1
|
Adams B, Sinayskiy I, Agarwal S, Petruccione F. Entanglement and coherence in pure and doped Posner molecules. Sci Rep 2025; 15:12559. [PMID: 40221481 PMCID: PMC11993700 DOI: 10.1038/s41598-025-96487-5] [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: 11/05/2024] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
The potential role of spin in biological systems is a primary topic in quantum biology. However, much of this research focuses on electron spin. A recent hypothesis suggests that nuclear spin may be better suited to biological processes, being less sensitive to decoherence. The hypothesis details how phosphorus nuclei might be prepared in a spin entangled state, how this entanglement is protected by assembly into calcium phosphate (Posner) molecules, and how this entanglement might modulate calcium ion production and concomitant neural activation. In this paper we investigate the robustness of quantum effects such as coherence and entanglement in Posner molecules. We investigate how these effects are directly dependent on specific parameters such as spin-spin coupling strengths and Posner molecule symmetry. We also investigate how lithium isotope doped Posner molecules differentially modulate quantum resources such as coherence and entanglement and whether this is a viable explanation for lithium's mechanism of action in bipolar disease. Finally we illustrate how entanglement might possibly be preserved through exploitation of the biological environment.
Collapse
Affiliation(s)
- Betony Adams
- National Institute for Theoretical and Computational Sciences, Stellenbosch, South Africa.
- School of Data Science and Computational Thinking and Department of Physics, Stellenbosch University, Stellenbosch, South Africa.
| | - Ilya Sinayskiy
- National Institute for Theoretical and Computational Sciences, Stellenbosch, South Africa
- Quantum Research Group, University of KwaZulu-Natal, Durban, South Africa
| | - Shivang Agarwal
- The Department of Electrical and Computer Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Francesco Petruccione
- National Institute for Theoretical and Computational Sciences, Stellenbosch, South Africa
- School of Data Science and Computational Thinking and Department of Physics, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
2
|
Xiong G, Sachdeva M, Yosipovitch G, Ziv M, Dodiuk-Gad RP. Pruritus and Neuropsychiatric Symptoms Among Patients with Darier Disease-An Overlooked and Interconnected Challenge. J Clin Med 2025; 14:1818. [PMID: 40142630 PMCID: PMC11942685 DOI: 10.3390/jcm14061818] [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: 12/23/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
(1) Background: Darier disease (DD) is a rare autosomal dominant disorder caused by mutations in ATP2A2, a gene that encodes the sarco(endo)plasmic reticulum calcium-ATPase 2 enzyme, which disrupts calcium homeostasis in keratinocytes. Pruritus, a frequently overlooked symptom in DD, can lead to physical and emotional complications, especially in patients with DD who are genetically predisposed to psychiatric comorbidities. (2) Methods: This study aimed to analyze pruritus and other related symptoms in patients with DD and explore their correlation with neuropsychiatric conditions, psychological challenges, disease severity, and body surface area (BSA) involvement through a retrospective review of a tertiary center. (3) Results: Data from 76 patients (equal gender distribution, mean age 44 years) revealed a prevalence of pruritus of 90.8%, surpassing symptoms such as pain (34.3%) and malodor (43.4%). Burning sensations due to DD lesions were significantly correlated with the diagnosis of comorbid neuropsychiatric conditions (p = 0.047) and psychiatric medication use (p = 0.019). While pruritus correlated with disease severity and %BSA involvement, the findings were not statistically significant. Patients reporting pruritus had a significantly higher Dermatology Life Quality Index symptom score (2.4 ± 1.0), which is defined as the presence of itch, soreness, pain, or stinging, than those who did not (1.5 ± 0.6), indicating accurate symptom reporting. (4) Conclusions: In conclusion, a striking majority of patients with DD experience pruritus, with higher prevalence among those with neuropsychiatric challenges, severe Darier disease, and greater %BSA skin involvement. Clinicians should recognize pruritus as a key therapeutic target and adopt comprehensive treatment approaches that both address the neuropsychiatric comorbidities and the added psychological burden of pruritus in patients with DD.
Collapse
Affiliation(s)
- Grace Xiong
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada;
| | - Muskaan Sachdeva
- Division of Dermatology, Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Gil Yosipovitch
- Miami Itch Centre, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami, Leonard M. Miller School of Medicine, Coral Gables, FL 33136, USA;
| | - Michael Ziv
- Department of Dermatology, Emek Medical Centre, Rappaport Faculty of Medicine, Technion—Institute of Technology, Haifa 1834111, Israel
| | - Roni P. Dodiuk-Gad
- Division of Dermatology, Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Dermatology, Emek Medical Centre, Rappaport Faculty of Medicine, Technion—Institute of Technology, Haifa 1834111, Israel
| |
Collapse
|
3
|
Zhang L, Zhang C, Yan H, Han Y, Xu C, Liang J, Li R, Chen N, Liang W, Huang W, Xie G, Guo W. Changes in degree centrality and its associated genes: A longitudinal study of patients with schizophrenia undergoing pharmacological treatment. Schizophr Res 2025; 277:130-139. [PMID: 40058280 DOI: 10.1016/j.schres.2025.03.009] [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: 11/12/2024] [Revised: 01/07/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND The role of degree centrality (DC) in schizophrenia (SCZ), its trajectory following pharmacological treatment, and its potential as a prognostic biomarker and genetic mechanism remain unclear. METHODS We recruited 51 healthy controls (HC) and 56 patients with SCZ. Additionally, the SCZ patients underwent three months of antipsychotic medication treatment. We collected resting-state functional magnetic resonance imaging data, clinical variables, and conducted analyses using support vector machines, support vector regression, and gene expression correlation analysis. RESULTS Our study revealed that SCZ patients had generally reduced DC values in the cerebral cortex compared to HC at baseline, with increased DC values observed in the left occipital gyrus. After three months of treatment, SCZ patients exhibited a significant decrease in DC values in the left fusiform gyrus and an increase in the left inferior parietal gyrus. Variations in DC values in SCZ patients were associated with multiple genes, primarily enriched in molecular functions. CONCLUSION Changes in DC values in the right inferior occipital/fusiform gyrus and right calcarine/middle occipital gyrus may serve as neuroimaging markers to differentiate between HC and SCZ patients. Additionally, DC values in the left middle/postcentral gyrus could be used to predict treatment outcomes. Transcriptome-neuroimaging spatial correlation analysis provides valuable insights into the neurobiological mechanisms underlying SCZ pathology.
Collapse
Affiliation(s)
- Linna Zhang
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, Guangdong 528000, China
| | - Chunguo Zhang
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, Guangdong 528000, China
| | - Haohao Yan
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Yiding Han
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Caixia Xu
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, Guangdong 528000, China
| | - Jiaquan Liang
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, Guangdong 528000, China
| | - Runyi Li
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, Guangdong 528000, China
| | - Ningning Chen
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, Guangdong 528000, China
| | - Wenting Liang
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, Guangdong 528000, China
| | - Wei Huang
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, Guangdong 528000, China
| | - Guojun Xie
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, Guangdong 528000, China.
| | - Wenbin Guo
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
4
|
Rao ER, Thaxton T, Gama E, Godfrey J, Wei C, Lin Q, Li Y, Hejazi Pastor DP, Hansel C, Du X, Gomez CM. Calcium channel-coupled transcription factors facilitate direct nuclear signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.09.637126. [PMID: 39990342 PMCID: PMC11844367 DOI: 10.1101/2025.02.09.637126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
VGCCs play crucial roles within the CNS, in maintaining cell excitability, enabling activity- dependent neuronal development, and forming long-term memory by regulating Ca 2+ influx. The intracellular carboxyl-terminal domains of VGCC α1 subunits help regulate VGCC function. Emerging evidence suggests that some VGCC C-termini have functions independent of channel gating and exist as stable proteins. Here, we demonstrate that all VGCC gene family members express bicistronic mRNA transcripts that produce functionally distinct C-terminal proteins (CTPs) in tandem with full-length VGCC α1 subunits. Two of these CTPs, α1CCT and α1ACT, cycle to and from the nucleus in a Ca 2+ - and calmodulin-dependent fashion. α1CCT, α1ACT, and α1HCT regulate chromatin accessibility and/or bind directly to genes, regulating gene networks involved in neuronal differentiation and synaptic function in a Ca 2+ -dependent manner. This study elucidates a conserved process of coordinated protein expression within the VGCC family, coupling the channel function with VGCC C-terminal transcription factors.
Collapse
|
5
|
Onu CJ, Adu M, Chakkour M, Kumar V, Greenberg ML. Inositol Phosphates and Synthesizing Enzymes: Implications in Neurodegenerative Disorders. Biomolecules 2025; 15:225. [PMID: 40001529 PMCID: PMC11853280 DOI: 10.3390/biom15020225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Inositol is a vital sugar molecule involved in numerous signaling pathways required for cellular homeostasis and cell survival. Myo-inositol and its phospho-derivatives, inositol phosphates (IPs), are the most prevalent forms of inositol found in living cells. They are involved in regulating ion channels, metabolic flux, stress response, and other key biological processes. While emerging research has highlighted the significant roles of inositol phosphates in immunity, cancer, and metabolic diseases, there is a lack of comprehensive reviews on their roles in psychiatric and neurological disorders. This review aims to fill that gap by analyzing the existing literature on the importance of inositol phosphates in severe psychiatric and neurological conditions such as Parkinson's disease, Alzheimer's disease, bipolar disorder, amyotrophic lateral sclerosis, schizophrenia, and Huntington's disease, underscoring the potential to pave the way for new treatment regimens for these debilitating disorders targeting inositol pathways.
Collapse
Affiliation(s)
| | | | | | | | - Miriam L. Greenberg
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA (M.A.); (V.K.)
| |
Collapse
|
6
|
Sánchez-Florentino ZA, Romero-Martínez BS, Flores-Soto E, Montaño LM, Sommer B, Valdés-Tovar M, Argueta J, Calixto E, Aquino-Gálvez A, Castillejos-López M, Serrano H, Gomez-Verjan JC, López-Riquelme GO, Benítez-King GA, Jaimez R, Solís-Chagoyán H. Altered PLCβ/IP 3/Ca 2+ Signaling Pathway Activated by GPRCs in Olfactory Neuronal Precursor Cells Derived from Patients Diagnosed with Schizophrenia. Biomedicines 2024; 12:2343. [PMID: 39457654 PMCID: PMC11504003 DOI: 10.3390/biomedicines12102343] [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: 09/13/2024] [Revised: 10/08/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Schizophrenia (SZ) is a multifactorial chronic psychiatric disorder with a worldwide prevalence of 1%. Altered expression of PLCβ occurs in SZ patients, suggesting alterations in the PLCβ/IP3/Ca2+ signaling pathway. This cascade regulates critical cellular processes in all cell types, including the neuronal lineage; however, there is scarce evidence regarding the functionality of this transduction signaling in neuronal cells derived from SZ patients. Objective: We evaluated the functionality of the PLCβ/IP3/Ca2+ pathway in olfactory neuronal precursor cells (hONPCs) obtained from SZ patients. Methods: Cryopreserved hONPCs isolated from SZ patients and healthy subjects (HS) were thawed. The cellular types in subcultures were corroborated by immunodetection of the multipotency and lineage markers SOX-2, Musashi-1, nestin, and β-III tubulin. The PLCβ/IP3/Ca2+ pathway was activated by GPCR (Gq) ligands (ATP, UTP, serotonin, and epinephrine). In addition, PLCβ and IP3R were directly stimulated by perfusing cells with the activators m-3M3FBS and ADA, respectively. Cytosolic Ca2+ was measured by microfluorometry and by Ca2+ imaging. The amount and subcellular distribution of the PLCβ1 and PLCβ3 isoforms were evaluated by confocal immunofluorescence. IP3 concentration was measured by ELISA. Results: The results show that the increase of cytosolic Ca2+ triggered by GPCR ligands or directly through either PLCβ or IP3R activation was significantly lower in SZ-derived hONPCs, regarding HS-derived cells. Moreover, the relative amount of the PLCβ1 and PLCβ3 isoforms and IP3 production stimulated with m-3M3FBS were reduced in SZ-derived cells. Conclusions: Our results suggest an overall functional impairment in the PLCβ/IP3/Ca2+ signaling pathway in SZ-derived hONPCs.
Collapse
Affiliation(s)
- Zuly A. Sánchez-Florentino
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, CP, Mexico;
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, CP, Mexico; (J.A.); (G.A.B.-K.)
| | - Bianca S. Romero-Martínez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, CP, Mexico; (B.S.R.-M.); (E.F.-S.); (L.M.M.)
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, CP, Mexico; (B.S.R.-M.); (E.F.-S.); (L.M.M.)
| | - Luis M. Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, CP, Mexico; (B.S.R.-M.); (E.F.-S.); (L.M.M.)
| | - Bettina Sommer
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City 14080, CP, Mexico;
| | - Marcela Valdés-Tovar
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, CP, Mexico;
| | - Jesús Argueta
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, CP, Mexico; (J.A.); (G.A.B.-K.)
| | - Eduardo Calixto
- Departamento de Neurobiología, Dirección de Investigación en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, CP, Mexico;
| | - Arnoldo Aquino-Gálvez
- Laboratorio de Biología Molecular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City 14080, CP, Mexico;
| | - Manuel Castillejos-López
- Unidad de Epidemiología Hospitalaria e Infectología, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City 14080, CP, Mexico;
| | - Héctor Serrano
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, CP, Mexico;
| | - Juan C. Gomez-Verjan
- Dirección de Investigación, Instituto Nacional de Geriatría, Mexico City 10200, CP, Mexico;
| | - Germán O. López-Riquelme
- Laboratorio de Socioneurobiologia, Centro de Investigación en Ciencias Cognitivas, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, CP, Mexico;
| | - Gloria A. Benítez-King
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, CP, Mexico; (J.A.); (G.A.B.-K.)
| | - Ruth Jaimez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, CP, Mexico; (B.S.R.-M.); (E.F.-S.); (L.M.M.)
| | - Héctor Solís-Chagoyán
- Laboratorio de Neurobiología Cognitiva, Centro de Investigación en Ciencias Cognitivas, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, CP, Mexico
| |
Collapse
|
7
|
Meng L, Ouyang Z, Chen Y, Huang C, Yu Y, Fan R. Low-dose BPA-induced neuronal energy metabolism dysfunction and apoptosis mediated by PINK1/parkin mitophagy pathway in juvenile rats. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 929:172655. [PMID: 38653419 DOI: 10.1016/j.scitotenv.2024.172655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Bisphenol A (BPA) is related to neurological disorders involving mitochondrial dysfunction, while the mechanism remains elusive. Therefore, we explored it through in vitro and in vivo experiments. In vitro, hippocampal neurons derived from neonatal rats of different genders were exposed to 1-100 nM and 100 μM BPA, autophagy activator Rapa and inhibitor 3-MA for 7 d. The results suggested that even nanomolar BPA (1-100 nM) disturbed Ca2+ homeostasis and damaged the integrity of mitochondrial cristae in neurons (p < 0.05). Furthermore, BPA increased the number of autophagic lysosomes, LC3II/LC3I ratio, and p62 expression, and decreased parkin expression (p < 0.05), suggesting that the entry of damaged mitochondria into autophagic pathway was prompted, while the autophagic degradation pathway was blocked. This further disrupts neuronal energy metabolism and promotes neuronal apoptosis. However, Rapa attenuated the adverse effects caused by BPA, while 3-MA exacerbated these reactions. In vivo, exposure of juvenile rats to 0.5, 50, 5000 μg/kg‧bw/day BPA during PND 7-21 markedly impaired the structure of hippocampal mitochondria, increased the number of autophagosomes, the rate of neuronal apoptosis, and the expression levels of pro-apoptotic proteins Cyt C, Bax, Bak1, and Caspase3, and decreased the expression of anti-apoptotic protein Bcl2 (p < 0.05). Particularly, male rats are more sensitive to low-dose BPA than females. Overall, environmental-doses BPA can induce the imbalance of energy metabolism in hippocampal neurons via PINK1/parkin mitophagy, thereby inducing their apoptosis. Importantly, this study provides a theoretical basis for attenuating BPA-related neurological diseases.
Collapse
Affiliation(s)
- Lingxue Meng
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China; State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou 510655, China
| | - Zedong Ouyang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Yuxin Chen
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Chengmeng Huang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Yunjiang Yu
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou 510655, China
| | - Ruifang Fan
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
8
|
Huang Z, Zhang L, Li Y, Yu Y, Shen Y, Sun X, Lou K, Luo H, Meng Z, Li H, Wei Y. Population Pharmacodynamic Models of Risperidone on PANSS Total Scores and Prolactin Levels in Schizophrenia. Pharmaceuticals (Basel) 2024; 17:148. [PMID: 38399363 PMCID: PMC10891722 DOI: 10.3390/ph17020148] [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: 12/20/2023] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
Currently, research predominantly focuses on evaluating clinical effects at specific time points while neglecting underlying patterns within the treatment process. This study aims to analyze the dynamic alterations in PANSS total scores and prolactin levels in patients with schizophrenia treated with risperidone, along with the influencing covariates. Using data from an 8-week randomized, double-blind, multicenter clinical trial, a population pharmacodynamic model was established for the PANSS total scores of and prolactin levels in patients treated with risperidone. The base model employed was the Emax model. Covariate selection was conducted using a stepwise forward inclusion and backward elimination approach. A total of 144 patients were included in this analysis, with 807 PANSS total scores and 531 prolactin concentration values. The PANSS total scores of the patients treated with risperidone decreased over time, fitting a proportionally parameterized sigmoid Emax model with covariates including baseline score, course of the disease, gender, plasma calcium ions, and lactate dehydrogenase levels. The increase in prolactin levels conformed to the ordinary Emax model, with covariates encompassing course of the disease, gender, weight, red blood cell count, and triglyceride levels. The impacts of the baseline scores and the course of the disease on the reduction of the PANSS scores, as well as the influence of gender on the elevation of prolactin levels, each exceeded 20%. This study provides valuable quantitative data regarding PANSS total scores and prolactin levels among patients undergoing risperidone treatment across various physiological conditions.
Collapse
Affiliation(s)
- Zhiwei Huang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
| | - Lei Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
| | - Yan Li
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
| | - Yimin Yu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
| | - Yifeng Shen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
| | - Xiujia Sun
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
| | - Kun Lou
- CSPC Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., Ltd., Shijiazhuang 050000, China
| | - Hongmei Luo
- CSPC Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., Ltd., Shijiazhuang 050000, China
| | - Zhibin Meng
- CSPC Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., Ltd., Shijiazhuang 050000, China
| | - Huafang Li
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
- Shanghai Clinical Research Center for Mental Health, Shanghai 200030, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, China
| | - Yumei Wei
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (Z.H.)
| |
Collapse
|
9
|
Punchaichira TJ, Kukshal P, Bhatia T, Deshpande SN, Thelma BK. Effect of rs1108580 of DBH and rs1006737 of CACNA1C on Cognition and Tardive Dyskinesia in a North Indian Schizophrenia Cohort. Mol Neurobiol 2023; 60:6826-6839. [PMID: 37493923 DOI: 10.1007/s12035-023-03496-4] [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: 04/15/2023] [Accepted: 07/10/2023] [Indexed: 07/27/2023]
Abstract
Genetic perturbations in dopamine neurotransmission and calcium signaling pathways are implicated in the etiology of schizophrenia. We aimed to test the association of a functional splice variant each in Dopamine β-Hydroxylase (DBH; rs1108580) and Calcium voltage-gated channel subunit alpha1 C (CACNA1C; rs1006737) genes in these pathways with schizophrenia (506 cases, 443 controls); Abnormal Involuntary Movement Scale (AIMS) scores in subjects assessed for tardive dyskinesia (76 TD-positive, 95 TD-negative) and Penn Computerized Neurocognitive Battery (PennCNB) scores (334 cases, 234 controls). The effect of smoking status and SNP genotypes on AIMS scores were assessed using ANOVA; health status and SNP genotypes on three performance functions of PennCNB cognitive domains were assessed by ANCOVA with age and sex as covariates. Association with Positive and Negative Syndrome Scale (PANSS) scores in the TD cohort and cognitive scores in healthy controls of the cognition cohort were tested by linear regression. None of the markers were associated with schizophrenia. Smoking status [F(2, 139) = 10.6; p = 5 × 10-5], rs1006737 [F(2, 139) = 7.1; p = 0.001], TD status*smoking [F(2, 139) = 8.0; p = 5.0 × 10-4] and smoking status*rs1006737 [F(4, 139) = 2.7; p = 0.03] had an effect on AIMS score. Furthermore, rs1006737 was associated with orofacial [F(2, 139) = 4.6; p = 0.01] and limb-truncal TD [(F(2, 139) = 3.8; p = 0.02]. Main effect of rs1108580 on working memoryprocessing speed [F(2, 544) = 3.8; p = 0.03] and rs1006737 on spatial abilityefficiency [F(1, 550) = 9.4; p = 0.02] was identified. Health status*rs1006737 interaction had an effect on spatial memoryprocessing speed [F(1, 550) = 6.9; p = 0.01]. Allelic/genotypic association (p = 0.01/0.03) of rs1006737 with disorganized/concrete factor and allelic association of rs1108580 (p = 0.04) with a depressive factor of PANSS was observed in the TD-negative subcohort. Allelic association of rs1006737 with sensorimotor dexterityaccuracy (p = 0.03), attentionefficiency (p = 0.05), and spatial abilityefficiency (p = 0.02); allelic association of rs1108580 with face memoryaccuracy (p = 0.05) and emotionefficiency (p = 0.05); and allelic/genotypic association with emotionaccuracy (p = 0.003/0.009) were observed in healthy controls of the cognition cohort. These association findings may have direct implications for personalized medicine and cognitive remediation.
Collapse
Affiliation(s)
| | - Prachi Kukshal
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India
- Sri Sathya Sai Sanjeevani International Centre for Child Heart Care & Research, Palwal, Haryana, 121102, India
| | - Triptish Bhatia
- Department of Psychiatry, Postgraduate Institute of Medical Education and Research-Dr. Ram Manohar Lohia Hospital, Baba Kharak Singh Marg, Connaught Place, New Delhi, 110001, India
| | - Smita Neelkanth Deshpande
- Department of Psychiatry, Postgraduate Institute of Medical Education and Research-Dr. Ram Manohar Lohia Hospital, Baba Kharak Singh Marg, Connaught Place, New Delhi, 110001, India
| | - B K Thelma
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India.
| |
Collapse
|
10
|
Baj J, Bargieł J, Cabaj J, Skierkowski B, Hunek G, Portincasa P, Flieger J, Smoleń A. Trace Elements Levels in Major Depressive Disorder-Evaluation of Potential Threats and Possible Therapeutic Approaches. Int J Mol Sci 2023; 24:15071. [PMID: 37894749 PMCID: PMC10606638 DOI: 10.3390/ijms242015071] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
The multifactorial etiology of major depressive disorder (MDD) includes biological, environmental, genetic, and psychological aspects. Recently, there has been an increasing interest in metallomic studies in psychiatry, aiming to evaluate the role of chosen trace elements in the MDD etiology as well as the progression of symptoms. This narrative review aims to summarize the available literature on the relationship between the concentration of chosen elements in the serum of patients with MDD and the onset and progression of this psychiatric condition. The authors reviewed PubMed, Web of Science, and Scopus databases searching for elements that had been investigated so far and further evaluated them in this paper. Ultimately, 15 elements were evaluated, namely, zinc, magnesium, selenium, iron, copper, aluminium, cadmium, lead, mercury, arsenic, calcium, manganese, chromium, nickel, and phosphorus. The association between metallomic studies and psychiatry has been developing dynamically recently. According to the results of current research, metallomics might act as a potential screening tool for patients with MDD while at the same time providing an assessment of the severity of symptoms. Either deficiencies or excessive amounts of chosen elements might be associated with the progression of depressive symptoms or even the onset of the disease among people predisposed to MDD.
Collapse
Affiliation(s)
- Jacek Baj
- Department of Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| | - Julia Bargieł
- Student Research Group of Department of Epidemiology and Clinical Research Methodology, Medical University of Lublin, Radziwiłłowska 11, 20-080 Lublin, Poland; (J.B.); (J.C.); (B.S.)
| | - Justyna Cabaj
- Student Research Group of Department of Epidemiology and Clinical Research Methodology, Medical University of Lublin, Radziwiłłowska 11, 20-080 Lublin, Poland; (J.B.); (J.C.); (B.S.)
| | - Bartosz Skierkowski
- Student Research Group of Department of Epidemiology and Clinical Research Methodology, Medical University of Lublin, Radziwiłłowska 11, 20-080 Lublin, Poland; (J.B.); (J.C.); (B.S.)
| | - Gabriela Hunek
- Student Research Group of Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland;
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy;
| | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland;
| | - Agata Smoleń
- Department of Epidemiology and Clinical Research Methodology, Medical University of Lublin, 20-080 Lublin, Poland;
| |
Collapse
|
11
|
Case KC, Beltman RJ, Pflum MKH, Greenberg ML. Valproate regulates inositol synthesis by reducing expression of myo-inositol-3-phosphate synthase. Sci Rep 2023; 13:14844. [PMID: 37684289 PMCID: PMC10491628 DOI: 10.1038/s41598-023-41936-2] [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: 03/25/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023] Open
Abstract
Inositol depletion is a hypothesized mechanism of action of mood stabilization drugs used in the treatment of bipolar disorder. It was previously reported that the mood stabilizer valproate (VPA) increased phosphorylation of myo-inositol-3-phosphate synthases (MIPS), the rate limiting enzyme of inositol synthesis. Phosphosites were identified and examination of site-directed mutants suggested that phosphorylation leads to decreased enzymatic activity. In this study, we examined the extent of MIPS phosphorylation in response to VPA and used two interaction screens to identify protein kinases that interact with MIPS. Using an epitope tagged MIPS construct, we determined the fraction of phosphorylated MIPS to be very low (less than 2% of total), and we could not detect phosphorylation of untagged MIPS in response to VPA. In vitro analyses of phosphorylation revealed that putative protein kinases, PKC and CKII, have low specificity toward MIPS. These findings suggest that VPA likely depletes inositol via a mechanism other than MIPS phosphorylation. Consistent with this, mRNA levels of the MIPS-encoding gene INO1 and MIPS protein levels were significantly reduced during the mid-log growth phase in response to VPA treatment. These findings suggest that the mechanism whereby VPA causes inositol depletion is by reducing expression of the rate-limiting enzyme MIPS.
Collapse
Affiliation(s)
- Kendall C Case
- Department of Biological Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Rachel J Beltman
- Department of Chemistry, Wayne State University, Detroit, MI, 48202, USA
| | - Mary Kay H Pflum
- Department of Chemistry, Wayne State University, Detroit, MI, 48202, USA
| | - Miriam L Greenberg
- Department of Biological Sciences, Wayne State University, Detroit, MI, 48202, USA.
| |
Collapse
|
12
|
Machado-Vieira R, Courtes AC, Zarate CA, Henter ID, Manji HK. Non-canonical pathways in the pathophysiology and therapeutics of bipolar disorder. Front Neurosci 2023; 17:1228455. [PMID: 37592949 PMCID: PMC10427509 DOI: 10.3389/fnins.2023.1228455] [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: 05/24/2023] [Accepted: 07/17/2023] [Indexed: 08/19/2023] Open
Abstract
Bipolar disorder (BD) is characterized by extreme mood swings ranging from manic/hypomanic to depressive episodes. The severity, duration, and frequency of these episodes can vary widely between individuals, significantly impacting quality of life. Individuals with BD spend almost half their lives experiencing mood symptoms, especially depression, as well as associated clinical dimensions such as anhedonia, fatigue, suicidality, anxiety, and neurovegetative symptoms. Persistent mood symptoms have been associated with premature mortality, accelerated aging, and elevated prevalence of treatment-resistant depression. Recent efforts have expanded our understanding of the neurobiology of BD and the downstream targets that may help track clinical outcomes and drug development. However, as a polygenic disorder, the neurobiology of BD is complex and involves biological changes in several organelles and downstream targets (pre-, post-, and extra-synaptic), including mitochondrial dysfunction, oxidative stress, altered monoaminergic and glutamatergic systems, lower neurotrophic factor levels, and changes in immune-inflammatory systems. The field has thus moved toward identifying more precise neurobiological targets that, in turn, may help develop personalized approaches and more reliable biomarkers for treatment prediction. Diverse pharmacological and non-pharmacological approaches targeting neurobiological pathways other than neurotransmission have also been tested in mood disorders. This article reviews different neurobiological targets and pathophysiological findings in non-canonical pathways in BD that may offer opportunities to support drug development and identify new, clinically relevant biological mechanisms. These include: neuroinflammation; mitochondrial function; calcium channels; oxidative stress; the glycogen synthase kinase-3 (GSK3) pathway; protein kinase C (PKC); brain-derived neurotrophic factor (BDNF); histone deacetylase (HDAC); and the purinergic signaling pathway.
Collapse
Affiliation(s)
- Rodrigo Machado-Vieira
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center, Houston, TX, United States
| | - Alan C. Courtes
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center, Houston, TX, United States
| | - Carlos A. Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Ioline D. Henter
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Husseini K. Manji
- Deparment of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
13
|
Zhang Z, Lin H, Feng Z, Xie H, Liu P, Shu Y, Jia Z, Zhang S. Impaired calcium channel function and pronounced hippocampal atrophy in a schizophrenia patient with cognitive impairment carrying Presenilin-2 Ser130Leu mutation: A case report and literature review. Schizophr Res 2023; 258:78-80. [PMID: 37517367 DOI: 10.1016/j.schres.2023.07.024] [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: 05/16/2023] [Revised: 07/12/2023] [Accepted: 07/23/2023] [Indexed: 08/01/2023]
Affiliation(s)
- Ziyi Zhang
- Department of Neurology, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu 610041, Sichuan Province, PR China
| | - Hua Lin
- Department of Nuclear Medicine, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu 610041, Sichuan Province, PR China
| | - Zijuan Feng
- Department of Neurology, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu 610041, Sichuan Province, PR China
| | - Hongsheng Xie
- Department of Nuclear Medicine, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu 610041, Sichuan Province, PR China
| | - Peng Liu
- Department of Emergency, West China Hospital of Sichuan University, No. 37 Guo Xue Xiang, Chengdu 610041, Sichuan Province, PR China
| | - Yang Shu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Zhiyun Jia
- Department of Nuclear Medicine, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu 610041, Sichuan Province, PR China.
| | - Shuting Zhang
- Department of Neurology, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu 610041, Sichuan Province, PR China.
| |
Collapse
|
14
|
Xu K, Zheng P, Zhao S, Feng J, Pu J, Wang J, Zhao S, Wang H, Chen J, Xie P. Altered MANF and RYR2 concentrations associated with hypolipidemia in the serum of patients with schizophrenia. J Psychiatr Res 2023; 163:142-149. [PMID: 37210832 DOI: 10.1016/j.jpsychires.2023.05.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/12/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023]
Abstract
Schizophrenia (SCZ) is associated with abnormal serum lipid profiles, but their relationship is poorly understood. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an important regulator of lipid metabolism. Previous studies have shown its involvement in the pathogenesis of numerous neuropsychiatric disorders, while its role in SCZ is still unknown. Therefore, this study was conducted to examine serum MANF levels in patients with SCZ, and to investigate the potential relationship between MANF, serum lipid levels and SCZ. The results showed that total cholesterol (TC) levels were significantly lower in 225 patients with SCZ than in 233 healthy controls (HCs). According to Ingenuity Pathway Analysis, hypolipidemia is associated with SCZ via MANF/ryanodine receptor 2 (RYR2) pathway. This theory was supported by another sample set, which showed significantly lower MANF levels and higher RYR2 levels in the serum of 170 SCZ patients compared to 80 HCs. Moreover, MANF and RYR2 levels both were significantly correlated with the severity of psychotic symptoms and TC levels. In addition, a model consisting of MANF and RYR2 was found to be effective in distinguishing SCZ patients from HCs. These findings suggested that the MANF/RYR2 pathway might serve as a bridge between hypolipidemia and SCZ, and MANF and RYR2 held promise as biomarkers for SCZ.
Collapse
Affiliation(s)
- Ke Xu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Peng Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuang Zhao
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Jinzhou Feng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Juncai Pu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiubing Wang
- Department of Clinical Laboratory, Chongqing Mental Health Centre, Chongqing, China
| | - Shuqian Zhao
- Department of Clinical Psychology, Chongqing Mental Health Centre, Chongqing, China
| | - Haiyang Wang
- National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Key Laboratory of Psychoseomadsy, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Jianjun Chen
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.
| | - Peng Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
15
|
Zhang J, Wang X, Duan H, Chen C, Lu Z, Zhang D, Li S. The Association of Calcium Signaling Pathway Gene Variants, Bone Mineral Density and Mild Cognitive Impairment in Elderly People. Genes (Basel) 2023; 14:genes14040828. [PMID: 37107586 PMCID: PMC10137633 DOI: 10.3390/genes14040828] [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: 02/22/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
The association of calcium signaling pathway gene variants, bone mineral density (BMD) and mild cognitive impairment (MCI) is poorly understood so far. A total of 878 participants from Qingdao city were recruited in this study. According to the candidate gene selection method, 58 single nucleotide polymorphisms (SNPs) in eight calcium signaling genes were selected. The association between gene polymorphisms and MCI was revealed by using multiple genetic models. Polygenic risk scores (PRS) were used to summarize the effects of the whole gene. Logistic regression was used to analyze the association between each PRS and MCI. The multiplicative interaction term in the regression models was used to estimate the interaction effects between the PRS and BMD. We observed significant associations of rs6877893 (NR3C1), rs6448456 (CCKAR), and rs723672 (CACNA1C) polymorphisms with MCI. The PRSs of NR3C1 (OR = 4.012, 95% CI = 1.722-9.347, p < 0.001), PRKCA (OR = 1.414, 95% CI = 1.083-1.845, p = 0.011) and TRPM1 (OR = 3.253, 95% CI = 1.116-9.484, p = 0.031) were associated with an increased risk of developing MCI, and the PRS of total genes (OR = 0.330, 95% CI = 0.224-0.485, p < 0.001) was associated with a decreased risk of developing MCI. In interaction effect analysis, the interaction effect of PRKCA and BMD was significant. Genetic variations of the calcium signaling pathway were associated with MCI in older people. There was an interaction effect between PRKCA gene variants and BMD on MCI.
Collapse
Affiliation(s)
- Jiesong Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266012, China
| | - Xueyan Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266012, China
| | - Haiping Duan
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao 266033, China
| | - Chen Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266012, China
| | - Zhonghai Lu
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266012, China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266012, China
| | - Suyun Li
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266012, China
| |
Collapse
|
16
|
Li Q, Xu X, Qian Y, Cai H, Zhao W, Zhu J, Yu Y. Resting-state brain functional alterations and their genetic mechanisms in drug-naive first-episode psychosis. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2023; 9:13. [PMID: 36841861 PMCID: PMC9968350 DOI: 10.1038/s41537-023-00338-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/07/2023] [Indexed: 02/27/2023]
Abstract
Extensive research has established the presence of resting-state brain functional damage in psychosis. However, the genetic mechanisms of such disease phenotype are yet to be unveiled. We investigated resting-state brain functional alterations in patients with drug-naive first-episode psychosis (DFP) by performing a neuroimaging meta-analysis of 8 original studies comprising 500 patients and 469 controls. Combined with the Allen Human Brain Atlas, we further conducted transcriptome-neuroimaging spatial correlations to identify genes whose expression levels were linked to brain functional alterations in DFP, followed by a range of gene functional characteristic analyses. Meta-analysis revealed a mixture of increased and decreased brain function in widespread areas including the default-mode, visual, motor, striatal, and cerebellar systems in DFP. Moreover, these brain functional alterations were spatially associated with the expression of 1662 genes, which were enriched for molecular functions, cellular components, and biological processes of the cerebral cortex, as well as psychiatric disorders including schizophrenia. Specific expression analyses demonstrated that these genes were specifically expressed in the brain tissue, in cortical neurons and immune cells, and during nearly all developmental periods. Concurrently, the genes could construct a protein-protein interaction network supported by hub genes and were linked to multiple behavioral domains including emotion, attention, perception, and motor. Our findings provide empirical evidence for the notion that brain functional damage in DFP involves a complex interaction of polygenes with various functional characteristics.
Collapse
Affiliation(s)
- Qian Li
- grid.459419.4Department of Radiology, Chaohu Hospital of Anhui Medical University, 238000 Hefei, China ,grid.412679.f0000 0004 1771 3402Department of Radiology, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, China ,Research Center of Clinical Medical Imaging, Anhui Province, 230032 Hefei, China ,Anhui Provincial Institute of Translational Medicine, 230032 Hefei, China
| | - Xiaotao Xu
- grid.412679.f0000 0004 1771 3402Department of Radiology, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, China ,Research Center of Clinical Medical Imaging, Anhui Province, 230032 Hefei, China ,Anhui Provincial Institute of Translational Medicine, 230032 Hefei, China
| | - Yinfeng Qian
- grid.412679.f0000 0004 1771 3402Department of Radiology, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, China ,Research Center of Clinical Medical Imaging, Anhui Province, 230032 Hefei, China ,Anhui Provincial Institute of Translational Medicine, 230032 Hefei, China
| | - Huanhuan Cai
- grid.412679.f0000 0004 1771 3402Department of Radiology, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, China ,Research Center of Clinical Medical Imaging, Anhui Province, 230032 Hefei, China ,Anhui Provincial Institute of Translational Medicine, 230032 Hefei, China
| | - Wenming Zhao
- grid.412679.f0000 0004 1771 3402Department of Radiology, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, China ,Research Center of Clinical Medical Imaging, Anhui Province, 230032 Hefei, China ,Anhui Provincial Institute of Translational Medicine, 230032 Hefei, China
| | - Jiajia Zhu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China. .,Research Center of Clinical Medical Imaging, Anhui Province, 230032, Hefei, China. .,Anhui Provincial Institute of Translational Medicine, 230032, Hefei, China.
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China. .,Research Center of Clinical Medical Imaging, Anhui Province, 230032, Hefei, China. .,Anhui Provincial Institute of Translational Medicine, 230032, Hefei, China.
| |
Collapse
|
17
|
Xiang S, Wang R, Hua L, Song J, Qian S, Jin Y, Zhang B, Ding X. Assessment of Bidirectional Relationships between Mental Illness and Rheumatoid Arthritis: A Two-Sample Mendelian Randomization Study. J Clin Med 2023; 12:944. [PMID: 36769592 PMCID: PMC9917759 DOI: 10.3390/jcm12030944] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
A correlation between mental illness and systemic rheumatoid arthritis (RA) has been observed in several prior investigations. However, little is known about the causative relationship between them. The present study aimed to systematically investigate the potential association between genetically determined mental illness and RA. Two-sample bidirectional Mendelian randomization (MR) analysis was performed using publicly released genome-wide association studies (GWAS). We selected independent genetic variants associated with four mental illnesses (bipolar disorder, broad depression, major depression, and anxiety) as instrumental variables. The inverse variance weighted (IVW) method was used as the primary analysis to assess the causal relationship between mental illness and RA. Results of the IVW analysis suggested that genetic predisposition to bipolar disorder was associated with a decreased risk of RA (odds ratio [OR] = 0.825, 95% CI = 0.716 to 0.95, p = 0.007). Furthermore, we did not find a significant causal effect of RA on bipolar disorder in the reverse MR analysis (p > 0.05). In addition, our study found no evidence of a bidirectional causal relationship between genetically predicted broad depression, major depression, anxiety, and RA (p > 0.05). The genetically proxied bipolar disorder population has a lower RA risk, which may indicate that there is a hidden mechanism for inhibiting the pathogenesis of RA in bipolar disorder. However, results do not support a causal connection between depression, anxiety, and RA.
Collapse
Affiliation(s)
- Shate Xiang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Rongyun Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Lijiangshan Hua
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jie Song
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Suhai Qian
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yibo Jin
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Bingyue Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xinghong Ding
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
18
|
Dashti S, Nahavandi A. Neuroprotective effects of aripiprazole in stress-induced depressive-like behavior: Possible role of CACNA1C. J Chem Neuroanat 2022; 126:102170. [PMID: 36270562 DOI: 10.1016/j.jchemneu.2022.102170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/07/2022] [Accepted: 10/15/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Depression is the most common psychiatric disorder. Recently, aripiprazole, a novel antipsychotic drug, has been approved as the adjunctive therapy for the Treatment-Resistant Depression (TRD). However, the mechanisms underlying the antidepressant effects of aripiprazole are not fully known. Besides the involvement of calcium signaling dysregulations in the pathophysiology of depression, there is some evidence of overexpressed CACNA1C (the gene encoding the Cav1.2 channels) following chronic stress in the brain regions, which involved in emotional and stress responses. Based on the data indicating the aripiprazole's effects on intracellular calcium levels, this study aimed to investigate the mechanisms of therapeutic effects of aripiprazole, by a focus on the modulation of CACNA1C expression, in the rat stress-induced model of depression. METHODS Using Chronic Unpredictable Mild Stress (CUMS) model of depression, we examined the effects of aripiprazole on depressive and anxiety-like behaviors (by forced swimming test and elevated plus maze), serum IL-6 (Elisa), and cell survival (Nissl staining). In addition, CACNA1C, BDNF, and TrkB expression in the PFC and hippocampus (RT-qPCR), as well as BDNF and GAP-43 protein levels in the hippocampus (Immunohistofluorescence), have been assayed. RESULTS Our data indicated that aripiprazole could improve anxiety and depressive-like behaviors, decrease the serum levels of IL-6 and hippocampal cell death following CUMS. In addition, we showed the significant modulation on overexpressed CACNA1C, as well as downregulated BDNF and GAP-43 expression DISCUSSION: These results demonstrate that aripiprazole may promote synaptic plasticity by improving the expression of BDNF and gap-43. In addition, inflammation reduction and CACNA1C expression downregulation may be some of mechanisms by which aripiprazole alleviates chronic stress-induced hippocampal cell death and play its pivotal antidepressant role.
Collapse
Affiliation(s)
- Somayeh Dashti
- Department of Physiology, Faculty of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Arezo Nahavandi
- Department of Physiology, Faculty of Medicine, Iran University of Medical Science, Tehran, Iran; Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Science, Tehran, Iran; Neuroscience Research Center, Iran University of Medical Science, Tehran, Iran.
| |
Collapse
|
19
|
Harrison PJ, Husain SM, Lee H, Los Angeles AD, Colbourne L, Mould A, Hall NAL, Haerty W, Tunbridge EM. CACNA1C (Ca V1.2) and other L-type calcium channels in the pathophysiology and treatment of psychiatric disorders: Advances from functional genomics and pharmacoepidemiology. Neuropharmacology 2022; 220:109262. [PMID: 36154842 DOI: 10.1016/j.neuropharm.2022.109262] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/09/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022]
Abstract
A role for voltage-gated calcium channels (VGCCs) in psychiatric disorders has long been postulated as part of a broader involvement of intracellular calcium signalling. However, the data were inconclusive and hard to interpret. We review three areas of research that have markedly advanced the field. First, there is now robust genomic evidence that common variants in VGCC subunit genes, notably CACNA1C which encodes the L-type calcium channel (LTCC) CaV1.2 subunit, are trans-diagnostically associated with psychiatric disorders including schizophrenia and bipolar disorder. Rare variants in these genes also contribute to the risk. Second, pharmacoepidemiological evidence supports the possibility that calcium channel blockers, which target LTCCs, might have beneficial effects on the onset or course of these disorders. This is especially true for calcium channel blockers that are brain penetrant. Third, long-range sequencing is revealing the repertoire of full-length LTCC transcript isoforms. Many novel and abundant CACNA1C isoforms have been identified in human and mouse brain, including some which are enriched compared to heart or aorta, and predicted to encode channels with differing functional and pharmacological properties. These isoforms may contribute to the molecular mechanisms of genetic association to psychiatric disorders. They may also enable development of therapeutic agents that can preferentially target brain LTCC isoforms and be of potential value for psychiatric indications.
Collapse
Affiliation(s)
- Paul J Harrison
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK.
| | - Syed M Husain
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Hami Lee
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK
| | | | - Lucy Colbourne
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Arne Mould
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Nicola A L Hall
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Wilfried Haerty
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, UK; School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Elizabeth M Tunbridge
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK
| |
Collapse
|
20
|
Pallier PN, Ferrara M, Romagnolo F, Ferretti MT, Soreq H, Cerase A. Chromosomal and environmental contributions to sex differences in the vulnerability to neurological and neuropsychiatric disorders: Implications for therapeutic interventions. Prog Neurobiol 2022; 219:102353. [PMID: 36100191 DOI: 10.1016/j.pneurobio.2022.102353] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/22/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
Abstract
Neurological and neuropsychiatric disorders affect men and women differently. Multiple sclerosis, Alzheimer's disease, anxiety disorders, depression, meningiomas and late-onset schizophrenia affect women more frequently than men. By contrast, Parkinson's disease, autism spectrum condition, attention-deficit hyperactivity disorder, Tourette's syndrome, amyotrophic lateral sclerosis and early-onset schizophrenia are more prevalent in men. Women have been historically under-recruited or excluded from clinical trials, and most basic research uses male rodent cells or animals as disease models, rarely studying both sexes and factoring sex as a potential source of variation, resulting in a poor understanding of the underlying biological reasons for sex and gender differences in the development of such diseases. Putative pathophysiological contributors include hormones and epigenetics regulators but additional biological and non-biological influences may be at play. We review here the evidence for the underpinning role of the sex chromosome complement, X chromosome inactivation, and environmental and epigenetic regulators in sex differences in the vulnerability to brain disease. We conclude that there is a pressing need for a better understanding of the genetic, epigenetic and environmental mechanisms sustaining sex differences in such diseases, which is critical for developing a precision medicine approach based on sex-tailored prevention and treatment.
Collapse
Affiliation(s)
- Patrick N Pallier
- Blizard Institute, Centre for Neuroscience, Surgery and Trauma, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK.
| | - Maria Ferrara
- Institute of Psychiatry, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy; Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States; Women's Brain Project (WBP), Switzerland
| | - Francesca Romagnolo
- Institute of Psychiatry, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | | | - Hermona Soreq
- The Edmond and Lily Safra Center of Brain Science, The Hebrew University of Jerusalem, 9190401, Israel
| | - Andrea Cerase
- EMBL-Rome, Via Ramarini 32, 00015 Monterotondo, RM, Italy; Blizard Institute, Centre for Genomics and Child Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; Department of Biology, University of Pisa, SS12 Abetone e Brennero 4, 56127 Pisa, Italy.
| |
Collapse
|
21
|
Lazcano P, Schmidtke MW, Onu C, Greenberg ML. Phosphatidic acid inhibits inositol synthesis by inducing nuclear translocation of kinase IP6K1 and repression of myo-inositol-3-P synthase. J Biol Chem 2022; 298:102363. [PMID: 35963434 PMCID: PMC9478396 DOI: 10.1016/j.jbc.2022.102363] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 11/17/2022] Open
Abstract
Inositol is an essential metabolite that serves as a precursor for structural and signaling molecules. Although perturbation of inositol homeostasis has been implicated in numerous human disorders, surprisingly little is known about how inositol levels are regulated in mammalian cells. A recent study in mouse embryonic fibroblasts demonstrated that nuclear translocation of inositol hexakisphosphate kinase 1 (IP6K1) mediates repression of myo-inositol-3-P synthase (MIPS), the rate-limiting inositol biosynthetic enzyme. Binding of IP6K1 to phosphatidic acid (PA) is required for this repression. Here, we elucidate the role of PA in IP6K1 repression. Our results indicate that increasing PA levels through pharmacological stimulation of phospholipase D (PLD) or direct supplementation of 18:1 PA induces nuclear translocation of IP6K1 and represses expression of the MIPS protein. We found that this effect was specific to PA synthesized in the plasma membrane, as endoplasmic reticulum–derived PA did not induce IP6K1 translocation. Furthermore, we determined that PLD-mediated PA synthesis can be stimulated by the master metabolic regulator 5′ AMP-activated protein kinase (AMPK). We show that activation of AMPK by glucose deprivation or by treatment with the mood-stabilizing drugs valproate or lithium recapitulated IP6K1 nuclear translocation and decreased MIPS expression. This study demonstrates for the first time that modulation of PA levels through the AMPK-PLD pathway regulates IP6K1-mediated repression of MIPS.
Collapse
Affiliation(s)
- Pablo Lazcano
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Michael W Schmidtke
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Chisom Onu
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Miriam L Greenberg
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States.
| |
Collapse
|
22
|
Colijn MA, Hrynchak M, Hrazdil CT, Willaeys V, White RF, Stowe RM. A 1p31.3 deletion encompassing the nuclear factor 1A gene presenting as possible temporal lobe epilepsy in association with schizoaffective disorder. Neurocase 2022; 28:382-387. [PMID: 36209511 DOI: 10.1080/13554794.2022.2132869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/10/2022]
Abstract
Chromosome 1p32-p31 deletion syndrome, which is characterized by a variety of neurodevelopmental abnormalities, is thought to occur as a result of nuclear factor 1A (NFIA) haploinsufficiency. We present a case of a right-handed 40-year-old female with a 1p31.3 deletion, who exhibited numerous common features of this syndrome, in addition to treatment resistant schizoaffective disorder and possible temporal lobe epilepsy, making her presentation unique. While neither psychosis nor temporal lobe epilepsy has been described in this syndrome previously, these conditions likely occurred in our patient as a result of NFIA haploinsufficiency.
Collapse
Affiliation(s)
- Mark A Colijn
- Department of Psychiatry, The University of Calgary, Calgary, AB, Canada
| | - Monica Hrynchak
- Molecular Cytogenetic Laboratory, Royal Columbian Hospital, The University of British Columbia, New Westminster, BC, Canada
| | - Chantelle T Hrazdil
- Division of Neurology, Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Veerle Willaeys
- BC Psychosis Program, British Columbia Mental Health & Substance Use Services, Vancouver, BC, Canada
| | - Randall F White
- BC Psychosis Program, British Columbia Mental Health & Substance Use Services, Vancouver, BC, Canada.,Department of Psychiatry, The University of British Columbia, Vancouver, BC, Canada
| | - Robert M Stowe
- BC Neuropsychiatry Program, Departments of Psychiatry and Neurology (Medicine), and Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
23
|
What Is Parvalbumin for? Biomolecules 2022; 12:biom12050656. [PMID: 35625584 PMCID: PMC9138604 DOI: 10.3390/biom12050656] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 12/28/2022] Open
Abstract
Parvalbumin (PA) is a small, acidic, mostly cytosolic Ca2+-binding protein of the EF-hand superfamily. Structural and physical properties of PA are well studied but recently two highly conserved structural motifs consisting of three amino acids each (clusters I and II), which contribute to the hydrophobic core of the EF-hand domains, have been revealed. Despite several decades of studies, physiological functions of PA are still poorly known. Since no target proteins have been revealed for PA so far, it is believed that PA acts as a slow calcium buffer. Numerous experiments on various muscle systems have shown that PA accelerates the relaxation of fast skeletal muscles. It has been found that oxidation of PA by reactive oxygen species (ROS) is conformation-dependent and one more physiological function of PA in fast muscles could be a protection of these cells from ROS. PA is thought to regulate calcium-dependent metabolic and electric processes within the population of gamma-aminobutyric acid (GABA) neurons. Genetic elimination of PA results in changes in GABAergic synaptic transmission. Mammalian oncomodulin (OM), the β isoform of PA, is expressed mostly in cochlear outer hair cells and in vestibular hair cells. OM knockout mice lose their hearing after 3–4 months. It was suggested that, in sensory cells, OM maintains auditory function, most likely affecting outer hair cells’ motility mechanisms.
Collapse
|
24
|
Inflammation and Nitro-oxidative Stress as Drivers of Endocannabinoid System Aberrations in Mood Disorders and Schizophrenia. Mol Neurobiol 2022; 59:3485-3503. [PMID: 35347586 DOI: 10.1007/s12035-022-02800-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 03/13/2022] [Indexed: 01/02/2023]
Abstract
The endocannabinoid system (ECS) is composed of the endocannabinoid ligands anandamide (AEA) and 2-arachidonoylgycerol (2-AG), their target cannabinoid receptors (CB1 and CB2) and the enzymes involved in their synthesis and metabolism (N-acyltransferase and fatty acid amide hydrolase (FAAH) in the case of AEA and diacylglycerol lipase (DAGL) and monoacylglycerol lipase (MAGL) in the case of 2-AG). The origins of ECS dysfunction in major neuropsychiatric disorders remain to be determined, and this paper explores the possibility that they may be associated with chronically increased nitro-oxidative stress and activated immune-inflammatory pathways, and it examines the mechanisms which might be involved. Inflammation and nitro-oxidative stress are associated with both increased CB1 expression, via increased activity of the NADPH oxidases NOX4 and NOX1, and increased CNR1 expression and DNA methylation; and CB2 upregulation via increased pro-inflammatory cytokine levels, binding of the transcription factor Nrf2 to an antioxidant response element in the CNR2 promoter region and the action of miR-139. CB1 and CB2 have antagonistic effects on redox signalling, which may result from a miRNA-enabled negative feedback loop. The effects of inflammation and oxidative stress are detailed in respect of AEA and 2-AG levels, via effects on calcium homeostasis and phospholipase A2 activity; on FAAH activity, via nitrosylation/nitration of functional cysteine and/or tyrosine residues; and on 2-AG activity via effects on MGLL expression and MAGL. Finally, based on these detailed molecular neurobiological mechanisms, it is suggested that cannabidiol and dimethyl fumarate may have therapeutic potential for major depressive disorder, bipolar disorder and schizophrenia.
Collapse
|
25
|
Lago SG, Bahn S. The druggable schizophrenia genome: from repurposing opportunities to unexplored drug targets. NPJ Genom Med 2022; 7:25. [PMID: 35338153 PMCID: PMC8956592 DOI: 10.1038/s41525-022-00290-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 02/04/2022] [Indexed: 12/04/2022] Open
Abstract
There have been no new drugs for the treatment of schizophrenia in several decades and treatment resistance represents a major unmet clinical need. The drugs that exist are based on serendipitous clinical observations rather than an evidence-based understanding of disease pathophysiology. In the present review, we address these bottlenecks by integrating common, rare, and expression-related schizophrenia risk genes with knowledge of the druggability of the human genome as a whole. We highlight novel drug repurposing opportunities, clinical trial candidates which are supported by genetic evidence, and unexplored therapeutic opportunities in the lesser-known regions of the schizophrenia genome. By identifying translational gaps and opportunities across the schizophrenia disease space, we discuss a framework for translating increasingly well-powered genetic association studies into personalized treatments for schizophrenia and initiating the vital task of characterizing clinically relevant drug targets in underexplored regions of the human genome.
Collapse
Affiliation(s)
- Santiago G Lago
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK.
| | - Sabine Bahn
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
26
|
Abstract
PURPOSE Development of new thymoleptic medications has primarily centered on anticonvulsants and antipsychotic drugs. Based on our studies of intracellular calcium ion signaling in mood disorders, we were interested in the use of novel medications that act on this mechanism of neuronal activation as potential mood stabilizers. METHOD We reviewed the dynamics of the calcium second messenger system and the international body of data demonstrating increased baseline and stimulated intracellular calcium levels in peripheral cells of patients with bipolar mood disorders. We then examined studies of the effect of established mood stabilizers on intracellular calcium ion levels and on mechanisms of mobilization of this second messenger. After summarizing studies of calcium channel blocking agents, whose primary action is to attenuate hyperactive intracellular calcium signaling, we considered clinical experience with this class of medications and the potential for further research. FINDINGS Established mood stabilizers normalize increased intracellular calcium ion levels in bipolar disorder patients. Most case series and controlled studies suggest an antimanic and possibly mood stabilizing effect of the calcium channel blocking medications verapamil and nimodipine, with fewer data on isradipine. A relatively low risk of teratogenicity and lack of cognitive adverse effects or weight gain suggest possible applications in pregnancy and in patients for whom these are considerations. IMPLICATIONS Medications that antagonize hyperactive intracellular signaling warrant more interest than they have received in psychiatry. Further experience will clarify the applications of these medications alone and in combination with more established mood stabilizers.
Collapse
|
27
|
Disrupted Cacna1c gene expression perturbs spontaneous Ca 2+ activity causing abnormal brain development and increased anxiety. Proc Natl Acad Sci U S A 2022; 119:2108768119. [PMID: 35135875 PMCID: PMC8851547 DOI: 10.1073/pnas.2108768119] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2021] [Indexed: 12/26/2022] Open
Abstract
The gene CACNA1C encodes for a calcium channel that has been linked to various psychiatric conditions, including schizophrenia and bipolar disorder, through hitherto unknown cellular mechanisms. Here, we report that deletion of Cacna1c in neurons of the developing brain disrupts spontaneous calcium activity and causes abnormal brain development and anxiety. Our results indicate that marginally alterations in the expression level of Cacna1c have major effects on the intrinsic spontaneous calcium activity of neural progenitors that play a crucial role in brain development. Thus, Cacna1c acts as a molecular switch that can increase susceptibility to psychiatric disease. The L-type voltage-gated Ca2+ channel gene CACNA1C is a risk gene for various psychiatric conditions, including schizophrenia and bipolar disorder. However, the cellular mechanism by which CACNA1C contributes to psychiatric disorders has not been elucidated. Here, we report that the embryonic deletion of Cacna1c in neurons destined for the cerebral cortex using an Emx1-Cre strategy disturbs spontaneous Ca2+ activity and causes abnormal brain development and anxiety. By combining computational modeling with electrophysiological membrane potential manipulation, we found that neural network activity was driven by intrinsic spontaneous Ca2+ activity in distinct progenitor cells expressing marginally increased levels of voltage-gated Ca2+ channels. MRI examination of the Cacna1c knockout mouse brains revealed volumetric differences in the neocortex, hippocampus, and periaqueductal gray. These results suggest that Cacna1c acts as a molecular switch and that its disruption during embryogenesis can perturb Ca2+ handling and neural development, which may increase susceptibility to psychiatric disease.
Collapse
|
28
|
Abstract
Bipolar disorder (BD) is a complex group of neuropsychiatric disorders, typically comprising both manic and depressive episodes. The underlying neuropathology of BD is not established, but a consistent feature is progressive thinning of cortical grey matter (GM) and white matter (WM) in specific pathways, due to loss of subpopulations of neurons and astrocytes, with accompanying disturbance of connectivity. Dysregulation of astrocyte homeostatic functions are implicated in BD, notably regulation of glutamate, calcium signalling, circadian rhythms and metabolism. Furthermore, the beneficial therapeutic effects of the frontline treatments for BD are due at least in part to their positive actions on astrocytes, notably lithium, valproic acid (VPA) and carbamazepine (CBZ), as well as antidepressants and antipsychotics that are used in the management of this disorder. Treatments for BD are ineffective in a large proportion of cases, and astrocytes represent new therapeutic targets that can also serve as biomarkers of illness progression and treatment responsiveness in BD.
Collapse
|
29
|
Suliman M, Schmidtke MW, Greenberg ML. The Role of the UPR Pathway in the Pathophysiology and Treatment of Bipolar Disorder. Front Cell Neurosci 2021; 15:735622. [PMID: 34531727 PMCID: PMC8439382 DOI: 10.3389/fncel.2021.735622] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/09/2021] [Indexed: 11/13/2022] Open
Abstract
Bipolar disorder (BD) is a mood disorder that affects millions worldwide and is associated with severe mood swings between mania and depression. The mood stabilizers valproate (VPA) and lithium (Li) are among the main drugs that are used to treat BD patients. However, these drugs are not effective for all patients and cause serious side effects. Therefore, better drugs are needed to treat BD patients. The main barrier to developing new drugs is the lack of knowledge about the therapeutic mechanism of currently available drugs. Several hypotheses have been proposed for the mechanism of action of mood stabilizers. However, it is still not known how they act to alleviate both mania and depression. The pathology of BD is characterized by mitochondrial dysfunction, oxidative stress, and abnormalities in calcium signaling. A deficiency in the unfolded protein response (UPR) pathway may be a shared mechanism that leads to these cellular dysfunctions. This is supported by reported abnormalities in the UPR pathway in lymphoblasts from BD patients. Additionally, studies have demonstrated that mood stabilizers alter the expression of several UPR target genes in mouse and human neuronal cells. In this review, we outline a new perspective wherein mood stabilizers exert their therapeutic mechanism by activating the UPR. Furthermore, we discuss UPR abnormalities in BD patients and suggest future research directions to resolve discrepancies in the literature.
Collapse
Affiliation(s)
- Mahmoud Suliman
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Michael W Schmidtke
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Miriam L Greenberg
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| |
Collapse
|
30
|
Clifton NE, Collado-Torres L, Burke EE, Pardiñas AF, Harwood JC, Di Florio A, Walters JTR, Owen MJ, O'Donovan MC, Weinberger DR, Holmans PA, Jaffe AE, Hall J. Developmental Profile of Psychiatric Risk Associated With Voltage-Gated Cation Channel Activity. Biol Psychiatry 2021; 90:399-408. [PMID: 33965196 PMCID: PMC8375582 DOI: 10.1016/j.biopsych.2021.03.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/26/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Recent breakthroughs in psychiatric genetics have implicated biological pathways onto which genetic risk for psychiatric disorders converges. However, these studies do not reveal the developmental time point(s) at which these pathways are relevant. METHODS We aimed to determine the relationship between psychiatric risk and developmental gene expression relating to discrete biological pathways. We used postmortem RNA sequencing data (BrainSeq and BrainSpan) from brain tissue at multiple prenatal and postnatal time points, with summary statistics from recent genome-wide association studies of schizophrenia, bipolar disorder, and major depressive disorder. We prioritized gene sets for overall enrichment of association with each disorder and then tested the relationship between the association of their constituent genes with their relative expression at each developmental stage. RESULTS We observed relationships between the expression of genes involved in voltage-gated cation channel activity during early midfetal, adolescence, and early adulthood time points and association with schizophrenia and bipolar disorder, such that genes more strongly associated with these disorders had relatively low expression during early midfetal development and higher expression during adolescence and early adulthood. The relationship with schizophrenia was strongest for the subset of genes related to calcium channel activity, while for bipolar disorder, the relationship was distributed between calcium and potassium channel activity genes. CONCLUSIONS Our results indicate periods during development when biological pathways related to the activity of calcium and potassium channels may be most vulnerable to the effects of genetic variants conferring risk for psychiatric disorders. Furthermore, they indicate key time points and potential targets for disorder-specific therapeutic interventions.
Collapse
Affiliation(s)
- Nicholas E Clifton
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom; MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom.
| | - Leonardo Collado-Torres
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland; Centre for Computational Biology, Johns Hopkins University Medical Campus, Baltimore, Maryland
| | - Emily E Burke
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland
| | - Antonio F Pardiñas
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Janet C Harwood
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Arianna Di Florio
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - James T R Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Michael J Owen
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom; MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Michael C O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland; Departments of Psychiatry, Neurology, Neuroscience and Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Peter A Holmans
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Andrew E Jaffe
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland; Centre for Computational Biology, Johns Hopkins University Medical Campus, Baltimore, Maryland; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom; MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
31
|
Fischer TT, Nguyen LD, Ehrlich BE. Neuronal calcium sensor 1 (NCS1) dependent modulation of neuronal morphology and development. FASEB J 2021; 35:e21873. [PMID: 34499766 DOI: 10.1096/fj.202100731r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/24/2021] [Accepted: 08/09/2021] [Indexed: 12/14/2022]
Abstract
Calcium (Ca2+ ) signaling is critical for neuronal functioning and requires the concerted interplay of numerous Ca2+ -binding proteins, including neuronal calcium sensor 1 (NCS1). Although an important role of NCS1 in neuronal processes and in neurodevelopmental and neurodegenerative diseases has been established, the underlying mechanisms remain enigmatic. Here, we systematically investigated the functions of NCS1 in the brain. Using Golgi-Cox staining, we observed a reduction in dendritic complexity and spine density in the prefrontal cortex and the dorsal hippocampus of Ncs1-/- mice, which may underlie concomitantly observed deficits in memory acquisition. Subsequent RNA sequencing of Ncs1-/- and Ncs1+/+ mouse brain tissues revealed that NCS1 modulates gene expression related to neuronal morphology and development. Investigation of developmental databases further supported a molecular role of NCS1 during brain development by identifying temporal gene expression patterns. Collectively, this study provides insights into NCS1-dependent signaling and lays the foundation for a better understanding of NCS1-associated diseases.
Collapse
Affiliation(s)
- Tom T Fischer
- Department of Pharmacology, Yale University, New Haven, Connecticut, USA.,Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Lien D Nguyen
- Department of Pharmacology, Yale University, New Haven, Connecticut, USA.,Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut, USA
| | - Barbara E Ehrlich
- Department of Pharmacology, Yale University, New Haven, Connecticut, USA.,Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut, USA.,Department of Celluar and Molecular Physiology, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
32
|
Salsaa M, Aziz K, Lazcano P, Schmidtke MW, Tarsio M, Hüttemann M, Reynolds CA, Kane PM, Greenberg ML. Valproate activates the Snf1 kinase in Saccharomyces cerevisiae by decreasing the cytosolic pH. J Biol Chem 2021; 297:101110. [PMID: 34428448 PMCID: PMC8449051 DOI: 10.1016/j.jbc.2021.101110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 11/27/2022] Open
Abstract
Valproate (VPA) is a widely used mood stabilizer, but its therapeutic mechanism of action is not understood. This knowledge gap hinders the development of more effective drugs with fewer side effects. Using the yeast model to elucidate the effects of VPA on cellular metabolism, we determined that the drug upregulated expression of genes normally repressed during logarithmic growth on glucose medium and increased levels of activated (phosphorylated) Snf1 kinase, the major metabolic regulator of these genes. VPA also decreased the cytosolic pH (pHc) and reduced glycolytic production of 2/3-phosphoglycerate. ATP levels and mitochondrial membrane potential were increased, and glucose-mediated extracellular acidification decreased in the presence of the drug, as indicated by a smaller glucose-induced shift in pH, suggesting that the major P-type proton pump Pma1 was inhibited. Interestingly, decreasing the pHc by omeprazole-mediated inhibition of Pma1 led to Snf1 activation. We propose a model whereby VPA lowers the pHc causing a decrease in glycolytic flux. In response, Pma1 is inhibited and Snf1 is activated, resulting in increased expression of normally repressed metabolic genes. These findings suggest a central role for pHc in regulating the metabolic program of yeast cells.
Collapse
Affiliation(s)
- Michael Salsaa
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, USA
| | - Kerestin Aziz
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, USA
| | - Pablo Lazcano
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, USA
| | - Michael W Schmidtke
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, USA
| | - Maureen Tarsio
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Christian A Reynolds
- Department of Emergency Medicine, School of Medicine, Wayne State University, Detroit, Michigan, USA; Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| | - Patricia M Kane
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Miriam L Greenberg
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, USA.
| |
Collapse
|
33
|
Suliman M, Schmidtke MW, Greenberg ML. A myo-inositol bioassay utilizing an auxotrophic strain of S. cerevisiae. J Microbiol Methods 2021; 189:106300. [PMID: 34389363 DOI: 10.1016/j.mimet.2021.106300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/06/2021] [Accepted: 08/08/2021] [Indexed: 11/16/2022]
Abstract
Myo-inositol is a six‑carbon sugar that is essential for the growth of mammalian cells and must be obtained through either extracellular uptake or de novo biosynthesis. The physiological importance of myo-inositol stems from its incorporation into phosphoinositides and inositol phosphates, which serve a variety of signaling, regulatory, and structural roles in cells. To study myo-inositol metabolism and function, it is essential to have a reliable method for assaying myo-inositol levels. However, current approaches to assay myo-inositol levels are time-consuming, expensive, and often unreliable. This article describes a simple new myo-inositol bioassay that utilizes an auxotrophic strain of S. cerevisiae to measure myo-inositol concentration in solutions. The accuracy of this method was confirmed by comparing assay values to those obtained by tandem mass spectrometry (LC-MS/MS). It is easy to perform, inexpensive, does not require sophisticated equipment, and is specific for myo-inositol.
Collapse
Affiliation(s)
- Mahmoud Suliman
- Wayne State University, Department of Biological Sciences, 5047 Gullen Mall, Detroit, MI 48202, USA
| | - Michael W Schmidtke
- Wayne State University, Department of Biological Sciences, 5047 Gullen Mall, Detroit, MI 48202, USA
| | - Miriam L Greenberg
- Wayne State University, Department of Biological Sciences, 5047 Gullen Mall, Detroit, MI 48202, USA.
| |
Collapse
|
34
|
Ithal D, Sukumaran SK, Bhattacharjee D, Vemula A, Nadella R, Mahadevan J, Sud R, Viswanath B, Purushottam M, Jain S. Exome hits demystified: The next frontier. Asian J Psychiatr 2021; 59:102640. [PMID: 33892377 DOI: 10.1016/j.ajp.2021.102640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/26/2021] [Indexed: 12/13/2022]
Abstract
Severe mental illnesses such as schizophrenia and bipolar disorder have complex inheritance patterns, involving both common and rare variants. Whole exome sequencing is a promising approach to find out the rare genetic variants. We had previously reported several rare variants in multiplex families with severe mental illnesses. The current article tries to summarise the biological processes and pattern of expression of genes harbouring the aforementioned variants, linking them to known clinical manifestations through a methodical narrative review. Of the 28 genes considered for this review from 7 families with multiple affected individuals, 6 genes are implicated in various neuropsychiatric manifestations including some variations in the brain morphology assessed by magnetic resonance imaging. Another 15 genes, though associated with neuropsychiatric manifestations, did not have established brain morphological changes whereas the remaining 7 genes did not have any previously recorded neuropsychiatric manifestations at all. Wnt/b-catenin signaling pathway was associated with 6 of these genes and PI3K/AKT, calcium signaling, ERK, RhoA and notch signaling pathways had at least 2 gene associations. We present a comprehensive review of biological and clinical knowledge about the genes previously reported in multiplex families with severe mental illness. A 'disease in dish approach' can be helpful to further explore the fundamental mechanisms.
Collapse
Affiliation(s)
- Dhruva Ithal
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Salil K Sukumaran
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Debanjan Bhattacharjee
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Alekhya Vemula
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Ravi Nadella
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Jayant Mahadevan
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Reeteka Sud
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Biju Viswanath
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Meera Purushottam
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India.
| | - Sanjeev Jain
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| |
Collapse
|
35
|
Jones GH, Rong C, Shariq AS, Mishra A, Machado-Vieira R. Intracellular Signaling Cascades in Bipolar Disorder. Curr Top Behav Neurosci 2021; 48:101-132. [PMID: 32860212 DOI: 10.1007/7854_2020_157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bipolar spectrum disorders carry a significant public health burden. Disproportionately high rates of suicide, incarceration, and comorbid medical conditions necessitate an extraordinary focus on understanding the intricacies of this disease. Elucidating granular, intracellular details seems to be a necessary preamble to advancing promising therapeutic opportunities. In this chapter, we review a wide range of intracellular mechanisms including mitochondrial energetics, calcium signaling, neuroinflammation, the microbiome, neurotransmitter metabolism, glycogen synthase kinase 3-beta (GSK3β), protein kinase C (PKC) and diacylglycerol (DAG), and neurotrophins (especially BDNF), as well as the glutamatergic, dopaminergic, purinergic, and neurohormonal systems. Owing to the relative lack of understanding and effective therapeutic options compared to the rest of the spectrum, special attention is paid in the chapter to the latest developments in bipolar depression. Likewise, from a therapeutic standpoint, special attention should be paid to the pervasive mechanistic actions of lithium as a means of amalgamating numerous, disparate cascades into a digestible cognitive topology.
Collapse
Affiliation(s)
- Gregory H Jones
- Department of Psychiatry, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Carola Rong
- Department of Psychiatry, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Aisha S Shariq
- Department of Psychiatry, Texas Tech University Health Science Center, El Paso, TX, USA
- Texas Tech University Health Science Center, Paul L. Foster School of Medicine, El Paso, TX, USA
| | - Abhinav Mishra
- Texas Tech University Health Science Center, Paul L. Foster School of Medicine, El Paso, TX, USA
| | - Rodrigo Machado-Vieira
- Department of Psychiatry, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
36
|
Levchenko A, Kanapin A, Samsonova A, Fedorenko OY, Kornetova EG, Nurgaliev T, Mazo GE, Semke AV, Kibitov AO, Bokhan NA, Gainetdinov RR, Ivanova SA. A genome-wide association study identifies a gene network associated with paranoid schizophrenia and antipsychotics-induced tardive dyskinesia. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110134. [PMID: 33065217 DOI: 10.1016/j.pnpbp.2020.110134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/10/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023]
Abstract
In the present study we conducted a genome-wide association study (GWAS) in a cohort of 505 patients with paranoid schizophrenia (SCZ), of which 95 had tardive dyskinesia (TD), and 503 healthy controls. Using data generated by the PsychENCODE Consortium (PEC) and other bioinformatic databases, we revealed a gene network, implicated in neurodevelopment and brain function, associated with both these disorders. Almost all these genes are in gene or isoform co-expression PEC network modules important for the functioning of the brain; the activity of these networks is also altered in SCZ, bipolar disorder and autism spectrum disorders. The associated PEC network modules are enriched for gene ontology terms relevant to the brain development and function (CNS development, neuron development, axon ensheathment, synapse, synaptic vesicle cycle, and signaling receptor activity) and to the immune system (inflammatory response). Results of the present study suggest that orofacial and limbtruncal types of TD seem to share the molecular network with SCZ. Paranoid SCZ and abnormal involuntary movements that indicate the orofacial type of TD are associated with the same genomic loci on chromosomes 3p22.2, 8q21.13, and 13q14.2. The limbtruncal type of TD is associated with a locus on chromosome 3p13 where the best functional candidate is FOXP1, a high-confidence SCZ gene. The results of this study shed light on common pathogenic mechanisms for SCZ and TD, and indicate that the pathogenesis of the orofacial and limbtruncal types of TD might be driven by interacting genes implicated in neurodevelopment.
Collapse
Affiliation(s)
- Anastasia Levchenko
- Theodosius Dobzhansky Center for Genome Bioinformatics, Saint Petersburg State University, Saint Petersburg, Russia.
| | - Alexander Kanapin
- Theodosius Dobzhansky Center for Genome Bioinformatics, Saint Petersburg State University, Saint Petersburg, Russia
| | - Anastasia Samsonova
- Theodosius Dobzhansky Center for Genome Bioinformatics, Saint Petersburg State University, Saint Petersburg, Russia
| | - Olga Yu Fedorenko
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia; National Research Tomsk Polytechnic University, Tomsk, Russia
| | - Elena G Kornetova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia; Siberian State Medical University, Tomsk, Russia
| | | | - Galina E Mazo
- Department of Endocrine Psychiatry, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, Saint Petersburg, Russia
| | - Arkadiy V Semke
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Alexander O Kibitov
- Department of Endocrine Psychiatry, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, Saint Petersburg, Russia; Laboratory of Molecular Genetics, Serbsky National Medical Research Center on Psychiatry and Addictions, Moscow, Russia
| | - Nikolay A Bokhan
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia; Siberian State Medical University, Tomsk, Russia; National Research Tomsk State University, Tomsk, Russia
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg, Russia
| | - Svetlana A Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia; National Research Tomsk Polytechnic University, Tomsk, Russia; Siberian State Medical University, Tomsk, Russia
| |
Collapse
|
37
|
Xia Y, Dai R, Wang K, Jiao C, Zhang C, Xu Y, Li H, Jing X, Chen Y, Jiang Y, Kopp RF, Giase G, Chen C, Liu C. Sex-differential DNA methylation and associated regulation networks in human brain implicated in the sex-biased risks of psychiatric disorders. Mol Psychiatry 2021; 26:835-848. [PMID: 30976086 PMCID: PMC6788945 DOI: 10.1038/s41380-019-0416-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 03/18/2019] [Accepted: 03/22/2019] [Indexed: 01/29/2023]
Abstract
Many psychiatric disorders are characterized by a strong sex difference, but the mechanisms behind sex-bias are not fully understood. DNA methylation plays important roles in regulating gene expression, ultimately impacting sexually different characteristics of the human brain. Most previous literature focused on DNA methylation alone without considering the regulatory network and its contribution to sex-bias of psychiatric disorders. Since DNA methylation acts in a complex regulatory network to connect genetic and environmental factors with high-order brain functions, we investigated the regulatory networks associated with different DNA methylation and assessed their contribution to the risks of psychiatric disorders. We compiled data from 1408 postmortem brain samples in 3 collections to identify sex-differentially methylated positions (DMPs) and regions (DMRs). We identified and replicated thousands of DMPs and DMRs. The DMR genes were enriched in neuronal related pathways. We extended the regulatory networks related to sex-differential methylation and psychiatric disorders by integrating methylation quantitative trait loci (meQTLs), gene expression, and protein-protein interaction data. We observed significant enrichment of sex-associated genes in psychiatric disorder-associated gene sets. We prioritized 2080 genes that were sex-biased and associated with psychiatric disorders, such as NRXN1, NRXN2, NRXN3, FDE4A, and SHANK2. These genes are enriched in synapse-related pathways and signaling pathways, suggesting that sex-differential genes of these neuronal pathways may cause the sex-bias of psychiatric disorders.
Collapse
Affiliation(s)
- Yan Xia
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Rujia Dai
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Kangli Wang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Chuan Jiao
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Chunling Zhang
- Department of Neuroscience, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Yuchen Xu
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Honglei Li
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xi Jing
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yu Chen
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Yi Jiang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Richard F Kopp
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Gina Giase
- Department of Public Health, University of Illinois at Chicago, Chicago, IL, USA
| | - Chao Chen
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA.
- National Clinical Research Center for Geriatric Disorders, the Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chunyu Liu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA.
- School of Psychology, Shaanxi Normal University, Xi'an, China.
| |
Collapse
|
38
|
Wollenhaupt-Aguiar B, Kapczinski F, Pfaffenseller B. Biological Pathways Associated with Neuroprogression in Bipolar Disorder. Brain Sci 2021; 11:brainsci11020228. [PMID: 33673277 PMCID: PMC7918818 DOI: 10.3390/brainsci11020228] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
There is evidence suggesting clinical progression in a subset of patients with bipolar disorder (BD). This progression is associated with worse clinical outcomes and biological changes. Molecular pathways and biological markers of clinical progression have been identified and may explain the progressive changes associated with this disorder. The biological basis for clinical progression in BD is called neuroprogression. We propose that the following intertwined pathways provide the biological basis of neuroprogression: inflammation, oxidative stress, impaired calcium signaling, endoplasmic reticulum and mitochondrial dysfunction, and impaired neuroplasticity and cellular resilience. The nonlinear interaction of these pathways may worsen clinical outcomes, cognition, and functioning. Understanding neuroprogression in BD is crucial for identifying novel therapeutic targets, preventing illness progression, and ultimately promoting better outcomes.
Collapse
Affiliation(s)
- Bianca Wollenhaupt-Aguiar
- Department of Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, ON L8N 3K7, Canada; (B.W.-A.); (F.K.)
- Mood Disorders Program, St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 3K7, Canada
| | - Flavio Kapczinski
- Department of Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, ON L8N 3K7, Canada; (B.W.-A.); (F.K.)
- Mood Disorders Program, St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 3K7, Canada
- Neuroscience Graduate Program, McMaster University, Hamilton, ON L8S 4L8, Canada
- Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-903, Brazil
- Department of Psychiatry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Brazil
| | - Bianca Pfaffenseller
- Department of Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, ON L8N 3K7, Canada; (B.W.-A.); (F.K.)
- Mood Disorders Program, St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 3K7, Canada
- Correspondence:
| |
Collapse
|
39
|
Comprehensive somatosensory and neurological phenotyping of NCS1 knockout mice. Sci Rep 2021; 11:2372. [PMID: 33504822 PMCID: PMC7840744 DOI: 10.1038/s41598-021-81650-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
Neuronal calcium sensor 1 (NCS1) regulates a wide range of cellular functions throughout the mammalian nervous systems. Altered NCS1 expression is associated with neurodevelopmental and neurodegenerative diseases. Previous studies focused on affective and cognitive behaviors in NCS1 knockout (KO) mice, but little is known about the physiological and pathological states associated with the loss of NCS1 in the peripheral nervous system. We previously reported that NCS1 expression was reduced following paclitaxel-induced peripheral neuropathy. Here, we comprehensively investigated the phenotypes of NCS1-KO mice through a battery of behavioral tests examining both central and peripheral nervous systems. Generally, only mild differences were observed in thermal sensation and memory acquisition between NCS1-WT and -KO male mice, but not in female mice. No differences were observed in motor performance, affective behaviors, and hearing in both sexes. These results suggest that NCS1 plays a modulatory role in sensory perceptions and cognition, particularly in male mice. NCS1 has been proposed as a pharmacological target for various diseases. Therefore, the sex-specific effects of NCS1 loss may be of clinical interest. As we examined a constitutive KO model, future studies focusing on various conditional KO models will further elucidate the precise physiological significance of NCS1.
Collapse
|
40
|
Reis de Assis D, Szabo A, Requena Osete J, Puppo F, O’Connell KS, A. Akkouh I, Hughes T, Frei E, A. Andreassen O, Djurovic S. Using iPSC Models to Understand the Role of Estrogen in Neuron-Glia Interactions in Schizophrenia and Bipolar Disorder. Cells 2021; 10:209. [PMID: 33494281 PMCID: PMC7909800 DOI: 10.3390/cells10020209] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/08/2020] [Accepted: 01/19/2021] [Indexed: 01/04/2023] Open
Abstract
Schizophrenia (SCZ) and bipolar disorder (BIP) are severe mental disorders with a considerable disease burden worldwide due to early age of onset, chronicity, and lack of efficient treatments or prevention strategies. Whilst our current knowledge is that SCZ and BIP are highly heritable and share common pathophysiological mechanisms associated with cellular signaling, neurotransmission, energy metabolism, and neuroinflammation, the development of novel therapies has been hampered by the unavailability of appropriate models to identify novel targetable pathomechanisms. Recent data suggest that neuron-glia interactions are disturbed in SCZ and BIP, and are modulated by estrogen (E2). However, most of the knowledge we have so far on the neuromodulatory effects of E2 came from studies on animal models and human cell lines, and may not accurately reflect many processes occurring exclusively in the human brain. Thus, here we highlight the advantages of using induced pluripotent stem cell (iPSC) models to revisit studies of mechanisms underlying beneficial effects of E2 in human brain cells. A better understanding of these mechanisms opens the opportunity to identify putative targets of novel therapeutic agents for SCZ and BIP. In this review, we first summarize the literature on the molecular mechanisms involved in SCZ and BIP pathology and the beneficial effects of E2 on neuron-glia interactions. Then, we briefly present the most recent developments in the iPSC field, emphasizing the potential of using patient-derived iPSCs as more relevant models to study the effects of E2 on neuron-glia interactions.
Collapse
Affiliation(s)
- Denis Reis de Assis
- NORMENT, Institute of Clinical Medicine, University of Oslo & Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway; (A.S.); (J.R.O.); (F.P.); (K.S.O.); (I.A.A.); (T.H.); (E.F.); (O.A.A.)
- Department of Medical Genetics, Oslo University Hospital, 0450 Oslo, Norway
| | - Attila Szabo
- NORMENT, Institute of Clinical Medicine, University of Oslo & Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway; (A.S.); (J.R.O.); (F.P.); (K.S.O.); (I.A.A.); (T.H.); (E.F.); (O.A.A.)
- Department of Medical Genetics, Oslo University Hospital, 0450 Oslo, Norway
| | - Jordi Requena Osete
- NORMENT, Institute of Clinical Medicine, University of Oslo & Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway; (A.S.); (J.R.O.); (F.P.); (K.S.O.); (I.A.A.); (T.H.); (E.F.); (O.A.A.)
- Department of Medical Genetics, Oslo University Hospital, 0450 Oslo, Norway
| | - Francesca Puppo
- NORMENT, Institute of Clinical Medicine, University of Oslo & Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway; (A.S.); (J.R.O.); (F.P.); (K.S.O.); (I.A.A.); (T.H.); (E.F.); (O.A.A.)
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Kevin S. O’Connell
- NORMENT, Institute of Clinical Medicine, University of Oslo & Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway; (A.S.); (J.R.O.); (F.P.); (K.S.O.); (I.A.A.); (T.H.); (E.F.); (O.A.A.)
| | - Ibrahim A. Akkouh
- NORMENT, Institute of Clinical Medicine, University of Oslo & Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway; (A.S.); (J.R.O.); (F.P.); (K.S.O.); (I.A.A.); (T.H.); (E.F.); (O.A.A.)
- Department of Medical Genetics, Oslo University Hospital, 0450 Oslo, Norway
| | - Timothy Hughes
- NORMENT, Institute of Clinical Medicine, University of Oslo & Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway; (A.S.); (J.R.O.); (F.P.); (K.S.O.); (I.A.A.); (T.H.); (E.F.); (O.A.A.)
- Department of Medical Genetics, Oslo University Hospital, 0450 Oslo, Norway
| | - Evgeniia Frei
- NORMENT, Institute of Clinical Medicine, University of Oslo & Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway; (A.S.); (J.R.O.); (F.P.); (K.S.O.); (I.A.A.); (T.H.); (E.F.); (O.A.A.)
- Department of Medical Genetics, Oslo University Hospital, 0450 Oslo, Norway
| | - Ole A. Andreassen
- NORMENT, Institute of Clinical Medicine, University of Oslo & Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway; (A.S.); (J.R.O.); (F.P.); (K.S.O.); (I.A.A.); (T.H.); (E.F.); (O.A.A.)
- Division of Mental Health and Addiction, Oslo University Hospital, 0372 Oslo, Norway
| | - Srdjan Djurovic
- NORMENT, Institute of Clinical Medicine, University of Oslo & Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway; (A.S.); (J.R.O.); (F.P.); (K.S.O.); (I.A.A.); (T.H.); (E.F.); (O.A.A.)
- NORMENT, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| |
Collapse
|
41
|
Shonibare DO, Patel RR, Islam AH, Metcalfe AWS, Fiksenbaum L, Freeman N, MacIntosh BJ, Kennedy JL, Goldstein BI. Neurostructural phenotypes of CACNA1C rs1006737 in adolescents with bipolar disorder and healthy controls. Prog Neuropsychopharmacol Biol Psychiatry 2021; 104:110071. [PMID: 32800865 DOI: 10.1016/j.pnpbp.2020.110071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 06/05/2020] [Accepted: 08/09/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Investigate the effects of CACNA1C rs1006737 on cortical and subcortical neurostructural phenotypes in Caucasian bipolar disorder (BD) and healthy control (HC) adolescents. METHODS Seventy-one adolescents (14-20 years; 38BD, 33HC) underwent 3-Tesla Magnetic Resonance Imaging (MRI). Region of interest (ROI) and vertex-wise analyses examined cortical volume, surface area (SA), and thickness, as well as subcortical volume. ROIs included the ventromedial prefrontal cortex (vmPFC), ventrolateral prefrontal cortex (vlPFC), anterior cingulate cortex (ACC), putamen, and amygdala. General linear models included main effects of diagnosis and rs1006737, and an interaction term, controlling for age, sex, and total intracranial volume. RESULTS Vertex-wise analysis found significant BD-by-rs1006737 interactions for prefrontal and occipital regions such that BD A-carriers were found to have greater SA relative to BD non-carriers, while HC A-carriers had reduced SA relative to HC non-carriers. ROI analysis found an interaction in the ACC such that BD A-carriers were found to have greater SA relative to BD non-carriers, while no significant difference was found in HCs. Main effects of rs1006737 were also found on ACC SA from ROI analysis, and occipital SA from vertex-wise analysis, such that A-carriers had larger SA relative to non-carriers in both of these regions. CONCLUSIONS The current study identified neurostructural intermediate phenotypes relevant to the impact of CACNA1C rs1006737 on adolescent BD. Further investigation is warranted into the neurofunctional and neurocognitive relevance of rs1006737 associations with BD-specific elevations in regional SA.
Collapse
Affiliation(s)
- Daniel O Shonibare
- Centre for Youth Bipolar Disorder, Sunnybrook Health Sciences Centre, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Ronak R Patel
- Centre for Youth Bipolar Disorder, Sunnybrook Health Sciences Centre, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Alvi H Islam
- Centre for Youth Bipolar Disorder, Sunnybrook Health Sciences Centre, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Arron W S Metcalfe
- Centre for Youth Bipolar Disorder, Sunnybrook Health Sciences Centre, Toronto, Canada; Hurvitz Brain Sciences, Sunnybrook Research Institute, Toronto, Canada; Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Canada
| | - Lisa Fiksenbaum
- Centre for Youth Bipolar Disorder, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Natalie Freeman
- Tanenbaum Centre for Pharmacogenetics, Centre for Addiction and Mental Health, Toronto, Canada
| | - Bradley J MacIntosh
- Hurvitz Brain Sciences, Sunnybrook Research Institute, Toronto, Canada; Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Canada; Department of Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - James L Kennedy
- Institute of Medical Science, University of Toronto, Toronto, Canada; Tanenbaum Centre for Pharmacogenetics, Centre for Addiction and Mental Health, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Benjamin I Goldstein
- Centre for Youth Bipolar Disorder, Sunnybrook Health Sciences Centre, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada.
| |
Collapse
|
42
|
Harrison PJ, Hall N, Mould A, Al-Juffali N, Tunbridge EM. Cellular calcium in bipolar disorder: systematic review and meta-analysis. Mol Psychiatry 2021; 26:4106-4116. [PMID: 31801967 PMCID: PMC8550977 DOI: 10.1038/s41380-019-0622-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/20/2019] [Accepted: 11/22/2019] [Indexed: 12/31/2022]
Abstract
Calcium signalling has long been implicated in bipolar disorder, especially by reports of altered intracellular calcium ion concentrations ([Ca2+]). However, the evidence has not been appraised critically. We carried out a systematic review and meta-analysis of studies of cellular calcium indices in bipolar disorder. 2281 records were identified and 117 screened, of which 32 were eligible and 21 were suitable for meta-analyses. The latter each involved up to 642 patients and 404 control subjects. We found that basal free intracellular [Ca2+] is increased in bipolar disorder, both in platelets and in lymphocytes. The effect size is 0.55, with an estimated elevation of 29%. It is observed in medication-free patients. It is present in mania and bipolar depression, but data are equivocal for euthymia. Cells from bipolar disorder individuals also show an enhanced [Ca2+] response to stimulation with 5-HT or thrombin, by an estimated 25%, with an effect size of 0.63. In studies which included other diagnoses, intracellular basal [Ca2+] was higher in bipolar disorder than in unipolar depression, but not significantly different from schizophrenia. Functional parameters of cellular Ca2+ (e.g. calcium transients), and neuronal [Ca2+], have been much less investigated, and no firm conclusions can be drawn. In summary, there is a robust, medium effect size elevation of basal and stimulated free intracellular [Ca2+] in bipolar disorder. The results suggest altered calcium functioning in the disorder, and encourage further investigations into the underlying mechanisms, and the implications for pathophysiology and therapeutics.
Collapse
Affiliation(s)
- Paul J. Harrison
- grid.4991.50000 0004 1936 8948Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX3 7JX UK ,grid.451190.80000 0004 0573 576XOxford Health NHS Foundation Trust, Oxford, UK
| | - Nicola Hall
- grid.4991.50000 0004 1936 8948Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX3 7JX UK
| | - Arne Mould
- grid.4991.50000 0004 1936 8948Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX3 7JX UK ,grid.451190.80000 0004 0573 576XOxford Health NHS Foundation Trust, Oxford, UK
| | - Noura Al-Juffali
- grid.4991.50000 0004 1936 8948Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX3 7JX UK ,grid.451190.80000 0004 0573 576XOxford Health NHS Foundation Trust, Oxford, UK
| | - Elizabeth M. Tunbridge
- grid.4991.50000 0004 1936 8948Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX3 7JX UK ,grid.451190.80000 0004 0573 576XOxford Health NHS Foundation Trust, Oxford, UK
| |
Collapse
|
43
|
Xu C, Wang X, Zhou S, Wu J, Geng Q, Ruan D, Qiu Y, Quan J, Ding R, Cai G, Wu Z, Zheng E, Yang J. Brain Transcriptome Analysis Reveals Potential Transcription Factors and Biological Pathways Associated with Feed Efficiency in Commercial DLY Pigs. DNA Cell Biol 2020; 40:272-282. [PMID: 33297854 DOI: 10.1089/dna.2020.6071] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Feed efficiency (FE) is one of the most important economic traits in the porcine industry. In this study, high-throughput RNA sequencing (RNA-seq) was first utilized for brain tissue transcriptome analysis in pigs to indicate the potential genes and biological pathways related to FE in pigs. A total of 8 pigs with either extremely high-FE group (HE-group) or low-FE group (LE-group) were selected from 225 Duroc × (Landrace × Yorkshire) (DLY) pigs for transcriptomic analysis. RNA-seq analysis was performed to determine differentially expressed genes (DEGs) between the HE- and LE-group, and 430 DEGs were identified in brain tissues of pigs (|log2(FoldChange)| > 1; adjusted p-values <0.05). Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that the DEGs were mainly enriched in synaptic signaling or transmission, and hormone secretion pathways, in which insulin secretion, and oxytocin signaling pathways were closely associated with FE by regulating feeding behavior and energy metabolism (adjusted p-values <0.05). Further, the transcription factors (TFs) analysis and gene co-expression network analysis indicated three hub differentially expressed TFs (NR2F2, TFAP2D, and HNF1B) that affected FE by mainly regulating feeding behavior, insulin sensitivity, or energy metabolism. Our findings suggest several potential TFs and biological pathways for further investigations of FE in pigs.
Collapse
Affiliation(s)
- Cineng Xu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Xingwang Wang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Shenping Zhou
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Jie Wu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Qian Geng
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Donglin Ruan
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Yibin Qiu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Jianping Quan
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Rongrong Ding
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Gengyuan Cai
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Zhenfang Wu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Enqin Zheng
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| | - Jie Yang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, P.R. China
| |
Collapse
|
44
|
Xy Ling N, Langendorf CG, Hoque A, Galic S, Loh K, Kemp BE, Gundlach AL, Oakhill JS, Scott JW. Functional analysis of an R311C variant of Ca 2+ -calmodulin-dependent protein kinase kinase-2 (CaMKK2) found as a de novo mutation in a patient with bipolar disorder. Bipolar Disord 2020; 22:841-848. [PMID: 32216002 DOI: 10.1111/bdi.12901] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVES Loss-of-function mutations in the gene encoding the calcium-calmodulin (Ca2+ -CaM)-dependent protein kinase kinase-2 (CaMKK2) enzyme are linked to bipolar disorder. Recently, a de novo arginine to cysteine (R311C) mutation in CaMKK2 was identified from a whole exome sequencing study of bipolar patients and their unaffected parents. The aim of the present study was to determine the functional consequences of the R311C mutation on CaMKK2 activity and regulation by Ca2+ -CaM. METHODS The effects of the R311C mutation on CaMKK2 activity and Ca2+ -CaM activation were examined using a radiolabeled adenosine triphosphate (ATP) kinase assay. We performed immunoblot analysis to determine whether the R311C mutation impacts threonine-85 (T85) autophosphorylation, an activating phosphorylation site on CaMKK2 that has also been implicated in bipolar disorder. We also expressed the R311C mutant in CaMKK2 knockout HAP1 cells and used immunoblot analysis and an MTS reduction assay to study its effects on Ca2+ -dependent downstream signaling and cell viability, respectively. RESULTS The R311C mutation maps to the conserved HRD motif within the catalytic loop of CaMKK2 and caused a marked reduction in kinase activity and Ca2+ -CaM activation. The R311C mutation virtually abolished T85 autophosphorylation in response to Ca2+ -CaM and exerted a dominant-negative effect in cells as it impaired the ability of wild-type CaMKK2 to initiate downstream signaling and maintain cell viability. CONCLUSIONS The highly disruptive, loss-of-function impact of the de novo R311C mutation in human CaMKK2 provides a compelling functional rationale for being considered a potential rare monogenic cause of bipolar disorder.
Collapse
Affiliation(s)
- Naomi Xy Ling
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia
| | - Christopher G Langendorf
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia
| | - Ashfaqul Hoque
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia
| | - Sandra Galic
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia
| | - Kim Loh
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia
| | - Bruce E Kemp
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia.,Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
| | - Andrew L Gundlach
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Jonathan S Oakhill
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia.,Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
| | - John W Scott
- St Vincent's Institute and Department of Medicine, The University of Melbourne, Fitzroy, Australia.,Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia.,The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| |
Collapse
|
45
|
Pandey GN, Rizavi HS, Ren X. Protein and mRNA expression of protein kinase C (PKC) in the postmortem brain of bipolar and schizophrenic subjects. J Psychiatr Res 2020; 130:362-371. [PMID: 32882578 PMCID: PMC7554203 DOI: 10.1016/j.jpsychires.2020.07.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 11/25/2022]
Abstract
Abnormalities of protein kinase C (PKC) have been implicated in the pathophysiology of bipolar (BP) illness. This is primarily based on studies of PKC in platelets of BP patients. Whether such abnormalities of PKC activity and isoforms exist in the brain is unclear. We have therefore determined PKC activity, protein and mRNA expression of PKC isoforms in the prefrontal cortex (PFC), cingulate cortex (CING) and temporal cortex (TEMP) from BP (n = 19), schizophrenic (SZ) (n = 20) and normal control (NC) (n = 25) subjects. The brain samples were obtained from the Harvard Brain Bank, and the subjects were diagnosed according to DSM-IV criteria. Protein levels were determined using Western blot technique and mRNA levels were determined using real-time PCR (qPCR) method. We found that there was a significant decrease in the PKC activity in the cytosol and membrane fractions of PFC and TEMP obtained from BP subjects but not from SZ subjects. When we compared the expression of PKC isozymes, we found that the protein and mRNA expression of several isozymes was significantly decreased in the PFC (i.e., PKCα, PKCβI, PKCβII and PKCε) and TEMP (i.e., PKCα, PKCβI, PKCβII, PKCε and PKCγ) of BP subjects, but not in the CING. Overall, there was no difference in the mRNA or protein expression of PKC isozymes between SZ and NC subjects in any of the three brain areas we studied. Our results show that there is a region-specific decrease of certain PKC isozymes in the membrane and cytosol fractions of BP but not SZ subjects.
Collapse
|
46
|
Mitochondria under the spotlight: On the implications of mitochondrial dysfunction and its connectivity to neuropsychiatric disorders. Comput Struct Biotechnol J 2020; 18:2535-2546. [PMID: 33033576 PMCID: PMC7522539 DOI: 10.1016/j.csbj.2020.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/30/2022] Open
Abstract
Neuropsychiatric disorders (NPDs) such as bipolar disorder (BD), schizophrenia (SZ) and mood disorder (MD) are hard to manage due to overlapping symptoms and lack of biomarkers. Risk alleles of BD/SZ/MD are emerging, with evidence suggesting mitochondrial (mt) dysfunction as a critical factor for disease onset and progression. Mood stabilizing treatments for these disorders are scarce, revealing the need for biomarker discovery and artificial intelligence approaches to design synthetically accessible novel therapeutics. Here, we show mt involvement in NPDs by associating 245 mt proteins to BD/SZ/MD, with 7 common players in these disease categories. Analysis of over 650 publications suggests that 245 NPD-linked mt proteins are associated with 800 other mt proteins, with mt impairment likely to rewire these interactions. High dosage of mood stabilizers is known to alleviate manic episodes, but which compounds target mt pathways is another gap in the field that we address through mood stabilizer-gene interaction analysis of 37 prescriptions and over-the-counter psychotropic treatments, which we have refined to 15 mood-stabilizing agents. We show 26 of the 245 NPD-linked mt proteins are uniquely or commonly targeted by one or more of these mood stabilizers. Further, induced pluripotent stem cell-derived patient neurons and three-dimensional human brain organoids as reliable BD/SZ/MD models are outlined, along with multiomics methods and machine learning-based decision making tools for biomarker discovery, which remains a bottleneck for precision psychiatry medicine.
Collapse
|
47
|
Manduchi E, Fu W, Romano JD, Ruberto S, Moore JH. Embedding covariate adjustments in tree-based automated machine learning for biomedical big data analyses. BMC Bioinformatics 2020; 21:430. [PMID: 32998684 PMCID: PMC7528347 DOI: 10.1186/s12859-020-03755-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/15/2020] [Indexed: 12/03/2022] Open
Abstract
Background A typical task in bioinformatics consists of identifying which features are associated with a target outcome of interest and building a predictive model. Automated machine learning (AutoML) systems such as the Tree-based Pipeline Optimization Tool (TPOT) constitute an appealing approach to this end. However, in biomedical data, there are often baseline characteristics of the subjects in a study or batch effects that need to be adjusted for in order to better isolate the effects of the features of interest on the target. Thus, the ability to perform covariate adjustments becomes particularly important for applications of AutoML to biomedical big data analysis.
Results We developed an approach to adjust for covariates affecting features and/or target in TPOT. Our approach is based on regressing out the covariates in a manner that avoids ‘leakage’ during the cross-validation training procedure. We describe applications of this approach to toxicogenomics and schizophrenia gene expression data sets. The TPOT extensions discussed in this work are available at https://github.com/EpistasisLab/tpot/tree/v0.11.1-resAdj. Conclusions In this work, we address an important need in the context of AutoML, which is particularly crucial for applications to bioinformatics and medical informatics, namely covariate adjustments. To this end we present a substantial extension of TPOT, a genetic programming based AutoML approach. We show the utility of this extension by applications to large toxicogenomics and differential gene expression data. The method is generally applicable in many other scenarios from the biomedical field.
Collapse
Affiliation(s)
- Elisabetta Manduchi
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA. .,Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Weixuan Fu
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Joseph D Romano
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Stefano Ruberto
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason H Moore
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
48
|
Lago SG, Tomasik J, van Rees GF, Ramsey JM, Haenisch F, Cooper JD, Broek JA, Suarez-Pinilla P, Ruland T, Auyeug B, Mikova O, Kabacs N, Arolt V, Baron-Cohen S, Crespo-Facorro B, Bahn S. Exploring the neuropsychiatric spectrum using high-content functional analysis of single-cell signaling networks. Mol Psychiatry 2020; 25:2355-2372. [PMID: 30038233 DOI: 10.1038/s41380-018-0123-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 05/04/2018] [Accepted: 05/25/2018] [Indexed: 12/26/2022]
Abstract
Neuropsychiatric disorders overlap in symptoms and share genetic risk factors, challenging their current classification into distinct diagnostic categories. Novel cross-disorder approaches are needed to improve our understanding of the heterogeneous nature of neuropsychiatric diseases and overcome existing bottlenecks in their diagnosis and treatment. Here we employ high-content multi-parameter phospho-specific flow cytometry, fluorescent cell barcoding and automated sample preparation to characterize ex vivo signaling network responses (n = 1764) measured at the single-cell level in B and T lymphocytes across patients diagnosed with four major neuropsychiatric disorders: autism spectrum condition (ASC), bipolar disorder (BD), major depressive disorder (MDD), and schizophrenia (SCZ; n = 25 each), alongside matched healthy controls (n = 100). We identified 25 nodes (individual cell subtype-epitope-ligand combinations) significantly altered relative to the control group, with variable overlap between different neuropsychiatric diseases and heterogeneously expressed at the level of each individual patient. Reconstruction of the diagnostic categories from the altered nodes revealed an overlapping neuropsychiatric spectrum extending from MDD on one end, through BD and SCZ, to ASC on the other end. Network analysis showed that although the pathway structure of the epitopes was broadly preserved across the clinical groups, there were multiple discrete alterations in network connectivity, such as disconnections within the antigen/integrin receptor pathway and increased negative regulation within the Akt1 pathway in CD4+ T cells from ASC and SCZ patients, in addition to increased correlation of Stat1 (pY701) and Stat5 (pY694) responses in B cells from BD and MDD patients. Our results support the "dimensional" approach to neuropsychiatric disease classification and suggest potential novel drug targets along the neuropsychiatric spectrum.
Collapse
Affiliation(s)
- Santiago G Lago
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Jakub Tomasik
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Geertje F van Rees
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Jordan M Ramsey
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Frieder Haenisch
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Jason D Cooper
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Jantine A Broek
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Paula Suarez-Pinilla
- Department of Psychiatry, Marqués de Valdecilla University Hospital, IDIVAL, School of Medicine, University of Cantabria, Santander, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Santander, Spain
| | - Tillmann Ruland
- Department of Psychiatry and Psychotherapy, University of Münster, Münster, Germany
| | - Bonnie Auyeug
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK.,Psychology Department, Edinburgh University, Scotland, UK
| | - Olya Mikova
- Foundation Biological Psychiatry, Sofia, Bulgaria
| | - Nikolett Kabacs
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| | - Volker Arolt
- Department of Psychiatry and Psychotherapy, University of Münster, Münster, Germany
| | - Simon Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK.,CLASS Clinic, Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| | - Benedicto Crespo-Facorro
- Department of Psychiatry, Marqués de Valdecilla University Hospital, IDIVAL, School of Medicine, University of Cantabria, Santander, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Santander, Spain
| | - Sabine Bahn
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
49
|
Moon AL, Brydges NM, Wilkinson LS, Hall J, Thomas KL. Cacna1c Hemizygosity Results in Aberrant Fear Conditioning to Neutral Stimuli. Schizophr Bull 2020; 46:1231-1238. [PMID: 31910256 PMCID: PMC7505182 DOI: 10.1093/schbul/sbz127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
CACNA1C, a gene that encodes an alpha-1 subunit of L-type voltage-gated calcium channels, has been strongly associated with psychiatric disorders including schizophrenia and bipolar disorder. An important objective is to understand how variation in this gene can lead to an increased risk of psychopathology. Altered associative learning has also been implicated in the pathology of psychiatric disorders, particularly in the manifestation of psychotic symptoms. In this study, we utilize auditory-cued fear memory paradigms in order to investigate whether associative learning is altered in rats hemizygous for the Cacna1c gene. Cacna1c hemizygous (Cacna1c+/-) rats and their wild-type littermates were exposed to either delay, trace, or unpaired auditory fear conditioning. All rats received a Context Recall (24 h post-conditioning) and a Cue Recall (48 h post-conditioning) to test their fear responses. In the delay condition, which results in strong conditioning to the cue in wild-type animals, Cacna1c+/- rats showed increased fear responses to the context. In the trace condition, which results in strong conditioning to the context in wild-type animals, Cacna1c+/- rats showed increased fear responses to the cue. Finally, in the unpaired condition, Cacna1c+/- rats showed increased fear responses to both context and cue. These results indicate that Cacna1c heterozygous rats show aberrantly enhanced fear responses to inappropriate cues, consistent with key models of psychosis.
Collapse
Affiliation(s)
- Anna L Moon
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Nichola M Brydges
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
| | - Lawrence S Wilkinson
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
- School of Psychology, Cardiff University, Cardiff, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Kerrie L Thomas
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
- School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
50
|
Levchenko A, Vyalova NM, Nurgaliev T, Pozhidaev IV, Simutkin GG, Bokhan NA, Ivanova SA. NRG1, PIP4K2A, and HTR2C as Potential Candidate Biomarker Genes for Several Clinical Subphenotypes of Depression and Bipolar Disorder. Front Genet 2020; 11:936. [PMID: 33193575 PMCID: PMC7478333 DOI: 10.3389/fgene.2020.00936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 07/27/2020] [Indexed: 12/20/2022] Open
Abstract
GSK3B, BDNF, NGF, NRG1, HTR2C, and PIP4K2A play important roles in molecular mechanisms of psychiatric disorders. GSK3B occupies a central position in these molecular mechanisms and is also modulated by psychotropic drugs. BDNF regulates a number of key aspects in neurodevelopment and synaptic plasticity. NGF exerts a trophic action and is implicated in cerebral alterations associated with psychiatric disorders. NRG1 is active in neural development, synaptic plasticity, and neurotransmission. HTR2C is another important psychopharmacological target. PIP4K2A catalyzes the phosphorylation of PI5P to form PIP2, the latter being implicated in various aspects of neuronal signal transduction. In the present study, the six genes were sequenced in a cohort of 19 patients with bipolar affective disorder, 41 patients with recurrent depressive disorder, and 55 patients with depressive episode. The study revealed a number of genetic variants associated with antidepressant treatment response, time to recurrence of episodes, and depression severity. Namely, alleles of rs35641374 and rs10508649 (NRG1 and PIP4K2A) may be prognostic biomarkers of time to recurrence of depressive and manic/mixed episodes among patients with bipolar affective disorder. Alleles of NC_000008.11:g.32614509_32614510del, rs61731109, and rs10508649 (also NRG1 and PIP4K2A) seem to be predictive biomarkers of response to pharmacological antidepressant treatment on the 28th day assessed by the HDRS-17 or CGI-I scale. In particular, the allele G of rs10508649 (PIP4K2A) may increase resistance to antidepressant treatment and be at the same time protective against recurrent manic/mixed episodes. These results support previous data indicating a biological link between resistance to antidepressant treatment and mania. Bioinformatic functional annotation of associated variants revealed possible impact for transcriptional regulation of PIP4K2A. In addition, the allele A of rs2248440 (HTR2C) may be a prognostic biomarker of depression severity. This allele decreases expression of the neighboring immune system gene IL13RA2 in the putamen according to the GTEx portal. The variant rs2248440 is near rs6318 (previously associated with depression and effects of psychotropic drugs) that is an eQTL for the same gene and tissue. Finally, the study points to several protein interactions relevant in the pathogenesis of mood disorders. Functional studies using cellular or animal models are warranted to support these results.
Collapse
Affiliation(s)
- Anastasia Levchenko
- Theodosius Dobzhansky Center for Genome Bioinformatics, Saint Petersburg State University, Saint Petersburg, Russia
| | - Natalia M Vyalova
- Tomsk National Research Medical Center, Mental Health Research Institute, Russian Academy of Sciences, Tomsk, Russia
| | - Timur Nurgaliev
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg, Russia
| | - Ivan V Pozhidaev
- Tomsk National Research Medical Center, Mental Health Research Institute, Russian Academy of Sciences, Tomsk, Russia
| | - German G Simutkin
- Tomsk National Research Medical Center, Mental Health Research Institute, Russian Academy of Sciences, Tomsk, Russia
| | - Nikolay A Bokhan
- Tomsk National Research Medical Center, Mental Health Research Institute, Russian Academy of Sciences, Tomsk, Russia.,National Research Tomsk State University, Tomsk, Russia.,Siberian State Medical University, Tomsk, Russia
| | - Svetlana A Ivanova
- Tomsk National Research Medical Center, Mental Health Research Institute, Russian Academy of Sciences, Tomsk, Russia.,Siberian State Medical University, Tomsk, Russia.,National Research Tomsk Polytechnic University, Tomsk, Russia
| |
Collapse
|