changes in theta band measures were positively correlated with working memory improvement in the active PPC group, while no such correlations were observed in the active DLPFC group or the sham tDCS group. Our results suggest that tDCS targeting the PPC relative to the DLPFC during concurrent cognitive performance may improve working memory in schizophrenia, meriting further investigation. The improvement in working memory appears to be linked to enhanced N-methyl-d-aspartate receptor functioning.
Collapse
Affiliation(s)
- Wenpeng Hou
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Fuchun Zhou
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qi Wang
- Fengtai Mental Health Center, Beijing, China
| | - Hang Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Xiangqin Qin
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yushen Ding
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Fang Dong
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qijing Bo
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Anning Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Liang Zhang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Zhenzhu Chen
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Zhimin Wang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Xianbin Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Jimmy Lee
- Institute of Mental Health, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Chuanyue Wang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
21
|
Vergallito A, Gesi C, Torriero S. Intermittent Theta Burst Stimulation Combined with Cognitive Training to Improve Negative Symptoms and Cognitive Impairment in Schizophrenia: A Pilot Study. Brain Sci 2024; 14:683. [PMID: 39061423 PMCID: PMC11274516 DOI: 10.3390/brainsci14070683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Schizophrenia is a chronic psychiatric disorder severely affecting patients' functioning and quality of life. Unlike positive symptoms, cognitive impairment and negative symptoms cannot be treated pharmacologically and represent consistent predictors of the illness's prognosis. Cognitive remediation (CR) interventions have been applied to target these symptoms. Brain stimulation also provides promising yet preliminary results in reducing negative symptoms, whereas its effect on cognitive impairment remains heterogeneous. Here, we combined intermittent theta burst stimulation (iTBS) with CR to improve negative symptoms and cognitive impairment in schizophrenia spectrum patients. One hundred eligible patients were invited, and twenty-one participated. We randomized them into four groups, manipulating the stimulation condition (real vs. sham) and CR (no training vs. training). We delivered fifteen iTBS sessions over the left dorsolateral prefrontal cortex for three weeks, followed (or not) by 50 min of training. Consensus-based clinical and cognitive assessment was administered at baseline and after the treatment, plus at three follow-ups occurring one, three, and six months after the intervention. Mixed-model analyses were run on cognitive and negative symptom scores. The preliminary findings highlighted a marginal modulation of iTBS on negative symptoms, whereas CR improved isolated cognitive functions. We herein discuss the limitations and strengths of the methodological approach.
Collapse
Affiliation(s)
- Alessandra Vergallito
- Department of Psychology & Neuromi, University of Milano-Bicocca, 20126 Milan, Italy
| | - Camilla Gesi
- Department of Mental Health and Addictions, ASST Fatebenefratelli-Sacco, 20157 Milan, Italy (S.T.)
| | - Sara Torriero
- Department of Mental Health and Addictions, ASST Fatebenefratelli-Sacco, 20157 Milan, Italy (S.T.)
| |
Collapse
|
22
|
Kritzer MF, Adler A, Locklear M. Androgen effects on mesoprefrontal dopamine systems in the adult male brain. Neuroscience 2024:S0306-4522(24)00306-3. [PMID: 38977069 DOI: 10.1016/j.neuroscience.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Epidemiological data show that males are more often and/or more severely affected by symptoms of prefrontal cortical dysfunction in schizophrenia, Parkinson's disease and other disorders in which dopamine circuits associated with the prefrontal cortex are dysregulated. This review focuses on research showing that these dopamine circuits are powerfully regulated by androgens. It begins with a brief overview of the sex differences that distinguish prefrontal function in health and prefrontal dysfunction or decline in aging and/or neuropsychiatric disease. This review article then spotlights data from human subjects and animal models that specifically identify androgens as potent modulators of prefrontal cortical operations and of closely related, functionally critical measures of prefrontal dopamine level or tone. Candidate mechanisms by which androgens dynamically control mesoprefrontal dopamine systems and impact prefrontal states of hypo- and hyper-dopaminergia in aging and disease are then considered. This is followed by discussion of a working model that identifies a key locus for androgen modulation of mesoprefrontal dopamine systems as residing within the prefrontal cortex itself. The last sections of this review critically consider the ways in which the organization and regulation of mesoprefrontal dopamine circuits differ in the adult male and female brain, and highlights gaps where more research is needed.
Collapse
Affiliation(s)
- Mary F Kritzer
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, United States.
| | - Alexander Adler
- Department of Oncology and Immuno-Oncology, Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, United States
| | | |
Collapse
|
23
|
Mana L, Schwartz-Pallejà M, Vila-Vidal M, Deco G. Overview on cognitive impairment in psychotic disorders: From impaired microcircuits to dysconnectivity. Schizophr Res 2024; 269:132-143. [PMID: 38788432 DOI: 10.1016/j.schres.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Schizophrenia's cognitive deficits, often overshadowed by positive symptoms, significantly contribute to the disorder's morbidity. Increasing attention highlights these deficits as reflections of neural circuit dysfunction across various cortical regions. Numerous connectivity alterations linked to cognitive symptoms in psychotic disorders have been reported, both at the macroscopic and microscopic level, emphasizing the potential role of plasticity and microcircuits impairment during development and later stages. However, the heterogeneous clinical presentation of cognitive impairment and diverse connectivity findings pose challenges in summarizing them into a cohesive picture. This review aims to synthesize major cognitive alterations, recent insights into network structural and functional connectivity changes and proposed mechanisms and microcircuit alterations underpinning these symptoms, particularly focusing on neurodevelopmental impairment, E/I balance, and sleep disturbances. Finally, we will also comment on some of the most recent and promising therapeutic approaches that aim to target these mechanisms to address cognitive symptoms. Through this comprehensive exploration, we strive to provide an updated and nuanced overview of the multiscale connectivity impairment underlying cognitive impairment in psychotic disorders.
Collapse
Affiliation(s)
- L Mana
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain.
| | - M Schwartz-Pallejà
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Department of Experimental and Health Science, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Eurecat, Technology Center of Catalonia, Multimedia Technologies, Barcelona, Spain.
| | - M Vila-Vidal
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Computational Biology and Complex Systems Group, Department of Physics, Universitat Politècnica de Catalunya, Barcelona, Spain.
| | - G Deco
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Institució Catalana de la Recerca i Estudis Avançats (ICREA), Passeig Lluís Companys 23, Barcelona 08010, Spain.
| |
Collapse
|
24
|
Gonzalez Burgos G, Miyamae T, Nishihata Y, Krimer OL, Wade K, Fish KN, Arion D, Cai ZL, Xue M, Stauffer WR, Lewis DA. Synaptic alterations in pyramidal cells following genetic manipulation of neuronal excitability in monkey prefrontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598658. [PMID: 38915638 PMCID: PMC11195287 DOI: 10.1101/2024.06.12.598658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
In schizophrenia, layer 3 pyramidal neurons (L3PNs) in the dorsolateral prefrontal cortex (DLPFC) are thought to receive fewer excitatory synaptic inputs and to have lower expression levels of activity-dependent genes and of genes involved in mitochondrial energy production. In concert, these findings from previous studies suggest that DLPFC L3PNs are hypoactive in schizophrenia, disrupting the patterns of activity that are crucial for working memory, which is impaired in the illness. However, whether lower PN activity produces alterations in inhibitory and/or excitatory synaptic strength has not been tested in the primate DLPFC. Here, we decreased PN excitability in rhesus monkey DLPFC in vivo using adeno-associated viral vectors (AAVs) to produce Cre recombinase-mediated overexpression of Kir2.1 channels, a genetic silencing tool that efficiently decreases neuronal excitability. In acute slices prepared from DLPFC 7-12 weeks post-AAV microinjections, Kir2.1-overexpressing PNs had a significantly reduced excitability largely attributable to highly specific effects of the AAV-encoded Kir2.1 channels. Moreover, recordings of synaptic currents showed that Kir2.1-overexpressing DLPFC PNs had reduced strength of excitatory synapses whereas inhibitory synaptic inputs were not affected. The decrease in excitatory synaptic strength was not associated with changes in dendritic spine number, suggesting that excitatory synapse quantity was unaltered in Kir2.1-overexpressing DLPFC PNs. These findings suggest that, in schizophrenia, the excitatory synapses on hypoactive L3PNs are weaker and thus might represent a substrate for novel therapeutic interventions. Significance Statement In schizophrenia, dorsolateral prefrontal cortex (DLPFC) pyramidal neurons (PNs) have both transcriptional and structural alterations that suggest they are hypoactive. PN hypoactivity is thought to produce synaptic alterations in schizophrenia, however the effects of lower neuronal activity on synaptic function in primate DLPFC have not been examined. Here, we used, for the first time in primate neocortex, adeno-associated viral vectors (AAVs) to reduce PN excitability with Kir2.1 channel overexpression and tested if this manipulation altered the strength of synaptic inputs onto the Kir2.1-overexpressing PNs. Recordings in DLPFC slices showed that Kir2.1 overexpression depressed excitatory (but not inhibitory), synaptic currents, suggesting that, in schizophrenia, the hypoactivity of PNs might be exacerbated by reduced strength of the excitatory synapses they receive.
Collapse
|
25
|
Guimond S, Van Rheenen TE. Editorial: Progress in understanding cognitive dysfunction in severe psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110989. [PMID: 38458345 DOI: 10.1016/j.pnpbp.2024.110989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Affiliation(s)
- Synthia Guimond
- Department of Psychoeducation and Psychology, University of Quebec in Outaouais, Gatineau, Canada; The Royal's Institute of Mental Health Research, Department of Psychiatry, University of Ottawa, Ottawa, Canada.
| | - Tamsyn E Van Rheenen
- Department of Psychiatry, University of Melbourne, Melbourne, Australia; Centre for Mental Health and Brain Sciences, Swinburne University, Melbourne, Australia
| |
Collapse
|
26
|
Yang KC, Yang BH, Liu MN, Liou YJ, Chou YH. Cognitive impairment in schizophrenia is associated with prefrontal-striatal functional hypoconnectivity and striatal dopaminergic abnormalities. J Psychopharmacol 2024; 38:515-525. [PMID: 38853592 DOI: 10.1177/02698811241257877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
BACKGROUND A better understanding of the mechanisms underlying cognitive impairment in schizophrenia is imperative, as it causes poor functional outcomes and a lack of effective treatments. AIMS This study aimed to investigate the relationships of two proposed main pathophysiology of schizophrenia, altered prefrontal-striatal connectivity and the dopamine system, with cognitive impairment and their interactions. METHODS Thirty-three patients with schizophrenia and 27 healthy controls (HCs) who are right-handed and matched for age and sex were recruited. We evaluated their cognition, functional connectivity (FC) between the dorsolateral prefrontal cortex (DLPFC)/middle frontal gyrus (MiFG) and striatum, and the availability of striatal dopamine transporter (DAT) using a cognitive battery investigating attention, memory, and executive function, resting-state functional magnetic resonance imaging with group independent component analysis and single-photon emission computed tomography with 99mTc-TRODAT. RESULTS Patients with schizophrenia exhibited poorer cognitive performance, reduced FC between DLPFC/MiFG and the caudate nucleus (CN) or putamen, decreased DAT availability in the left CN, and decreased right-left DAT asymmetry in the CN compared to HCs. In patients with schizophrenia, altered imaging markers are associated with cognitive impairments, especially the relationship between DLPFC/MiFG-putamen FC and attention and between DAT asymmetry in the CN and executive function. CONCLUSIONS This study is the first to demonstrate how prefrontal-striatal hypoconnectivity and altered striatal DAT markers are associated with different domains of cognitive impairment in schizophrenia. More research is needed to evaluate their complex relationships and potential therapeutic implications.
Collapse
Affiliation(s)
- Kai-Chun Yang
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Bang-Hung Yang
- Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mu-N Liu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ying-Jay Liou
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yuan-Hwa Chou
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Psychiatry, Taichung Veterans General Hospital, Taichung, Taiwan
- The Human Brain Research Center, Taichung Veterans General Hospital, Taichung, Taiwan
| |
Collapse
|
27
|
Biačková N, Adamová A, Klírová M. Transcranial alternating current stimulation in affecting cognitive impairment in psychiatric disorders: a review. Eur Arch Psychiatry Clin Neurosci 2024; 274:803-826. [PMID: 37682331 PMCID: PMC11127835 DOI: 10.1007/s00406-023-01687-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/21/2023] [Indexed: 09/09/2023]
Abstract
Transcranial alternating current stimulation (tACS) is a non-invasive brain stimulation method that, through its manipulation of endogenous oscillations, can affect cognition in healthy adults. Given the fact that both endogenous oscillations and cognition are impaired in various psychiatric diagnoses, tACS might represent a suitable intervention. We conducted a search of Pubmed and Web of Science databases and reviewed 27 studies where tACS is used in psychiatric diagnoses and cognition change is evaluated. TACS is a safe and well-tolerated intervention method, suitable for multiple-sessions protocols. It can be administered at home, individualized according to the patient''s anatomical and functional characteristics, or used as a marker of disease progression. The results are varying across diagnoses and applied protocols, with some protocols showing a long-term effect. However, the overall number of studies is small with a great variety of diagnoses and tACS parameters, such as electrode montage or used frequency. Precise mechanisms of tACS interaction with pathophysiological processes are only partially described and need further research. Currently, tACS seems to be a feasible method to alleviate cognitive impairment in psychiatric patients; however, a more robust confirmation of efficacy of potential protocols is needed to introduce it into clinical practise.
Collapse
Affiliation(s)
- Nina Biačková
- Neurostimulation Department, National Institute of Mental Health, Klecany, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Andrea Adamová
- Neurostimulation Department, National Institute of Mental Health, Klecany, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Monika Klírová
- Neurostimulation Department, National Institute of Mental Health, Klecany, Czech Republic.
- Third Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
28
|
Huuki-Myers LA, Spangler A, Eagles NJ, Montgomery KD, Kwon SH, Guo B, Grant-Peters M, Divecha HR, Tippani M, Sriworarat C, Nguyen AB, Ravichandran P, Tran MN, Seyedian A, Hyde TM, Kleinman JE, Battle A, Page SC, Ryten M, Hicks SC, Martinowich K, Collado-Torres L, Maynard KR. A data-driven single-cell and spatial transcriptomic map of the human prefrontal cortex. Science 2024; 384:eadh1938. [PMID: 38781370 PMCID: PMC11398705 DOI: 10.1126/science.adh1938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 12/06/2023] [Indexed: 05/25/2024]
Abstract
The molecular organization of the human neocortex historically has been studied in the context of its histological layers. However, emerging spatial transcriptomic technologies have enabled unbiased identification of transcriptionally defined spatial domains that move beyond classic cytoarchitecture. We used the Visium spatial gene expression platform to generate a data-driven molecular neuroanatomical atlas across the anterior-posterior axis of the human dorsolateral prefrontal cortex. Integration with paired single-nucleus RNA-sequencing data revealed distinct cell type compositions and cell-cell interactions across spatial domains. Using PsychENCODE and publicly available data, we mapped the enrichment of cell types and genes associated with neuropsychiatric disorders to discrete spatial domains.
Collapse
Affiliation(s)
- Louise A Huuki-Myers
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Abby Spangler
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Nicholas J Eagles
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Kelsey D Montgomery
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Sang Ho Kwon
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Boyi Guo
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Melissa Grant-Peters
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Heena R Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Chaichontat Sriworarat
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Annie B Nguyen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Prashanthi Ravichandran
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21218, USA
| | - Matthew N Tran
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Arta Seyedian
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Alexis Battle
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21218, USA
- Department of Computer Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Stephanie C Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Mina Ryten
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Stephanie C Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21218, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD 21218, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Leonardo Collado-Torres
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
29
|
Chen X, Xiu H, Hou Y, Chen X, Liu F, Tu S. High-Frequency Repetitive Transcranial Magnetic Stimulation on Overall Cognition in Patients With Poststroke Cognitive Impairment: A Systematic Review and Meta-analysis. Am J Phys Med Rehabil 2024; 103:418-427. [PMID: 38113027 DOI: 10.1097/phm.0000000000002377] [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: 12/21/2023]
Abstract
OBJECTIVE The aim of the study is to comprehensively assess the recovery effects of high-frequency repetitive transcranial magnetic stimulation in patients with poststroke cognitive impairment. METHODS Six English and four Chinese databases were searched for relevant studies published up to January 2022. Randomized controlled trials of patients with poststroke cognitive impairment treated with high-frequency repetitive transcranial magnetic stimulation were included. Included studies were assessed for the risk of bias through the Cochrane Intervention Systematic Review Manual 5.1.0. The meta-analysis was performed using RevMan 5.4 software. The PRISMA 2020 guidelines were followed. RESULTS Sixty-one randomized controlled trials (4012 patients) were included. Montreal Cognitive Assessment Score, Mini-Mental State Examination score, event-related potential P300 (P300) amplitude, Loewenstein Occupational Therapy Cognitive Assessment score, and total effective rate of cognitive function were higher in the high-frequency repetitive transcranial magnetic stimulation group than in the control group at the end of the treatment period, and scores of Alzheimer Disease Assessment Scale-Cognitive Subscale and P300 latency were lower in the high-frequency repetitive transcranial magnetic stimulation group than in the control group. Fifty studies had selected the dorsolateral prefrontal cortex as the stimulation site for high-frequency repetitive transcranial magnetic stimulation. CONCLUSIONS Compared with nonrepetitive transcranial magnetic stimulation or sham repetitive transcranial magnetic stimulation, high-frequency repetitive transcranial magnetic stimulation not only improves the overall cognitive function of poststroke cognitive impairment patients but also has better rehabilitation results.
Collapse
Affiliation(s)
- Xin Chen
- From the Nursing College, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | | | | | | | | | | |
Collapse
|
30
|
Zhu J, Li J, Zhou L, Xu L, Pu C, Huang B, Zhou Q, Lin Y, Tang Y, Yang L, Shi C. Eye movements as predictor of cognitive improvement after cognitive remediation therapy in patients with schizophrenia. Front Psychiatry 2024; 15:1395198. [PMID: 38690204 PMCID: PMC11059054 DOI: 10.3389/fpsyt.2024.1395198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 03/26/2024] [Indexed: 05/02/2024] Open
Abstract
Aim Baseline cognitive functions of patients predicted the efficacy of cognitive remediation therapy (CRT), but results are mixed. Eye movement is a more objective and advanced assessment of cognitive functions than neuropsychological testing. We aimed to investigate the applicability of eye movements in predicting cognitive improvement after patients with schizophrenia were treated with CRT. Methods We recruited 79 patients with schizophrenia to complete 8 weeks of CRT and assessed their cognitive improvement outcomes. Eye movements were assessed by prosaccades, antisaccades, and free-viewing tasks at baseline, and neuropsychological tests in four cognitive domains were assessed before and after treatment to calculate treatment outcomes. Predictors of demographic information, clinical characteristics, and eye movement measures at baseline on cognitive improvement outcomes were analyzed using logistic regression analysis. We further compared the predictive performance between eye movement measurements and neuropsychological test regarding the effect of CRT on cognitive improvement, and explored factors that could be affect the treatment outcomes in different cognitive domains. Results As operationally defined, 33 patients showed improved in cognition (improved group) and 46 patients did not (non-improved group) after CRT. Patients with schizophrenia being employed, lower directional error rate in antisaccade task, and lower the gap effect (i.e., the difference in saccadic latency between the gap condition and overlap condition) in prosaccade task at baseline predicted cognitive improvement in CRT. However, performance in the free-viewing task not associated with cognitive improvement in patients in CRT. Our results show that eye-movement prediction model predicted the effect of CRT on cognitive improvement in patients with schizophrenia better than neuropsychological prediction model in CRT. In addition, baseline eye-movements, cognitive reserve, antipsychotic medication dose, anticholinergic cognitive burden change, and number of training sessions were associated with improvements in four cognitive domains. Conclusion Eye movements as a non-invasiveness, objective, and sensitive method of evaluating cognitive function, and combined saccadic measurements in pro- and anti-saccades tasks could be more beneficial than free-viewing task in predicting the effect of CRT on cognitive improvement in patients with schizophrenia.
Collapse
Affiliation(s)
- Jiahui Zhu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Peking University, Beijing, China
| | - Jinhao Li
- Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Li Zhou
- Faculty of Education, East China Normal University, Shanghai, China
| | - Lingzi Xu
- Research and Development Department, Infinite Brain Technologies, Beijing, China
| | - Chengcheng Pu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Peking University, Beijing, China
| | - Bingjie Huang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Peking University, Beijing, China
| | - Qi Zhou
- The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yunhan Lin
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Peking University, Beijing, China
| | - Yajing Tang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Peking University, Beijing, China
| | - Liu Yang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Peking University, Beijing, China
| | - Chuan Shi
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Peking University, Beijing, China
| |
Collapse
|
31
|
Negrón-Oyarzo I, Dib T, Chacana-Véliz L, López-Quilodrán N, Urrutia-Piñones J. Large-scale coupling of prefrontal activity patterns as a mechanism for cognitive control in health and disease: evidence from rodent models. Front Neural Circuits 2024; 18:1286111. [PMID: 38638163 PMCID: PMC11024307 DOI: 10.3389/fncir.2024.1286111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/11/2024] [Indexed: 04/20/2024] Open
Abstract
Cognitive control of behavior is crucial for well-being, as allows subject to adapt to changing environments in a goal-directed way. Changes in cognitive control of behavior is observed during cognitive decline in elderly and in pathological mental conditions. Therefore, the recovery of cognitive control may provide a reliable preventive and therapeutic strategy. However, its neural basis is not completely understood. Cognitive control is supported by the prefrontal cortex, structure that integrates relevant information for the appropriate organization of behavior. At neurophysiological level, it is suggested that cognitive control is supported by local and large-scale synchronization of oscillatory activity patterns and neural spiking activity between the prefrontal cortex and distributed neural networks. In this review, we focus mainly on rodent models approaching the neuronal origin of these prefrontal patterns, and the cognitive and behavioral relevance of its coordination with distributed brain systems. We also examine the relationship between cognitive control and neural activity patterns in the prefrontal cortex, and its role in normal cognitive decline and pathological mental conditions. Finally, based on these body of evidence, we propose a common mechanism that may underlie the impaired cognitive control of behavior.
Collapse
Affiliation(s)
- Ignacio Negrón-Oyarzo
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Tatiana Dib
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Lorena Chacana-Véliz
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Doctorado en Ciencias Mención en Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Nélida López-Quilodrán
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Doctorado en Ciencias Mención en Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jocelyn Urrutia-Piñones
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Doctorado en Ciencias Mención en Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
32
|
Li L, Liu C, Pan W, Wang W, Jin W, Ren Y, Ma X. Repetitive Transcranial Magnetic Stimulation for Working Memory Deficits in Schizophrenia: A Systematic Review of Randomized Controlled Trials. Neuropsychiatr Dis Treat 2024; 20:649-662. [PMID: 38528855 PMCID: PMC10962363 DOI: 10.2147/ndt.s450303] [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: 11/17/2023] [Accepted: 03/15/2024] [Indexed: 03/27/2024] Open
Abstract
Working memory (WM) deficits are a significant component of neurocognitive impairment in individuals with schizophrenia (SCZ). Two previous meta-analyses, conducted on randomized controlled trials (RCTs), examined the effectiveness of repetitive transcranial magnetic stimulation (rTMS) in addressing WM deficits in individuals diagnosed with SCZ. However, the conclusions drawn from these analyses were inconsistent. Additionally, the commonly used random effects (RE) models might underestimate statistical errors, attributing a significant portion of perceived heterogeneity between studies to variations in study quality. Therefore, this review utilized both RE and quality effects (QE) models to assess relevant RCTs comparing TMS with sham intervention in terms of clinical outcomes. A comprehensive literature search was conducted using PubMed and Scopus databases, resulting in the inclusion of 13 studies for data synthesis. Overall, regardless of whether the RE or QE model was used, eligible RCTs suggested that the TMS and sham groups exhibited comparable therapeutic effects after treatment. The current state of research regarding the use of rTMS as a treatment for WM deficits in patients with SCZ remains in its preliminary phase. Furthermore, concerning the mechanism of action, the activation of brain regions focused on the dorsolateral prefrontal cortex and alterations in gamma oscillations may hold significant relevance in the therapeutic application of rTMS for addressing WM impairments. Finally, we believe that the application of closed-loop neuromodulation may contribute to the optimization of rTMS for WM impairment in patients with SCZ.
Collapse
Affiliation(s)
- Li Li
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Chaomeng Liu
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Weigang Pan
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Wen Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Wenqing Jin
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Yanping Ren
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Xin Ma
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
33
|
Meneghetti N, Vannini E, Mazzoni A. Rodents' visual gamma as a biomarker of pathological neural conditions. J Physiol 2024; 602:1017-1048. [PMID: 38372352 DOI: 10.1113/jp283858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 01/23/2024] [Indexed: 02/20/2024] Open
Abstract
Neural gamma oscillations (indicatively 30-100 Hz) are ubiquitous: they are associated with a broad range of functions in multiple cortical areas and across many animal species. Experimental and computational works established gamma rhythms as a global emergent property of neuronal networks generated by the balanced and coordinated interaction of excitation and inhibition. Coherently, gamma activity is strongly influenced by the alterations of synaptic dynamics which are often associated with pathological neural dysfunctions. We argue therefore that these oscillations are an optimal biomarker for probing the mechanism of cortical dysfunctions. Gamma oscillations are also highly sensitive to external stimuli in sensory cortices, especially the primary visual cortex (V1), where the stimulus dependence of gamma oscillations has been thoroughly investigated. Gamma manipulation by visual stimuli tuning is particularly easy in rodents, which have become a standard animal model for investigating the effects of network alterations on gamma oscillations. Overall, gamma in the rodents' visual cortex offers an accessible probe on dysfunctional information processing in pathological conditions. Beyond vision-related dysfunctions, alterations of gamma oscillations in rodents were indeed also reported in neural deficits such as migraine, epilepsy and neurodegenerative or neuropsychiatric conditions such as Alzheimer's, schizophrenia and autism spectrum disorders. Altogether, the connections between visual cortical gamma activity and physio-pathological conditions in rodent models underscore the potential of gamma oscillations as markers of neuronal (dys)functioning.
Collapse
Affiliation(s)
- Nicolò Meneghetti
- The Biorobotics Institute, Scuola Superiore Sant'Anna, Pisa, Italy
- Department of Excellence for Robotics and AI, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Eleonora Vannini
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy
| | - Alberto Mazzoni
- The Biorobotics Institute, Scuola Superiore Sant'Anna, Pisa, Italy
- Department of Excellence for Robotics and AI, Scuola Superiore Sant'Anna, Pisa, Italy
| |
Collapse
|
34
|
Arime Y, Saitoh Y, Ishikawa M, Kamiyoshihara C, Uchida Y, Fujii K, Takao K, Akiyama K, Ohkawa N. Activation of prefrontal parvalbumin interneurons ameliorates working memory deficit even under clinically comparable antipsychotic treatment in a mouse model of schizophrenia. Neuropsychopharmacology 2024; 49:720-730. [PMID: 38049583 PMCID: PMC10876596 DOI: 10.1038/s41386-023-01769-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 10/06/2023] [Accepted: 11/01/2023] [Indexed: 12/06/2023]
Abstract
One of the critical unmet medical needs in schizophrenia is the treatment for cognitive deficits. However, the neural circuit mechanisms of them remain unresolved. Previous studies utilizing animal models of schizophrenia did not consider the fact that patients with schizophrenia generally cannot discontinue antipsychotic medication due to the high risk of relapse. Here, we used multi-dimensional approaches, including histological analysis of the prelimbic cortex (PL), LC-MS/MS-based in vivo dopamine D2 receptor occupancy analysis for antipsychotics, in vivo calcium imaging, and behavioral analyses of mice using chemogenetics to investigate neural mechanisms and potential therapeutic strategies for working memory deficit in a chronic phencyclidine (PCP) mouse model of schizophrenia. Chronic PCP administration led to alterations in excitatory and inhibitory synapses, specifically in dendritic spines of pyramidal neurons, vesicular glutamate transporter 1 (VGLUT1) positive terminals, and parvalbumin (PV) positive GABAergic interneurons located in layer 2-3 of the PL. Continuous administration of olanzapine, which achieved a sustained therapeutic window of dopamine D2 receptor occupancy (60-80%) in the striatum, did not ameliorate these synaptic abnormalities and working memory deficit in the chronic PCP-treated mice. We demonstrated that chemogenetic activation of PV neurons in the PL, as confirmed by in vivo calcium imaging, ameliorated working memory deficit in this model even under clinically comparable olanzapine treatment which by itself inhibited only PCP-induced psychomotor hyperactivity. Our study suggests that targeting prefrontal PV neurons could be a promising therapeutic intervention for cognitive deficits in schizophrenia in combination with antipsychotic medication.
Collapse
Affiliation(s)
- Yosefu Arime
- Division for Memory and Cognitive Function, Research Center for Advanced Medical Science, Comprehensive Research Facilities for Advanced Medical Science, Dokkyo Medical University, Tochigi, Japan.
- Department of Biological Psychiatry and Neuroscience, Dokkyo Medical University School of Medicine, Tochigi, Japan.
| | - Yoshito Saitoh
- Division for Memory and Cognitive Function, Research Center for Advanced Medical Science, Comprehensive Research Facilities for Advanced Medical Science, Dokkyo Medical University, Tochigi, Japan
| | - Mikiko Ishikawa
- Division for Memory and Cognitive Function, Research Center for Advanced Medical Science, Comprehensive Research Facilities for Advanced Medical Science, Dokkyo Medical University, Tochigi, Japan
- Department of Biological Psychiatry and Neuroscience, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Chikako Kamiyoshihara
- Division for Memory and Cognitive Function, Research Center for Advanced Medical Science, Comprehensive Research Facilities for Advanced Medical Science, Dokkyo Medical University, Tochigi, Japan
| | - Yasuo Uchida
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kazuki Fujii
- Department of Behavioral Physiology, Faculty of Medicine, University of Toyama, Toyama, Japan
- Life Science Research Center, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Keizo Takao
- Department of Behavioral Physiology, Faculty of Medicine, University of Toyama, Toyama, Japan
- Life Science Research Center, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Kazufumi Akiyama
- Department of Biological Psychiatry and Neuroscience, Dokkyo Medical University School of Medicine, Tochigi, Japan
- Kawada Hospital, Okayama, Japan
| | - Noriaki Ohkawa
- Division for Memory and Cognitive Function, Research Center for Advanced Medical Science, Comprehensive Research Facilities for Advanced Medical Science, Dokkyo Medical University, Tochigi, Japan.
| |
Collapse
|
35
|
Vergallito A, Gramano B, La Monica K, Giuliani L, Palumbo D, Gesi C, Torriero S. Combining transcranial magnetic stimulation with training to improve social cognition impairment in schizophrenia: a pilot randomized controlled trial. Front Psychol 2024; 15:1308971. [PMID: 38445059 PMCID: PMC10912559 DOI: 10.3389/fpsyg.2024.1308971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/16/2024] [Indexed: 03/07/2024] Open
Abstract
Schizophrenia is a severe, chronic mental disorder that profoundly impacts patients' everyday lives. The illness's core features include positive and negative symptoms and cognitive impairments. In particular, deficits in the social cognition domain showed a tighter connection to patients' everyday functioning than the other symptoms. Social remediation interventions have been developed, providing heterogeneous results considering the possibility of generalizing the acquired improvements in patients' daily activities. In this pilot randomized controlled trial, we investigated the feasibility of combining fifteen daily cognitive and social training sessions with non-invasive brain stimulation to boost the effectiveness of the two interventions. We delivered intermittent theta burst stimulation (iTBS) over the left dorsolateral prefrontal cortex (DLPFC). Twenty-one patients were randomized into four groups, varying for the assigned stimulation condition (real vs. sham iTBS) and the type of cognitive intervention (training vs. no training). Clinical symptoms and social cognition tests were administered at five time points, i.e., before and after the treatment, and at three follow-ups at one, three, and six months after the treatments' end. Preliminary data show a trend in improving the competence in managing emotion in participants performing the training. Conversely, no differences were found in pre and post-treatment scores for emotion recognition, theory of mind, and attribution of intentions scores. The iTBS intervention did not induce additional effects on individuals' performance. The methodological approach's novelty and limitations of the present study are discussed.
Collapse
Affiliation(s)
| | - Bianca Gramano
- Department of Mental Health and Addictions, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Kevin La Monica
- Department of Mental Health and Addictions, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Luigi Giuliani
- Department of Psychiatry, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Davide Palumbo
- Department of Psychiatry, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Camilla Gesi
- Department of Mental Health and Addictions, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Sara Torriero
- Department of Mental Health and Addictions, ASST Fatebenefratelli-Sacco, Milan, Italy
| |
Collapse
|
36
|
Lamsma J, Raine A, Kia SM, Cahn W, Arold D, Banaj N, Barone A, Brosch K, Brouwer R, Brunetti A, Calhoun VD, Chew QH, Choi S, Chung YC, Ciccarelli M, Cobia D, Cocozza S, Dannlowski U, Dazzan P, de Bartolomeis A, Di Forti M, Dumais A, Edmond JT, Ehrlich S, Evermann U, Flinkenflügel K, Georgiadis F, Glahn DC, Goltermann J, Green MJ, Grotegerd D, Guerrero-Pedraza A, Ha M, Hong EL, Hulshoff Pol H, Iasevoli F, Kaiser S, Kaleda V, Karuk A, Kim M, Kircher T, Kirschner M, Kochunov P, Kwon JS, Lebedeva I, Lencer R, Marques TR, Meinert S, Murray R, Nenadić I, Nguyen D, Pearlson G, Piras F, Pomarol-Clotet E, Pontillo G, Potvin S, Preda A, Quidé Y, Rodrigue A, Rootes-Murdy K, Salvador R, Skoch A, Sim K, Spalletta G, Spaniel F, Stein F, Thomas-Odenthal F, Tikàsz A, Tomecek D, Tomyshev A, Tranfa M, Tsogt U, Turner JA, van Erp TGM, van Haren NEM, van Os J, Vecchio D, Wang L, Wroblewski A, Nickl-Jockschat T. Structural brain abnormalities and aggressive behaviour in schizophrenia: Mega-analysis of data from 2095 patients and 2861 healthy controls via the ENIGMA consortium. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.04.24302268. [PMID: 38370846 PMCID: PMC10871467 DOI: 10.1101/2024.02.04.24302268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Background Schizophrenia is associated with an increased risk of aggressive behaviour, which may partly be explained by illness-related changes in brain structure. However, previous studies have been limited by group-level analyses, small and selective samples of inpatients and long time lags between exposure and outcome. Methods This cross-sectional study pooled data from 20 sites participating in the international ENIGMA-Schizophrenia Working Group. Sites acquired T1-weighted and diffusion-weighted magnetic resonance imaging scans in a total of 2095 patients with schizophrenia and 2861 healthy controls. Measures of grey matter volume and white matter microstructural integrity were extracted from the scans using harmonised protocols. For each measure, normative modelling was used to calculate how much patients deviated (in z-scores) from healthy controls at the individual level. Ordinal regression models were used to estimate the associations of these deviations with concurrent aggressive behaviour (as odds ratios [ORs] with 99% confidence intervals [CIs]). Mediation analyses were performed for positive symptoms (i.e., delusions, hallucinations and disorganised thinking), impulse control and illness insight. Aggression and potential mediators were assessed with the Positive and Negative Syndrome Scale, Scale for the Assessment of Positive Symptoms or Brief Psychiatric Rating Scale. Results Aggressive behaviour was significantly associated with reductions in total cortical volume (OR [99% CI] = 0.88 [0.78, 0.98], p = .003) and global white matter integrity (OR [99% CI] = 0.72 [0.59, 0.88], p = 3.50 × 10-5) and additional reductions in dorsolateral prefrontal cortex volume (OR [99% CI] = 0.85 [0.74, 0.97], p =.002), inferior parietal lobule volume (OR [99% CI] = 0.76 [0.66, 0.87], p = 2.20 × 10-7) and internal capsule integrity (OR [99% CI] = 0.76 [0.63, 0.92], p = 2.90 × 10-4). Except for inferior parietal lobule volume, these associations were largely mediated by increased severity of positive symptoms and reduced impulse control. Conclusions This study provides evidence that the co-occurrence of positive symptoms, poor impulse control and aggressive behaviour in schizophrenia has a neurobiological basis, which may inform the development of therapeutic interventions.
Collapse
Affiliation(s)
- Jelle Lamsma
- Department of Criminology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Psychiatry, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Adrian Raine
- Department of Criminology, University of Pennsylvania, Philadelphia, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, USA
| | - Seyed M. Kia
- Department of Psychiatry, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Cognitive Science and Artificial Intelligence, Tilburg University, Tilburg, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Wiepke Cahn
- Department of Psychiatry, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Dominic Arold
- Division of Psychological and Social Medicine and Developmental Neurosciences, TU Dresden, Germany
| | - Nerisa Banaj
- Laboratory of Neuropsychiatry, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Annarita Barone
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, USA
| | - Rachel Brouwer
- Department of Psychiatry, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Complex Trait Genetics, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Arturo Brunetti
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Vince D. Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science, Georgia State University, Georgia Institute of Technology and Emory University, Atlanta, USA
| | - Qian H. Chew
- Department of Research, Institute of Mental Health, Singapore
| | - Sunah Choi
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, South Korea
| | - Young-Chul Chung
- Department of Psychiatry, Jeonbuk National University, Jeonju, South Korea
- Department of Psychiatry, Jeonbuk National University Hospital, Jeonju, South Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Mariateresa Ciccarelli
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Derin Cobia
- Department of Psychology and Neuroscience Center, Brigham Young University, Provo, USA
| | - Sirio Cocozza
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Paola Dazzan
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Andrea de Bartolomeis
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Marta Di Forti
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Alexandre Dumais
- Department of Psychiatry and Addictology, University of Montreal, Montreal, Canada
- Institut Philippe-Pinel, Montreal, Canada
| | - Jesse T. Edmond
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science, Georgia State University, Georgia Institute of Technology and Emory University, Atlanta, USA
- Department of Psychology, Georgia State University, Atlanta, USA
| | - Stefan Ehrlich
- Division of Psychological and Social Medicine and Developmental Neurosciences, TU Dresden, Germany
- Department of Child and Adolescent Psychiatry and Psychotherapy, TU Dresden, Germany
| | - Ulrika Evermann
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Kira Flinkenflügel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Foivos Georgiadis
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zürich, Switzerland
| | - David C. Glahn
- Department of Psychiatry, Harvard Medical School, Harvard, USA
- Olin Neuropsychiatry Research Center, Institute of Living, Hartford, USA
| | - Janik Goltermann
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Melissa J. Green
- Neuroscience Research Australia, Randwick, Australia
- School of Clinical Medicine, University of New South Wales, Sydney, Australia
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | | | - Minji Ha
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, South Korea
| | - Elliot L. Hong
- Department of Psychiatry and Behavioral Science, UTHealth Houston, Houston, USA
| | - Hilleke Hulshoff Pol
- Department of Psychiatry, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Psychology, Utrecht University, Utrecht, the Netherlands
| | - Felice Iasevoli
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Stefan Kaiser
- Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Vasily Kaleda
- Department of Youth Psychiatry, Mental Health Research Center, Moscow, Russia
| | - Andriana Karuk
- FIDMAG Germanes Hospitalaries Research Foundation, Barcelona, Spain
| | - Minah Kim
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, South Korea
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Matthias Kirschner
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zürich, Switzerland
- Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
- Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada
| | - Peter Kochunov
- Department of Psychiatry and Behavioral Science, UTHealth Houston, Houston, USA
| | - Jun Soo Kwon
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, South Korea
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, South Korea
| | - Irina Lebedeva
- Laboratory of Neuroimaging and Multimodal Analysis, Mental Health Research Center, Moscow, Russia
| | - Rebekka Lencer
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry and Psychotherapy, University of Lübeck, Germany
| | - Tiago R. Marques
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
- Institute of Clinical Sciences, Imperial College London, London, UK
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Institute for Translational Neuroscience, University of Münster, Münster, Germany
| | - Robin Murray
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Dana Nguyen
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, USA
| | - Godfrey Pearlson
- Olin Neuropsychiatry Research Center, Institute of Living, Hartford, USA
| | - Fabrizio Piras
- Laboratory of Neuropsychiatry, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Edith Pomarol-Clotet
- FIDMAG Germanes Hospitalaries Research Foundation, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Carlos III Health Institute, Barcelona, Spain
| | - Giuseppe Pontillo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
- Department of Electrical Engineering and Information Technology, University of Naples Federico II, Naples, Italy
| | - Stéphane Potvin
- Department of Psychiatry and Addictology, University of Montreal, Montreal, Canada
- Centre de Recherche de l’Institut Universitaire en Santé Mentale de Montréal, Montreal, Canada
| | - Adrian Preda
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, USA
| | - Yann Quidé
- Neuroscience Research Australia, Randwick, Australia
- School of Psychology, University of New South Wales, Sydney, Australia
| | - Amanda Rodrigue
- Department of Psychiatry, Harvard Medical School, Harvard, USA
| | - Kelly Rootes-Murdy
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science, Georgia State University, Georgia Institute of Technology and Emory University, Atlanta, USA
- Department of Psychology, Georgia State University, Atlanta, USA
| | - Raymond Salvador
- FIDMAG Germanes Hospitalaries Research Foundation, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Carlos III Health Institute, Barcelona, Spain
| | - Antonin Skoch
- National Institute of Mental Health, Klecany, Czech Republic
| | - Kang Sim
- Department of Research, Institute of Mental Health, Singapore
| | | | - Filip Spaniel
- National Institute of Mental Health, Klecany, Czech Republic
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | | | - Andràs Tikàsz
- Department of Psychiatry and Addictology, University of Montreal, Montreal, Canada
- Centre de Recherche de l’Institut Universitaire en Santé Mentale de Montréal, Montreal, Canada
| | - David Tomecek
- National Institute of Mental Health, Klecany, Czech Republic
- Institute of Computer Science, Czech Academy of Sciences, Prague, Czech Republic
- Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Alexander Tomyshev
- Laboratory of Neuroimaging and Multimodal Analysis, Mental Health Research Center, Moscow, Russia
| | - Mario Tranfa
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Uyanga Tsogt
- Department of Psychiatry, Jeonbuk National University, Jeonju, South Korea
| | - Jessica A. Turner
- Psychiatry and Behavioral Health, Ohio State University Wexner Medical Center, Columbus, USA
| | - Theo G. M. van Erp
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, USA
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine, USA
| | - Neeltje E. M. van Haren
- Department of Psychiatry, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Child and Adolescent Psychiatry and Psychology, Erasmus MC Sophia, Rotterdam, the Netherlands
| | - Jim van Os
- Department of Psychiatry, University Medical Center Utrecht, Utrecht, the Netherlands
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- Department of Psychiatry and Neuropsychology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Daniela Vecchio
- Laboratory of Neuropsychiatry, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Lei Wang
- Psychiatry and Behavioral Health, Ohio State University Wexner Medical Center, Columbus, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Chicago, USA
| | - Adrian Wroblewski
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Thomas Nickl-Jockschat
- Department of Psychiatry, University of Iowa, Iowa City, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, USA
| |
Collapse
|
37
|
Naveed M, Chao OY, Hill JW, Yang YM, Huston JP, Cao R. Circadian neurogenetics and its implications in neurophysiology, behavior, and chronomedicine. Neurosci Biobehav Rev 2024; 157:105523. [PMID: 38142983 PMCID: PMC10872425 DOI: 10.1016/j.neubiorev.2023.105523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 12/26/2023]
Abstract
The circadian rhythm affects multiple physiological processes, and disruption of the circadian system can be involved in a range of disease-related pathways. The genetic underpinnings of the circadian rhythm have been well-studied in model organisms. Significant progress has been made in understanding how clock genes affect the physiological functions of the nervous system. In addition, circadian timing is becoming a key factor in improving drug efficacy and reducing drug toxicity. The circadian biology of the target cell determines how the organ responds to the drug at a specific time of day, thus regulating pharmacodynamics. The current review brings together recent advances that have begun to unravel the molecular mechanisms of how the circadian clock affects neurophysiological and behavioral processes associated with human brain diseases. We start with a brief description of how the ubiquitous circadian rhythms are regulated at the genetic, cellular, and neural circuit levels, based on knowledge derived from extensive research on model organisms. We then summarize the latest findings from genetic studies of human brain disorders, focusing on the role of human clock gene variants in these diseases. Lastly, we discuss the impact of common dietary factors and medications on human circadian rhythms and advocate for a broader application of the concept of chronomedicine.
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Owen Y Chao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Joseph P Huston
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Ruifeng Cao
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA; Department of Neurology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
38
|
Besteher B, Rocktäschel T, Garza AP, Machnik M, Ballez J, Helbing DL, Finke K, Reuken P, Güllmar D, Gaser C, Walter M, Opel N, Rita Dunay I. Cortical thickness alterations and systemic inflammation define long-COVID patients with cognitive impairment. Brain Behav Immun 2024; 116:175-184. [PMID: 38036270 DOI: 10.1016/j.bbi.2023.11.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/26/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023] Open
Abstract
As the heterogeneity of symptoms is increasingly recognized among long-COVID patients, it appears highly relevant to study potential pathophysiological differences along the different subtypes. Preliminary evidence suggests distinct alterations in brain structure and systemic inflammatory patterns in specific groups of long-COVID patients. To this end, we analyzed differences in cortical thickness and peripheral immune signature between clinical subgroups based on 3 T-MRI scans and signature inflammatory markers in n = 120 participants comprising healthy never-infected controls (n = 30), healthy COVID-19 survivors (n = 29), and subgroups of long-COVID patients with (n = 26) and without (n = 35) cognitive impairment according to screening with Montreal Cognitive Assessment. Whole-brain comparison of cortical thickness between the 4 groups was conducted by surface-based morphometry. We identified distinct cortical areas showing a progressive increase in cortical thickness across different groups, starting from healthy individuals who had never been infected with COVID-19, followed by healthy COVID-19 survivors, long-COVID patients without cognitive deficits (MoCA ≥ 26), and finally, long-COVID patients exhibiting significant cognitive deficits (MoCA < 26). These findings highlight the continuum of cortical thickness alterations associated with COVID-19, with more pronounced changes observed in individuals experiencing cognitive impairment (p < 0.05, FWE-corrected). Affected cortical regions covered prefrontal and temporal gyri, insula, posterior cingulate, parahippocampal gyrus, and parietal areas. Additionally, we discovered a distinct immunophenotype, with elevated levels of IL-10, IFNγ, and sTREM2 in long-COVID patients, especially in the group suffering from cognitive impairment. We demonstrate lingering cortical and immunological alterations in healthy and impaired subgroups of COVID-19 survivors. This implies a complex underlying pathomechanism in long-COVID and emphasizes the necessity to investigate the whole spectrum of post-COVID biology to determine targeted treatment strategies targeting specific sub-groups.
Collapse
Affiliation(s)
- Bianca Besteher
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany; German Center for Mental Health (DZPG), Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany.
| | - Tonia Rocktäschel
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany; German Center for Mental Health (DZPG), Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| | - Alejandra P Garza
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University, Magdeburg, Germany
| | - Marlene Machnik
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany
| | - Johanna Ballez
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany
| | - Dario-Lucas Helbing
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany; German Center for Mental Health (DZPG), Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| | - Kathrin Finke
- Department of Neurology, Jena University Hospital, Germany
| | - Philipp Reuken
- Department of Internal Medicine IV, Gastroenterology, Hepatology and Infectious Diseases, Jena University Hospital, Germany
| | - Daniel Güllmar
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital - Friedrich Schiller University Jena, Germany
| | - Christian Gaser
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany; German Center for Mental Health (DZPG), Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany; Department of Neurology, Jena University Hospital, Germany
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany; German Center for Mental Health (DZPG), Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| | - Nils Opel
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany; German Center for Mental Health (DZPG), Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| | - Ildiko Rita Dunay
- German Center for Mental Health (DZPG), Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany; Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University, Magdeburg, Germany
| |
Collapse
|
39
|
Sohal VS. Neurobiology of schizophrenia. Curr Opin Neurobiol 2024; 84:102820. [PMID: 38091860 DOI: 10.1016/j.conb.2023.102820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/23/2023] [Accepted: 11/19/2023] [Indexed: 02/18/2024]
Affiliation(s)
- Vikaas S Sohal
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, Center for Integrative Neuroscience, University of California, San Francisco, San Francisco, CA 94143-0444, USA.
| |
Collapse
|
40
|
Yao S, Harder A, Darki F, Chang YW, Li A, Nikouei K, Volpe G, Lundström JN, Zeng J, Wray N, Lu Y, Sullivan PF, Leffler JH. Connecting genomic results for psychiatric disorders to human brain cell types and regions reveals convergence with functional connectivity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.18.24301478. [PMID: 38410450 PMCID: PMC10896415 DOI: 10.1101/2024.01.18.24301478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Understanding the temporal and spatial brain locations etiological for psychiatric disorders is essential for targeted neurobiological research. Integration of genomic insights from genome-wide association studies with single-cell transcriptomics is a powerful approach although past efforts have necessarily relied on mouse atlases. Leveraging a comprehensive atlas of the adult human brain, we prioritized cell types via the enrichment of SNP-heritabilities for brain diseases, disorders, and traits, progressing from individual cell types to brain regions. Our findings highlight specific neuronal clusters significantly enriched for the SNP-heritabilities for schizophrenia, bipolar disorder, and major depressive disorder along with intelligence, education, and neuroticism. Extrapolation of cell-type results to brain regions reveals important patterns for schizophrenia with distinct subregions in the hippocampus and amygdala exhibiting the highest significance. Cerebral cortical regions display similar enrichments despite the known prefrontal dysfunction in those with schizophrenia highlighting the importance of subcortical connectivity. Using functional MRI connectivity from cases with schizophrenia and neurotypical controls, we identified brain networks that distinguished cases from controls that also confirmed involvement of the central and lateral amygdala, hippocampal body, and prefrontal cortex. Our findings underscore the value of single-cell transcriptomics in decoding the polygenicity of psychiatric disorders and offer a promising convergence of genomic, transcriptomic, and brain imaging modalities toward common biological targets.
Collapse
Affiliation(s)
- Shuyang Yao
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Arvid Harder
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Fahimeh Darki
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Yu-Wei Chang
- Department of Physics, University of Gothenburg, Gothenburg, Sweden
| | - Ang Li
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Kasra Nikouei
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Giovanni Volpe
- Department of Physics, University of Gothenburg, Gothenburg, Sweden
| | - Johan N Lundström
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | - Jian Zeng
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Naomi Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Yi Lu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Jens Hjerling Leffler
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
41
|
Vita A, Nibbio G, Barlati S. Pharmacological Treatment of Cognitive Impairment Associated With Schizophrenia: State of the Art and Future Perspectives. SCHIZOPHRENIA BULLETIN OPEN 2024; 5:sgae013. [PMID: 39144119 PMCID: PMC11207676 DOI: 10.1093/schizbullopen/sgae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Cognitive Impairment Associated with Schizophrenia (CIAS) represents one of the core dimensions of Schizophrenia Spectrum Disorders (SSD), with an important negative impact on real-world functional outcomes of people living with SSD. Treatment of CIAS represents a therapeutic goal of considerable importance, and while cognition-oriented evidence-based psychosocial interventions are available, effective pharmacological treatment could represent a game-changer in the lives of people with SSD. The present critical review reports and discusses the evidence regarding the effects of several pharmacological agents that are available in clinical practice or are under study, commenting on both current and future perspectives of CIAS treatment. In particular, the effects on CIAS of antipsychotic medications, anticholinergic medications, benzodiazepines, which are currently commonly used in the treatment of SSD, and of iclepertin, d-serine, luvadaxistat, xanomeline-trospium, ulotaront, anti-inflammatory molecules, and oxytocin, which are undergoing regulatory trials or can be considered as experimental agents, will be reported and discussed. Currently, available pharmacological agents do not appear to provide substantial benefits on CIAS, but accurate management of antipsychotic medications and avoiding treatments that can further exacerbate CIAS represent important strategies. Some molecules that are currently being investigated in Phase 2 and Phase 3 trials have provided very promising preliminary results, but more information is currently required to assess their effectiveness in real-world contexts and to provide clear recommendations regarding their use in clinical practice. The results of ongoing and future studies will reveal whether any of these molecules represents the awaited pharmacological game-changer in the treatment of CIAS.
Collapse
Affiliation(s)
- Antonio Vita
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Department of Mental Health and Addiction Services, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Gabriele Nibbio
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stefano Barlati
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Department of Mental Health and Addiction Services, ASST Spedali Civili of Brescia, Brescia, Italy
| |
Collapse
|
42
|
Thompson SM. Modulators of GABA A receptor-mediated inhibition in the treatment of neuropsychiatric disorders: past, present, and future. Neuropsychopharmacology 2024; 49:83-95. [PMID: 37709943 PMCID: PMC10700661 DOI: 10.1038/s41386-023-01728-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/14/2023] [Accepted: 08/08/2023] [Indexed: 09/16/2023]
Abstract
The predominant inhibitory neurotransmitter in the brain, γ-aminobutyric acid (GABA), acts at ionotropic GABAA receptors to counterbalance excitation and regulate neuronal firing. GABAA receptors are heteropentameric channels comprised from subunits derived from 19 different genes. GABAA receptors have one of the richest and well-developed pharmacologies of any therapeutic drug target, including agonists, antagonists, and positive and negative allosteric modulators (PAMs, NAMs). Currently used PAMs include benzodiazepine sedatives and anxiolytics, barbiturates, endogenous and synthetic neurosteroids, and general anesthetics. In this article, I will review evidence that these drugs act at several distinct binding sites and how they can be used to alter the balance between excitation and inhibition. I will also summarize existing literature regarding (1) evidence that changes in GABAergic inhibition play a causative role in major depression, anxiety, postpartum depression, premenstrual dysphoric disorder, and schizophrenia and (2) whether and how GABAergic drugs exert beneficial effects in these conditions, focusing on human studies where possible. Where these classical therapeutics have failed to exert benefits, I will describe recent advances in clinical and preclinical drug development. I will also highlight opportunities to advance a generation of GABAergic therapeutics, such as development of subunit-selective PAMs and NAMs, that are engendering hope for novel tools to treat these devastating conditions.
Collapse
Affiliation(s)
- Scott M Thompson
- Center for Novel Therapeutics, Department of Psychiatry, University of Colorado School of Medicine, 12700 E. 19th Ave., Aurora, CO, 80045, USA.
| |
Collapse
|
43
|
Han K, Liu G, Liu N, Li J, Li J, Cui L, Cheng M, Long J, Liao X, Tang Z, Liu Y, Liu J, Chen J, Lu H, Zhang H. Effects of Mobile Intelligent Cognitive Training for Patients with Post-Stroke Cognitive Impairment: A 12-Week, Multicenter, Randomized Controlled Study. J Alzheimers Dis 2024; 100:999-1015. [PMID: 38968051 DOI: 10.3233/jad-240356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024]
Abstract
Background The current application effects of computerized cognitive intervention are inconsistent and limited to hospital rehabilitation settings. Objective To investigate the effect of mobile intelligent cognitive training (MICT) on patients with post-stroke cognitive impairment (PSCI). Methods This study was a multicenter, prospective, open-label, blinded endpoint, cluster-randomized controlled trial (RCT). 518 PSCI patients were stratified and assigned to four rehabilitation settings, and then patients were randomized into experimental and control groups in each rehabilitation setting through cluster randomization. All patients received comprehensive management for PSCI, while the experimental group additionally received MICT intervention. Treatment was 30 minutes daily, 5 days per week, for 12 weeks. Cognitive function, activities of daily living (ADL), and quality of life (QOL) were assessed before the treatment, at weeks 6 and 12 post-treatment, and a 16-week follow-up. Results Linear Mixed Effects Models showed patients with PSCI were better off than pre-treatment patients on each outcome measure (p < 0.05). Additionally, the improvement of these outcomes in the experimental group was significantly better than in the control group at week 6 post-treatment and 16-week follow-up (p < 0.05). The rehabilitation setting also affected the cognitive efficacy of MICT intervention in improving PSCI patients, and the degree of improvement in each outcome was found to be highest in hospital, followed by community, nursing home, and home settings. Conclusions Long-term MICT intervention can improve cognition, ADL, and QOL in patients with PSCI, with sustained effects for at least one month. Notably, different rehabilitation settings affect the cognitive intervention efficacy of MICT on PSCI patients. However, this still needs to be further determined in future studies.
Collapse
Affiliation(s)
- Kaiyue Han
- School of Rehabilitation, Capital Medical University, Beijing, China
- Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
| | | | - Nan Liu
- Beijing Puren Hospital, Beijing, China
| | - Jiangyi Li
- Beijing Dongcheng District Kangfu One Two Three Health Training Center, Beijing, China
| | - Jianfeng Li
- Beijing Yangfangdian Hospital, Beijing, China
| | - Lihua Cui
- Beijing Fengtai District Jiaxiang Nursing-Home for the Elderly, Beijing, China
- Beijing Fengtai You Anmen Hospital, Beijing, China
| | - Ming Cheng
- Beijing Haidian District Guolilai Elderly Care Center, Beijing, China
| | - Junzi Long
- School of Rehabilitation, Capital Medical University, Beijing, China
- Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
- Changping Laboratory, Beijing, China
| | - Xingxing Liao
- School of Rehabilitation, Capital Medical University, Beijing, China
- Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
- Changping Laboratory, Beijing, China
| | - Zhiqing Tang
- School of Rehabilitation, Capital Medical University, Beijing, China
- Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
| | - Ying Liu
- School of Rehabilitation, Capital Medical University, Beijing, China
- Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
| | - Jiajie Liu
- School of Rehabilitation, Capital Medical University, Beijing, China
- Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
| | - Jiarou Chen
- Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Haitao Lu
- School of Rehabilitation, Capital Medical University, Beijing, China
- Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
| | - Hao Zhang
- School of Rehabilitation, Capital Medical University, Beijing, China
- Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
- University of Health and Rehabilitation Sciences, Qingdao, China
- Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
44
|
Hoptman MJ, Girgis RR, Javitt DC. Biomarkers for Cognitive Control, Response Inhibition, Aggressivity, Impulsivity, and Violence. ADVANCES IN NEUROBIOLOGY 2024; 40:725-756. [PMID: 39562462 DOI: 10.1007/978-3-031-69491-2_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Deficits in cognitive control contribute to behavioral impairments across neuropsychiatric disorders. Cognitive control is captured as a construct in the Research Domain Construct (RDoC) matrix and incorporate subdomains of goal selection, response selection, and performance monitoring. Relevant tasks for these subdomains include the "AX" version of the continuous performance task (goal selection) and the Go/NoGo and Stop-Signal reaction time tasks (response selection). Underlying mechanisms for these domains have been investigated intensively using fMRI and event-related potential (ERP) approaches, which provide candidate biomarkers for translational research. In RDoC, impulsive behaviors are provisionally assigned to the cognitive control/response selection construct, but other factors may also contribute. Impulsivity has gained increased importance over recent years due to its link to aggression and suicidality, which is mediated especially through the constructs of urgency and frustrative nonreward. These constructs, in turn, may be captured through scales such as the Urgency, (Lack of) Premeditation, (Lack of) Perseverance, and Sensation Seeking (UPPS-P) impulsivity scale and the Point Subtraction Aggression Paradigm (PSAP), respectively. At present, no validated biomarkers exist for either urgency or aggressivity. Potential directions for the development of predictive biomarkers for both targets are discussed.
Collapse
Affiliation(s)
- Matthew J Hoptman
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Ragy R Girgis
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Daniel C Javitt
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
45
|
Mittli D. Inflammatory processes in the prefrontal cortex induced by systemic immune challenge: Focusing on neurons. Brain Behav Immun Health 2023; 34:100703. [PMID: 38033612 PMCID: PMC10682838 DOI: 10.1016/j.bbih.2023.100703] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/30/2023] [Accepted: 11/04/2023] [Indexed: 12/02/2023] Open
Abstract
Peripheral immune challenge induces neurobiological alterations in the brain and related neuropsychiatric symptoms both in humans and other mammals. One of the best known physiological effects of systemic inflammation is sickness behavior. However, in addition to this depression-like state, there are other cognitive outcomes of peripherally induced neuroinflammation that can be linked to the dysfunction of higher-order cortical areas, such as the prefrontal cortex (PFC). As the physiological activity of the PFC is largely based on the balanced interplay of excitatory pyramidal cells and inhibitory interneurons, it may be hypothesized that neuroinflammatory processes result in a shift of excitatory/inhibitory balance, which is a common hallmark of several neuropsychiatric conditions. Indeed, many data suggest that peripherally induced neuroinflammation is strongly associated with molecular and functional changes in PFC neurons leading to disturbances in their synaptic networks. Different experimental approaches may cause some incongruence in the reviewed data. However, it is commonly agreed that acute systemic inflammation leads to changes in the excitatory/inhibitory balance in the PFC by proinflammatory signaling at the brain borders and in the brain parenchyma. These cellular changes result in altered local and brain-wide network activity inducing disturbances in the top-down control of goal-directed behavior and cognition regulated by the PFC. Lipopolysaccharide (LPS)-treated rodents are the most widely used experimental models of peripherally induced neuroinflammation, so the majority of the reviewed data come from studies utilizing the LPS model. This may limit their general interpretation regarding the neuronal effects of peripheral immune activation. In addition, several biological variables (e.g., sex, age) can influence the PFC effects of systemic immune challenge, not only the nature and severity of immune activation. Therefore, it would be desirable to investigate inflammation-related neuronal changes in the PFC using other models of systemic inflammation as well, and to focus on the targeted fine-tuning of the affected cell types via common molecular mechanisms of the immune and nervous systems.
Collapse
Affiliation(s)
- Dániel Mittli
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Physiology and Neurobiology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- InnoScience Ltd., Mátranovák, Hungary
| |
Collapse
|
46
|
Dienel SJ, Dowling KF, Barile Z, Bazmi HH, Liu A, Vespoli JC, Fish KN, Lewis DA. Diagnostic Specificity and Association With Cognition of Molecular Alterations in Prefrontal Somatostatin Neurons in Schizophrenia. JAMA Psychiatry 2023; 80:1235-1245. [PMID: 37647039 PMCID: PMC10469307 DOI: 10.1001/jamapsychiatry.2023.2972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/14/2023] [Indexed: 09/01/2023]
Abstract
Importance Individuals with schizophrenia (SZ) exhibit pronounced deficits in somatostatin (SST) messenger RNA (mRNA) levels in the dorsolateral prefrontal cortex (DLPFC). Molecularly distinct subtypes of SST neurons, located in the superficial and deep zones of the DLPFC, are thought to contribute to different functional processes of this region; understanding the specificity of SST alterations in SZ across these zones could inform the functional consequences of those alterations, including cognitive impairments characteristic of SZ. Objective To quantify mRNA levels of SST and related neuropeptides in the DLPFC in individuals with SZ, bipolar disorder (BPD), or major depressive disorder (MDD) and unaffected comparison individuals. Design, Setting, and Participants This case-control study, conducted from January 20, 2020, to March 30, 2022, used postmortem brain tissue specimens previously obtained from individuals with SZ, MDD, or BPD and unaffected individuals from a community population through 2 medical examiners' offices. Demographic, clinical, and educational information was ascertained through psychological autopsies. Exposures Diagnosis of SZ, BPD, or MDD. Main Outcome and Measures The main outcome was levels of SST and related neuropeptide mRNA in 2 DLPFC zones, examined using laser microdissection and quantitative polymerase chain reaction or fluorescent in situ hybridization (FISH). Findings were compared using educational attainment as a proxy measure of premorbid cognition. Results A total of 200 postmortem brain specimens were studied, including 65 from unaffected comparison individuals (42 [65%] male; mean [SD] age, 49.2 [14.1] years); 54 from individuals with SZ (37 [69%] male; mean [SD] age, 47.5 [13.3] years); 42 from individuals with MDD (24 [57%] male; mean [SD] age, 45.6 [12.1] years); and 39 from individuals with BPD (23 [59%] male; mean (SD) age, 46.2 [12.5] years). Compared with unaffected individuals, levels of SST mRNA were lower in both superficial (Cohen d, 0.68; 95% CI, 0.23-1.13; P = .004) and deep (Cohen d, 0.60; 95% CI, 0.16-1.04; P = .02) DLPFC zones in individuals with SZ; findings were confirmed using FISH. Levels of SST were lower only in the superficial zone in the group with MDD (Cohen d, 0.58; 95% CI, 0.14-1.02; P = .12), but the difference was not significant; SST levels were not lower in either zone in the BPD group. Levels of neuropeptide Y and tachykinin 1 showed similar patterns. Neuropeptide alterations in the superficial, but not deep, zone were associated with lower educational attainment only in the group with SZ (superficial: adjusted odds ratio, 1.71 [95% CI, 1.11-2.69]; P = .02; deep: adjusted odds ratio, 1.08 [95% CI, 0.64-1.84]; P = .77). Conclusions and Relevance The findings revealed diagnosis-specific patterns of molecular alterations in SST neurons in the DLPFC, suggesting that distinct disease processes are reflected in the differential vulnerability of SST neurons in individuals with SZ, MDD, and BPD. In SZ, alterations specifically in the superficial zone may be associated with cognitive dysfunction.
Collapse
Affiliation(s)
- Samuel J. Dienel
- Medical Scientist Training Program, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Kevin F. Dowling
- Medical Scientist Training Program, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zackery Barile
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - H. Holly Bazmi
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Amy Liu
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Julia C. Vespoli
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kenneth N. Fish
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David A. Lewis
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania
| |
Collapse
|
47
|
Zhuo C, Tian H, Zhu J, Fang T, Ping J, Wang L, Sun Y, Cheng L, Chen C, Chen G. Low-dose lithium adjunct to quetiapine improves cognitive task performance in mice with MK801-induced long-term cognitive impairment: Evidence from a pilot study. J Affect Disord 2023; 340:42-52. [PMID: 37506773 DOI: 10.1016/j.jad.2023.07.104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 07/04/2023] [Accepted: 07/23/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Low-dose lithium (LD-Li) has been shown to rescue cognitive impairment in mouse models of short-term mild cognitive impairment, dementia, and schizophrenia. However, few studies have characterized the effects of LD-Li, alone or in conjunction with anti-psychotics, in the mouse model of MK801-induced long term cognitive impairment. METHODS The present study used in vivo Ca2+ imaging and a battery of cognitive function assessments to investigate the long-term effects of LD-Li on cognition in mice exposed to repeated injections of MK801. Prefrontal Ca2+ activity was visualized to estimate alterations in neural activity in the model mice. Pre-pulse inhibition (PPI), novel object recognition (NOR), Morris water maze (MWM), and fear conditioning (FC) tasks were used to characterize cognitive performance; open field activity (OFA) testing was used to observe psychotic symptoms. Two treatment strategies were tested: LD-Li [250 mg/d human equivalent dose (HED)] adjunct to quetiapine (QTP; 600 mg/d HED); and QTP-monotherapy (mt; 600 mg/d HED). RESULTS Compared to the QTP-mt group, the LD-Li + QTP group showed greatly improved cognitive performance on all measures between experimental days 29 and 85. QTP-mt improved behavioral measures compared to untreated controls, but the effects persisted only from day 29 to day 43. These data suggest that LD-Li + QTP is superior to QTP-mt for improving long-term cognitive impairments in the MK801 mouse model. LIMITATIONS There is no medical consensus regarding lithium use in patients with schizophrenia. CONCLUSION More pre-clinical and clinical studies are needed to further investigate effective treatment strategies for patients with long-term cognitive impairments, such as chronic schizophrenia.
Collapse
Affiliation(s)
- Chuanjun Zhuo
- Key Laboratory of Sensory Information Processing Abnormalities in Schizophrenia (SIPAC_Lab), Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin 300140, China; Animal Imaging Center (AIC), Wenzhou Seventh Peoples Hospital, Wenzhou 325000, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjn Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin 300222, China.
| | - Hongjun Tian
- Key Laboratory of Sensory Information Processing Abnormalities in Schizophrenia (SIPAC_Lab), Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin 300140, China
| | - Jingjing Zhu
- Animal Imaging Center (AIC), Wenzhou Seventh Peoples Hospital, Wenzhou 325000, China
| | - Tao Fang
- Key Laboratory of Sensory Information Processing Abnormalities in Schizophrenia (SIPAC_Lab), Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin 300140, China
| | - Jing Ping
- Animal Imaging Center (AIC), Wenzhou Seventh Peoples Hospital, Wenzhou 325000, China
| | - Lina Wang
- Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjn Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin 300222, China
| | - Yun Sun
- Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjn Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin 300222, China
| | - Langlang Cheng
- Animal Imaging Center (AIC), Wenzhou Seventh Peoples Hospital, Wenzhou 325000, China
| | - Chunmian Chen
- Animal Imaging Center (AIC), Wenzhou Seventh Peoples Hospital, Wenzhou 325000, China
| | - Guangdong Chen
- Animal Imaging Center (AIC), Wenzhou Seventh Peoples Hospital, Wenzhou 325000, China
| |
Collapse
|
48
|
Hu X, Wang S, Zhou H, Li N, Zhong C, Luo W, Liu S, Fu F, Meng Y, Ding Z, Cheng B. Altered Functional Connectivity Strength in Distinct Brain Networks of Children With Early-Onset Schizophrenia. J Magn Reson Imaging 2023; 58:1617-1623. [PMID: 36932678 DOI: 10.1002/jmri.28682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 03/05/2023] [Accepted: 03/06/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Schizophrenia is regarded as a brain network or connectome disorder that is associated with neurodevelopment. Children with early-onset schizophrenia (EOS) provide an opportunity to evaluate the neuropathology of schizophrenia at a very early stage without potential confounding factors. But dysfunction in brain networks of schizophrenia is inconsistent. PURPOSE To identify abnormal functional connectivity (FC) in EOS patients and relationships with clinical symptoms, we aimed to reveal neuroimaging phenotypes of EOS. STUDY TYPE Prospective, cross-sectional. POPULATION Twenty-six female/22 male patients (age:14.3 ± 3.45 years) with first-episode EOS, 27 female/22 male age- and gender-matched healthy controls (HC) (age:14.1 ± 4.32). FIELD STRENGTH/SEQUENCE 3-T, resting-state (rs) gradient-echo echo-planar imaging and three-dimensional magnetization-prepared rapid gradient-echo imaging. ASSESSMENT Intelligence quotient (IQ) was measured by the Wechsler Intelligence Scale-Fourth edition for Children (WISC-IV). The clinical symptoms were evaluated by the Positive and Negative Syndrome Scale (PANSS). FC strength (FCS) from rs functional MRI (rsfMRI) was used to investigate functional integrity of global brain regions. In addition, associations between regionally altered FCS and clinical symptoms in EOS patients were examined. STATISTICAL TESTS Two-sample t-test controlling for sample size, diagnostic method, brain volume algorithm, and age of the subjects, Bonferroni correction, Pearson's correlation analysis. A P-value <0.05 with a minimum cluster size of 50 voxels was considered statistically significant. RESULTS Compared with HC, EOS patients had significantly lower total IQ scores (IQ:91.5 ± 16.1), increased FCS in the bilateral precuneus, left dorsolateral prefrontal cortex, left thalamus, and left parahippocampus (paraHIP), and decreased FCS in the right cerebellum posterior lobe and right superior temporal gyrus. The PANSS total score of EOS patients (PANSS total score:74.30 ± 7.23) was found to be positively correlated to FCS in the left paraHIP (r = 0.45). DATA CONCLUSION Our study revealed that disrupted FC of brain hubs illustrate multiple abnormalities in brain networks in EOS patients. EVIDENCE LEVEL 1 TECHNICAL EFFICACY STAGE: 2.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Rehabilitation Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Song Wang
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Hui Zhou
- Department of Rehabilitation Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Na Li
- Department of Rehabilitation Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Can Zhong
- Department of Rehabilitation Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Weiling Luo
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Sijia Liu
- School of Sociality and Psychology, Southwest Minzu University, Chengdu, China
| | - Fanghui Fu
- School of Sociality and Psychology, Southwest Minzu University, Chengdu, China
| | - Yajing Meng
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zhiyong Ding
- Department of Medical Imaging, Qujing Maternal and Child Health Care Hospital, Qujing, China
| | - Bochao Cheng
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
49
|
Hamzehpour L, Bohn T, Jaspers L, Grimm O. Exploring the link between functional connectivity of ventral tegmental area and physical fitness in schizophrenia and healthy controls. Eur Neuropsychopharmacol 2023; 76:77-86. [PMID: 37562082 DOI: 10.1016/j.euroneuro.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 08/12/2023]
Abstract
Decreased physical fitness and being overweight are highly prevalent in schizophrenia, represent a major risk factor for comorbid cardio-vascular diseases and decrease the life expectancy of the patients. Thus, it is important to understand the underlying mechanisms that link psychopathology and weight gain. We hypothesize that the dopaminergic reward system plays an important role in this. We analyzed the seed-based functional connectivity (FC) of the ventral tegmental area (VTA) in a group of schizophrenic patients (n=32) and age-, as well as gender-, matched healthy controls (n=27). We then correlated the resting-state results with physical fitness parameters, obtained in a fitness test, and psychopathology. The FC analysis revealed decreased functional connections between the VTA and the anterior cingulate cortex (ACC), as well as the dorsolateral prefrontal cortex, which negatively correlated with psychopathology, and increased FC between the VTA and the middle temporal gyrus in patients compared to healthy controls, which positively correlated with psychopathology. The decreased FC between the VTA and the ACC of the patient group further positively correlated with total body fat (p = .018, FDR-corr.) and negatively correlated with the overall physical fitness (p = .022). This study indicates a link between decreased physical fitness and higher body fat with functional dysconnectivity between the VTA and the ACC. These findings demonstrate that a dysregulated reward system might also be involved in comorbidities and could pave the way for future lifestyle therapy interventions.
Collapse
Affiliation(s)
- Lara Hamzehpour
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Heinrich-Hoffmann-Straße 10, 60528, Frankfurt am Main, Germany; Goethe University Frankfurt, Faculty 15 Biological Sciences, Frankfurt am Main, Germany.
| | - Tamara Bohn
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Heinrich-Hoffmann-Straße 10, 60528, Frankfurt am Main, Germany
| | - Lucia Jaspers
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Heinrich-Hoffmann-Straße 10, 60528, Frankfurt am Main, Germany
| | - Oliver Grimm
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Heinrich-Hoffmann-Straße 10, 60528, Frankfurt am Main, Germany
| |
Collapse
|
50
|
Schamiloglu S, Lewis E, Keeshen CM, Hergarden AC, Bender KJ, Whistler JL. Arrestin-3 Agonism at Dopamine D 3 Receptors Defines a Subclass of Second-Generation Antipsychotics That Promotes Drug Tolerance. Biol Psychiatry 2023; 94:531-542. [PMID: 36931452 PMCID: PMC10914650 DOI: 10.1016/j.biopsych.2023.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 02/09/2023] [Accepted: 03/02/2023] [Indexed: 03/19/2023]
Abstract
BACKGROUND Second-generation antipsychotics (SGAs) are frontline treatments for serious mental illness. Often, individual patients benefit only from some SGAs and not others. The mechanisms underlying this unpredictability in treatment efficacy remain unclear. All SGAs bind the dopamine D3 receptor (D3R) and are traditionally considered antagonists for dopamine receptor signaling. METHODS Here, we used a combination of two-photon calcium imaging, in vitro signaling assays, and mouse behavior to assess signaling by SGAs at D3R. RESULTS We report that some clinically important SGAs function as arrestin-3 agonists at D3R, resulting in modulation of calcium channels localized to the site of action potential initiation in prefrontal cortex pyramidal neurons. We further show that chronic treatment with an arrestin-3 agonist SGA, but not an antagonist SGA, abolishes D3R function through postendocytic receptor degradation by GASP1 (G protein-coupled receptor-associated sorting protein-1). CONCLUSIONS These results implicate D3R-arrestin-3 signaling as a source of SGA variability, highlighting the importance of including arrestin-3 signaling in characterizations of drug action. Furthermore, they suggest that postendocytic receptor trafficking that occurs during chronic SGA treatment may contribute to treatment efficacy.
Collapse
Affiliation(s)
- Selin Schamiloglu
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, California
| | - Elinor Lewis
- Neuroscience Graduate Group, University of California Davis, Davis, California; Center for Neuroscience, University of California Davis, Davis, California
| | - Caroline M Keeshen
- Neuroscience Graduate Group, University of California Davis, Davis, California; Center for Neuroscience, University of California Davis, Davis, California
| | - Anne C Hergarden
- Center for Neuroscience, University of California Davis, Davis, California
| | - Kevin J Bender
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, California; Department of Neurology, Kavli Institute for Fundamental Neuroscience, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California.
| | - Jennifer L Whistler
- Center for Neuroscience, University of California Davis, Davis, California; Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, California.
| |
Collapse
|