1
|
Ni L, Zhu Y, Lv L, Zhang R, Xie S, Zhang X. Peripheral blood miR-16-5p as a potential biomarker for distinguishing unmedicated bipolar disorder type II from major depressive disorder. J Affect Disord 2025:S0165-0327(25)00702-5. [PMID: 40274128 DOI: 10.1016/j.jad.2025.04.126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
OBJECTIVE major depressive disorder (MDD) and bipolar disorder type II (BD-II) are difficult to distinguish clinically due to similar depressive symptoms and unrecognizable hypomania symptoms in the early stages. The study aims to identify these two disorders in the early stages through differential expression of microRNAs. METHODS 93 subjects including 66 unmedicated patients (33 MDD, 33 BD-II), and 27 healthy controls (HC) were enrolled. At the time of enrollment, all subjects' demographic data, HAMD, HCL-32, and YMRS scales were assessed. 5 ml of peripheral blood for all subjects was collected for microRNA second-generation sequencing. MicroRNA differential expression, target gene GO and KEGG analyses were performed. RESULTS No statistical differences in demographic data were found except for age (BD-II < MDD, P = 0.002). In terms of clinical data, there are differences in the course of the disease (BD-II > MDD, P = 0.037) and the HCL-32 (BD-II > MDD, P < 0.01). A variance analysis of microRNA expressions across all three groups identified eight highly expressed differential miRNAs (P < 0.001), Pairwise comparisons revealed that the expression level of miR-16-5p was lower in both MDD group (P < 0.05) and BD-II group (P < 0.001) than in HC group, and it was even lower in BD-II group compared to MDD group (P < 0.01). The area under the curve (AUC) for miR-16-5p in differentiating BD-II from MDD groups was 0.723 (P = 0.003). CONCLUSIONS Peripheral blood miR-16-5p may serve as a potential biomarker for distinguishing unmedicated BD-II from MDD patients.
Collapse
Affiliation(s)
- Longyan Ni
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuanyuan Zhu
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lanlan Lv
- Department of Psychiatry, The Affiliated Xuzhou Oriental Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Rongrong Zhang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shiping Xie
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Xiangrong Zhang
- Department of Geriatric Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
2
|
Kukla-Bartoszek M, Piechota M, Suski M, Hajto J, Borczyk M, Basta-Kaim A, Głombik K. Integrated Profiling Identifies Long-Term Molecular Consequences of Prenatal Dexamethasone Treatment in the Rat Brain-Potential Triggers of Depressive Phenotype and Cognitive Impairment. Mol Neurobiol 2025; 62:5183-5201. [PMID: 39528842 PMCID: PMC11880045 DOI: 10.1007/s12035-024-04586-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Prenatal excess of glucocorticoids (GCs) is considered to be one of the highly impacting factors contributing to depression development. Although GCs are crucial for normal fetal development and their administration (mainly dexamethasone, DEX) is a life-saving procedure for those at risk of preterm delivery, exposure to excess levels of GCs during pregnancy can yield detrimental consequences. Therefore, we aimed to systematically investigate the brain molecular alterations triggered by prenatal DEX administration. We used a rat model of depression based on prenatal exposure to DEX and performed integrative multi-level methylomic, transcriptomic, and proteomic analyses of adult rats' brains (i.e., frontal cortex (FCx) and hippocampus (Hp)) to identify the outcomes of DEX action. Each of the investigated levels was significantly affected by DEX in the long-term manner. Particularly, we found 200 CpG islands to be differentially methylated in the FCx and 200 in the Hp of prenatally DEX-treated rats. Global transcriptomic analysis uncovered differential expression of transcripts mostly in FCx (271) and 1 in Hp, while proteomic study identified 146 differentially expressed proteins in FCx and 123 in Hp. Among the identified enriched molecular networks, we found altered pathways involved in synaptic plasticity (i.e., cAMP, calcium, and Wnt signaling pathways or tight junctions and adhesion molecules), which may contribute to cognitive impairment, observed in DEX-treated animals. Moreover, in the FCx, DEX administration in the prenatal period downregulates the expression of ribosome protein genes associated both with large and small ribosomal subunit assembly which can lead to a global decrease in translation and protein synthesis processes and, indirectly, alterations in the neurotransmission process.
Collapse
Affiliation(s)
- Magdalena Kukla-Bartoszek
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Polish Academy of Sciences, Maj Institute of Pharmacology, Smętna 12, 31-343, Kraków, Poland
| | - Marcin Piechota
- Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Maciej Suski
- Department of Pharmacology, Jagiellonian University Medical College, Faculty of Medicine, Grzegórzecka 16, 31-531, Kraków, Poland
- Centre for the Development of Therapies for Civilization and Age-Related Diseases CDT-CARD, Jagiellonian University Medical College, Skawińska 8, 31-066, Kraków, Poland
| | - Jacek Hajto
- Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Małgorzata Borczyk
- Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Polish Academy of Sciences, Maj Institute of Pharmacology, Smętna 12, 31-343, Kraków, Poland
| | - Katarzyna Głombik
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Polish Academy of Sciences, Maj Institute of Pharmacology, Smętna 12, 31-343, Kraków, Poland.
| |
Collapse
|
3
|
Panizzutti B, Bortolasci CC, Spolding B, Kidnapillai S, Connor T, Truong TT, Liu ZS, Hernández D, Gray L, Kim JH, Dean OM, Berk M, Walder K. Effect of antipsychotics on the focal adhesion pathway. World J Biol Psychiatry 2025; 26:146-152. [PMID: 39846496 DOI: 10.1080/15622975.2025.2453181] [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: 09/02/2024] [Revised: 01/05/2025] [Accepted: 01/09/2025] [Indexed: 01/24/2025]
Abstract
Focal adhesions and their dynamic nature are essential for various physiological processes, including the formation of neurites, synaptic function and plasticity. Alterations in these processes have been associated with schizophrenia and bipolar disorder. OBJECTIVES This study aimed to explore the impact of pharmacological treatments used for bipolar disorder and schizophrenia on the expression of genes involved in the focal adhesion pathway, addressing a gap in understanding the interaction between medication effects and disease pathophysiology. METHODS NT2-N (neuron-like) cells were exposed to treatment with amisulpride, aripiprazole, chlorpromazine, clozapine, haloperidol, olanzapine, quetiapine, risperidone, or vehicle for 24 h. Genome-wide mRNA expression was analysed using gene set enrichment analysis. RESULTS The analysis revealed that seven out of the eight drugs widely prescribed for bipolar disorder and schizophrenia downregulate the expression of genes associated with the focal adhesions pathway. Focal adhesion was the pathway with the most negative normalised enrichment score across all treatments. CONCLUSIONS Our results support the hypothesis that focal adhesion pathways may play a role in the pathophysiology of bipolar disorder and schizophrenia. Moreover, the data underscore the importance of differentiating medication effects from disease mechanisms in psychiatric research, a challenge compounded by the medicated state of most study participants.
Collapse
Affiliation(s)
- Bruna Panizzutti
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Chiara C Bortolasci
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Briana Spolding
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Srisaiyini Kidnapillai
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Timothy Connor
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Trang Tt Truong
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Zoe Sj Liu
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Damián Hernández
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Laura Gray
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Deakin University, Geelong, Australia
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Jee Hyun Kim
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Deakin University, Geelong, Australia
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Olivia M Dean
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Deakin University, Geelong, Australia
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Michael Berk
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Deakin University, Geelong, Australia
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
- Department of Psychiatry, Royal Melbourne Hospital, University of Melbourne, Parkville, Australia
- Centre of Youth Mental Health, University of Melbourne, Parkville, Australia
- Orygen Youth Health Research Centre, Parkville, Australia
| | - Ken Walder
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Deakin University, Geelong, Australia
| |
Collapse
|
4
|
Paribello P, Isayeva U, Pisanu C, Squassina A, Manchia M. Pharmacogenomics and response to lithium in bipolar disorder. Pharmacogenomics 2025; 25:689-706. [PMID: 39998910 PMCID: PMC11901374 DOI: 10.1080/14622416.2025.2470605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/19/2025] [Indexed: 02/27/2025] Open
Abstract
AIMS The present review explores the existing evidence on pharmacogenomic tests for prediction of lithium response in the treatment of bipolar disorder. We focused our research article on reports describing findings from genome-wide association studies, polygenic risk scores, and gene expression analyses associated with lithium response. METHODS We conducted a non-systematic review of studies from PubMed/Medline by using terms such as "pharmacogenomics," "GWAS," "gene expression," and "lithium response." Inclusion criteria focused on original research involving human subjects and assessing lithium response outcomes as well as in vitro studies. An extensive pearl-growing strategy was employed to further enlarge the scope of the piece by capitalizing on the knowledge of study authors on the subject. RESULTS The observed results, albeit promising, remain preliminary in terms of clinical relevance. Machine learning combining genetic and clinical data appears associated with moderate success in predicting lithium responsiveness. Gene expression studies and genome-wide association studies have helped identify possible targets of lithium and have the potential to support target-specific drug research. CONCLUSIONS Pharmacogenomics may support further discoveries in precision medicine further enlarging our understanding of the underlying mechanisms of lithium for its efficacy. However, clinical applications currently appear out of reach in the near future.
Collapse
Affiliation(s)
- Pasquale Paribello
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Ulker Isayeva
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Claudia Pisanu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
5
|
Chatzifrangkeskou M, Stanly T, Koennig D, Campos-Soares L, Eyres M, Hasson A, Perdiou A, Vendrell I, Fischer R, Das S, Gardner S, Go S, Futcher B, Newton A, Skourides P, Szele F, O’Neill E. ATR-hippo drives force signaling to nuclear F-actin and links mechanotransduction to neurological disorders. SCIENCE ADVANCES 2025; 11:eadr5683. [PMID: 39951537 PMCID: PMC11827640 DOI: 10.1126/sciadv.adr5683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025]
Abstract
The mechanical environment is sensed through cell-matrix contacts with the cytoskeleton, but how signals transit the nuclear envelope to affect cell fate decisions remains unknown. Nuclear actin coordinates chromatin motility during differentiation and genome maintenance, yet it remains unclear how nuclear actin responds to mechanical force. The DNA-damage kinase ataxia telangiectasia and Rad3-related protein (ATR) translocates to the nuclear envelope to protect the nucleus during cell motility or compression. Here, we show that ATR drives nuclear actin assembly via recruitment of Filamin-A to the inner nuclear membrane through binding of the hippo pathway scaffold and ATR substrate, RASSF1A. Moreover, we demonstrate how germline RASSF1 mutation disables nuclear mechanotransduction resulting in cerebral cortex thinning and associates with common psychological traits. Thus, defective mechanical-regulated pathways may contribute to complex neurological disorders.
Collapse
Affiliation(s)
- Maria Chatzifrangkeskou
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
- Department of Biological Sciences, University of Cyprus, P.O. Box 20537, 2109 Nicosia, Cyprus
| | - Tess Stanly
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Delia Koennig
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Luana Campos-Soares
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
- Department Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Michael Eyres
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Alexander Hasson
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Alexandra Perdiou
- Department of Biological Sciences, University of Cyprus, P.O. Box 20537, 2109 Nicosia, Cyprus
| | - Iolanda Vendrell
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Roman Fischer
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sayoni Das
- PrecisionLife, Bankside, Long Hanborough, Oxford OX29 8LJ, UK
| | - Steve Gardner
- PrecisionLife, Bankside, Long Hanborough, Oxford OX29 8LJ, UK
| | - Simei Go
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Ben Futcher
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Ashley Newton
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Paris Skourides
- Department of Biological Sciences, University of Cyprus, P.O. Box 20537, 2109 Nicosia, Cyprus
| | - Francis Szele
- Department Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Eric O’Neill
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
6
|
O'Callaghan LA, Blum CB, Powell K, Chess‐Williams R, McDermott C. From Psychiatry to Oncology: Exploring the Anti-Neoplastic Mechanisms of Aripiprazole and Its Potential Use in Cancer Treatment. Pharmacol Res Perspect 2025; 13:e70076. [PMID: 39939172 PMCID: PMC11821285 DOI: 10.1002/prp2.70076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/14/2025] Open
Abstract
Drug repurposing provides a cost-effective and time-saving approach to cancer therapy. Aripiprazole (ARI), a third-generation antipsychotic, has shown potential anticancer properties by modulating pathways central to tumor progression and resistance. This scoping review systematically examines evidence on ARI's anticancer effects, mechanisms of action, and translational potential. A systematic search of PubMed, EMBASE, SCOPUS, and Web of Science was conducted following PRISMA-ScR guidelines. Eligible studies included in vitro, in vivo, and clinical investigations. Data on cancer types, pathways, assays, and outcomes were extracted and synthesized to identify trends and gaps. Of 588 screened studies, 23 met inclusion criteria, spanning cancer types such as breast, colorectal, lung, and brain cancers. ARI modulates key pathways like PI3K/AKT/mTOR and Wnt/β-catenin, induces apoptosis through mitochondrial dysfunction and ER stress, and overcomes drug resistance by inhibiting P-glycoprotein activity and expression. It exhibits tumor-suppressive effects in vivo and synergizes with chemotherapy and radiotherapy. Retrospective population studies suggest ARI's prolactin-sparing properties may reduce the risk of hormone-sensitive cancers such as breast and endometrial cancer compared to antipsychotics with stronger dopamine receptor blockade. Additionally, ARI's ability to target multiple Hallmarks of Cancer highlights its promise as a repurposed anticancer agent. However, current evidence is primarily preclinical and observational, with limited clinical validation. Large-scale cohort studies and prospective trials are essential to confirm its efficacy and address translational challenges. By bridging these gaps, ARI could emerge as a valuable adjunctive therapy in oncology, leveraging its safety profile and versatility to address unmet needs in cancer treatment.
Collapse
Affiliation(s)
- Liam A. O'Callaghan
- Faculty of Health Sciences and MedicineBond UniversityRobinaQueenslandAustralia
| | - Ciara B. Blum
- School of Medicine and DentistryGriffith UniversitySouthportQueenslandAustralia
| | - Katie Powell
- Faculty of Health Sciences and MedicineBond UniversityRobinaQueenslandAustralia
| | - Russ Chess‐Williams
- Faculty of Health Sciences and MedicineBond UniversityRobinaQueenslandAustralia
| | - Catherine McDermott
- Faculty of Health Sciences and MedicineBond UniversityRobinaQueenslandAustralia
| |
Collapse
|
7
|
Liu ZSJ, Truong TTT, Bortolasci CC, Spolding B, Panizzutti B, Swinton C, Kim JH, Hernández D, Kidnapillai S, Gray L, Berk M, Dean OM, Walder K. The potential of baicalin to enhance neuroprotection and mitochondrial function in a human neuronal cell model. Mol Psychiatry 2024; 29:2487-2495. [PMID: 38503930 DOI: 10.1038/s41380-024-02525-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/21/2024]
Abstract
Baicalin is a flavone glycoside derived from flowering plants belonging to the Scutellaria genus. Previous studies have reported baicalin's anti-inflammatory and neuroprotective properties in rodent models, indicating the potential of baicalin in neuropsychiatric disorders where alterations in numerous processes are observed. However, the extent of baicalin's therapeutic effects remains undetermined in a human cell model, more specifically, neuronal cells to mimic the brain environment in vitro. As a proof of concept, we treated C8-B4 cells (murine cell model) with three different doses of baicalin (0.1, 1 and 5 μM) and vehicle control (DMSO) for 24 h after liposaccharide-induced inflammation and measured the levels of TNF-α in the medium by ELISA. NT2-N cells (human neuronal-like cell model) underwent identical baicalin treatment, followed by RNA extraction, genome-wide mRNA expression profiles and gene set enrichment analysis (GSEA). We also performed neurite outgrowth assays and mitochondrial flux bioanalysis (Seahorse) in NT2-N cells. We found that in C8-B4 cells, baicalin at ≥ 1 μM exhibited anti-inflammatory effects, lowering TNF-α levels in the cell culture media. In NT2-N cells, baicalin positively affected neurite outgrowth and transcriptionally up-regulated genes in the tricarboxylic acid cycle and the glycolysis pathway. Similarly, Seahorse analysis showed increased oxygen consumption rate in baicalin-treated NT2-N cells, an indicator of enhanced mitochondrial function. Together, our findings have confirmed the neuroprotective and mitochondria enhancing effects of baicalin in human-neuronal like cells. Given the increased prominence of mitochondrial mechanisms in diverse neuropsychiatric disorders and the paucity of mitochondrial therapeutics, this suggests the potential therapeutic application of baicalin in human neuropsychiatric disorders where these processes are altered.
Collapse
Affiliation(s)
- Zoe S J Liu
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, 3220, Australia.
| | - Trang T T Truong
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, 3220, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, 3220, Australia
| | - Briana Spolding
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, 3220, Australia
| | - Bruna Panizzutti
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, 3220, Australia
| | - Courtney Swinton
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, 3220, Australia
| | - Jee Hyun Kim
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, 3220, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, 3010, Australia
| | - Damián Hernández
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, 3220, Australia
| | - Srisaiyini Kidnapillai
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, 3220, Australia
| | - Laura Gray
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, 3220, Australia
| | - Michael Berk
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, 3220, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, 3010, Australia
| | - Olivia M Dean
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, 3220, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, 3010, Australia
| | - Ken Walder
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, 3220, Australia
| |
Collapse
|
8
|
Panizzutti B, Bortolasci CC, Spolding B, Kidnapillai S, Connor T, Martin SD, Truong TTT, Liu ZSJ, Gray L, Kowalski GM, McGee SL, Kim JH, Berk M, Walder K. Effects of antipsychotic drugs on energy metabolism. Eur Arch Psychiatry Clin Neurosci 2024; 274:1125-1135. [PMID: 38072867 DOI: 10.1007/s00406-023-01727-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/12/2023] [Indexed: 07/06/2024]
Abstract
Schizophrenia (SCZ) is a complex neuropsychiatric disorder associated with altered bioenergetic pathways and mitochondrial dysfunction. Antipsychotic medications, both first and second-generation, are commonly prescribed to manage SCZ symptoms, but their direct impact on mitochondrial function remains poorly understood. In this study, we investigated the effects of commonly prescribed antipsychotics on bioenergetic pathways in cultured neurons. We examined the impact of risperidone, aripiprazole, amisulpride, and clozapine on gene expression, mitochondrial bioenergetic profile, and targeted metabolomics after 24-h treatment, using RNA-seq, Seahorse XF24 Flux Analyser, and gas chromatography-mass spectrometry (GC-MS), respectively. Risperidone treatment reduced the expression of genes involved in oxidative phosphorylation, the tricarboxylic acid cycle, and glycolysis pathways, and it showed a tendency to decrease basal mitochondrial respiration. Aripiprazole led to dose-dependent reductions in various mitochondrial function parameters without significantly affecting gene expression. Aripiprazole, amisulpride and clozapine treatment showed an effect on the tricarboxylic acid cycle metabolism, leading to more abundant metabolite levels. Antipsychotic drug effects on mitochondrial function in SCZ are multifaceted. While some drugs have greater effects on gene expression, others appear to exert their effects through enzymatic post-translational or allosteric modification of enzymatic activity. Understanding these effects is crucial for optimising treatment strategies for SCZ. Novel therapeutic interventions targeting energy metabolism by post-transcriptional pathways might be more effective as these can more directly and efficiently regulate energy production.
Collapse
Affiliation(s)
- Bruna Panizzutti
- Deakin University, School of Medicine, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
| | - Chiara C Bortolasci
- Deakin University, School of Medicine, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
| | - Briana Spolding
- Deakin University, School of Medicine, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
| | - Srisaiyini Kidnapillai
- Deakin University, School of Medicine, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
| | - Timothy Connor
- Deakin University, School of Medicine, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
| | - Sheree D Martin
- Deakin University, School of Medicine, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
| | - Trang T T Truong
- Deakin University, School of Medicine, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
| | - Zoe S J Liu
- Deakin University, School of Medicine, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
| | - Laura Gray
- Deakin University, School of Medicine, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
| | - Greg M Kowalski
- Metabolic Research Unit, School of Medicine, Institute for Physical Activity and Nutrition, Waurn Ponds, Geelong, VIC, Australia
| | - Sean L McGee
- Deakin University, School of Medicine, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
| | - Jee Hyun Kim
- Deakin University, School of Medicine, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Michael Berk
- Deakin University, School of Medicine, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
- Barwon Health, University Hospital Geelong, Geelong, VIC, Australia
- Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia
- Orygen, The National Centre for Excellence in Youth Mental Health, Parkville, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Ken Walder
- Deakin University, School of Medicine, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia.
| |
Collapse
|
9
|
Bastawy EM, Eraslan IM, Voglsanger L, Suphioglu C, Walker AJ, Dean OM, Read JL, Ziemann M, Smith CM. Novel Insights into Changes in Gene Expression within the Hypothalamus in Two Asthma Mouse Models: A Transcriptomic Lung-Brain Axis Study. Int J Mol Sci 2024; 25:7391. [PMID: 39000495 PMCID: PMC11242700 DOI: 10.3390/ijms25137391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
Patients with asthma experience elevated rates of mental illness. However, the molecular links underlying such lung-brain crosstalk remain ambiguous. Hypothalamic dysfunction is observed in many psychiatric disorders, particularly those with an inflammatory component due to many hypothalamic regions being unprotected by the blood-brain barrier. To gain a better insight into such neuropsychiatric sequelae, this study investigated gene expression differences in the hypothalamus following lung inflammation (asthma) induction in mice, using RNA transcriptome profiling. BALB/c mice were challenged with either bacterial lipopolysaccharide (LPS, E. coli) or ovalbumin (OVA) allergens or saline control (n = 7 per group), and lung inflammation was confirmed via histological examination of postmortem lung tissue. The majority of the hypothalamus was micro-dissected, and total RNA was extracted for sequencing. Differential expression analysis identified 31 statistically significant single genes (false discovery rate FDR5%) altered in expression following LPS exposure compared to controls; however, none were significantly changed following OVA treatment, suggesting a milder hypothalamic response. When gene sets were examined, 48 were upregulated and 8 were downregulated in both asthma groups relative to controls. REACTOME enrichment analysis suggests these gene sets are involved in signal transduction metabolism, immune response and neuroplasticity. Interestingly, we identified five altered gene sets directly associated with neurotransmitter signaling. Intriguingly, many of these altered gene sets can influence mental health and or/neuroinflammation in humans. These findings help characterize the links between asthma-induced lung inflammation and the brain and may assist in identifying relevant pathways and therapeutic targets for future intervention.
Collapse
Affiliation(s)
- Eslam M Bastawy
- Faculty of Health, School of Medicine, Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong 3216, Australia
| | - Izel M Eraslan
- Faculty of Health, School of Medicine, Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong 3216, Australia
| | - Lara Voglsanger
- Faculty of Health, School of Medicine, Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong 3216, Australia
| | - Cenk Suphioglu
- Faculty of Science, Engineering and Built Environment, School of Life and Environmental Sciences, Deakin University, Geelong 3216, Australia
| | - Adam J Walker
- Faculty of Health, School of Medicine, Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong 3216, Australia
| | - Olivia M Dean
- Faculty of Health, School of Medicine, Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong 3216, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Melbourne 3052, Australia
| | - Justin L Read
- Faculty of Health, School of Medicine, Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong 3216, Australia
| | - Mark Ziemann
- Faculty of Science, Engineering and Built Environment, School of Life and Environmental Sciences, Deakin University, Geelong 3216, Australia
- Burnet Institute, Melbourne 3004, Australia
| | - Craig M Smith
- Faculty of Health, School of Medicine, Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong 3216, Australia
| |
Collapse
|
10
|
Shen HP, Dong X, Li ZB, Wu JZ, Zheng CM, Hu XJ, Qian C, Wang SP, Zhao YL, Li JC. Protein Profiles and Novel Molecular Biomarkers of Schizophrenia Based on 4D-DIA Proteomics. J Proteome Res 2024; 23:2376-2385. [PMID: 38856018 DOI: 10.1021/acs.jproteome.4c00040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Schizophrenia is a severe psychological disorder. The current diagnosis mainly relies on clinical symptoms and lacks laboratory evidence, which makes it very difficult to make an accurate diagnosis especially at an early stage. Plasma protein profiles of schizophrenia patients were obtained and compared with healthy controls using 4D-DIA proteomics technology. Furthermore, 79 DEPs were identified between schizophrenia and healthy controls. GO functional analysis indicated that DEPs were predominantly associated with responses to toxic substances and platelet aggregation, suggesting the presence of metabolic and immune dysregulation in patients with schizophrenia. KEGG pathway enrichment analysis revealed that DEPs were primarily enriched in the chemokine signaling pathway and cytokine receptor interactions. A diagnostic model was ultimately established, comprising three proteins, namely, PFN1, GAPDH and ACTBL2. This model demonstrated an AUC value of 0.972, indicating its effectiveness in accurately identifying schizophrenia. PFN1, GAPDH and ACTBL2 exhibit potential as biomarkers for the early detection of schizophrenia. The findings of our studies provide novel insights into the laboratory-based diagnosis of schizophrenia.
Collapse
Affiliation(s)
- Hui-Ping Shen
- Department of Psychiatry, Shaoxing Seventh People's Hospital, Shaoxing 312000, China
| | - Xiaotao Dong
- Major Disease Biomarker Research Laboratory, School of Basic Medical Science, Henan University, Kaifeng 475004, China
| | - Zhi-Bin Li
- Major Disease Biomarker Research Laboratory, School of Basic Medical Science, Henan University, Kaifeng 475004, China
| | - Jing-Zhu Wu
- Department of Psychiatry, Shaoxing Seventh People's Hospital, Shaoxing 312000, China
| | - Chun-Mei Zheng
- Department of Psychiatry, Shaoxing Seventh People's Hospital, Shaoxing 312000, China
| | - Xie-Jun Hu
- Department of Psychiatry, Shaoxing Seventh People's Hospital, Shaoxing 312000, China
| | - Chao Qian
- Department of Psychiatry, Shaoxing Seventh People's Hospital, Shaoxing 312000, China
| | - Sheng-Pang Wang
- Department of Psychiatry, Shaoxing Seventh People's Hospital, Shaoxing 312000, China
| | - Yu-Long Zhao
- Department of Psychiatry, Shaoxing Seventh People's Hospital, Shaoxing 312000, China
| | - Ji-Cheng Li
- Major Disease Biomarker Research Laboratory, School of Basic Medical Science, Henan University, Kaifeng 475004, China
- Institute of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
11
|
Del Casale A, Modesti MN, Gentile G, Guariglia C, Ferracuti S, Simmaco M, Borro M. Is the Hedgehog Pathway Involved in the Pathophysiology of Schizophrenia? A Systematic Review of Current Evidence of Neural Molecular Correlates and Perspectives on Drug Development. Curr Issues Mol Biol 2024; 46:5322-5336. [PMID: 38920990 PMCID: PMC11202070 DOI: 10.3390/cimb46060318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/09/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Among the pathophysiological correlates of schizophrenia, recent research suggests a potential role for the Hedgehog (Hh) signalling pathway, which has been traditionally studied in embryonic development and oncology. Its dysregulation may impact brain homeostasis, neuroplasticity, and potential involvement in neural processes. This systematic review provides an overview of the involvement of Hh signalling in the pathophysiology of schizophrenia and antipsychotic responses. We searched the PubMed and Scopus databases to identify peer-reviewed scientific studies focusing on Hh and schizophrenia, following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement, finally including eight studies, including three articles focused on patients with schizophrenia, two animal models of schizophrenia, two animal embryo studies, and one cellular differentiation study. The Hh pathway is crucial in the development of midbrain dopaminergic neurons, neuroplasticity mechanisms, regulating astrocyte phenotype and function, brain-derived neurotrophic factor expression, brain glutamatergic neural transmission, and responses to antipsychotics. Overall, results indicate an involvement of Hh in the pathophysiology of schizophrenia and antipsychotic responses, although an exiguity of studies characterises the literature. The heterogeneity between animal and human studies is another main limitation. Further research can lead to better comprehension and the development of novel personalised drug treatments and therapeutic interventions.
Collapse
Affiliation(s)
- Antonio Del Casale
- Department of Dynamic and Clinical Psychology and Health Studies, Faculty of Medicine and Psychology, Sapienza University of Rome, 00185 Rome, Italy;
- Unit of Psychiatry, Emergency and Admissions Department, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Martina Nicole Modesti
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, 00185 Rome, Italy
- Unit of Psychiatry, Mental Health Department, Santissimo Gonfalone Hospital, Local Health Service Roma 5, Monterotondo, 00015 Rome, Italy
| | - Giovanna Gentile
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
- Unit of Laboratory and Advanced Molecular Diagnostics, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Cecilia Guariglia
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, 00185 Rome, Italy
- Cognitive and Motor Rehabilitation and Neuroimaging Unit, Scientific Institute for Research, Hospitalization and Healthcare Fondazione Santa Lucia, 00179 Rome, Italy
| | - Stefano Ferracuti
- Department of Human Neuroscience, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00185 Rome, Italy;
- Unit of Risk Management, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Maurizio Simmaco
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
- Unit of Laboratory and Advanced Molecular Diagnostics, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Marina Borro
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
- Unit of Laboratory and Advanced Molecular Diagnostics, Sant’Andrea University Hospital, 00189 Rome, Italy
| |
Collapse
|
12
|
Chamera K, Curzytek K, Kamińska K, Trojan E, Leśkiewicz M, Tylek K, Regulska M, Basta-Kaim A. Insights into the Potential Impact of Quetiapine on the Microglial Trajectory and Inflammatory Response in Organotypic Cortical Cultures Derived from Rat Offspring. Biomedicines 2023; 11:biomedicines11051405. [PMID: 37239076 DOI: 10.3390/biomedicines11051405] [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: 03/09/2023] [Revised: 04/27/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Atypical antipsychotics currently constitute the first-line medication for schizophrenia, with quetiapine being one of the most commonly prescribed representatives of the group. Along with its specific affinity for multiple receptors, this compound exerts other biological characteristics, among which anti-inflammatory effects are strongly suggested. Simultaneously, published data indicated that inflammation and microglial activation could be diminished by stimulation of the CD200 receptor (CD200R), which takes place by binding to its ligand (CD200) or soluble CD200 fusion protein (CD200Fc). Therefore, in the present study, we sought to evaluate whether quetiapine could affect certain aspects of microglial activity, including the CD200-CD200R and CX3CL1-CX3CR1 axes, which are involved in the regulation of neuron-microglia interactions, as well as the expression of selected markers of the pro- and anti-inflammatory profile of microglia (Cd40, Il-1β, Il-6, Cebpb, Cd206, Arg1, Il-10 and Tgf-β). Concurrently, we examined the impact of quetiapine and CD200Fc on the IL-6 and IL-10 protein levels. The abovementioned aspects were investigated in organotypic cortical cultures (OCCs) prepared from the offspring of control rats (control OCCs) or those subjected to maternal immune activation (MIA OCCs), which is a widely implemented approach to explore schizophrenia-like disturbances in animals. The experiments were performed under basal conditions and after additional exposure to the bacterial endotoxin lipopolysaccharide (LPS), according to the "two-hit" hypothesis of schizophrenia. The results of our research revealed differences between control and MIA OCCs under basal conditions and in response to treatment with LPS in terms of lactate dehydrogenase and nitric oxide release as well as Cd200r, Il-1β, Il-6 and Cd206 expression. The additional stimulation with the bacterial endotoxin resulted in a notable change in the mRNA levels of pro- and anti-inflammatory microglial markers in both types of OCCs. Quetiapine diminished the influence of LPS on Il-1β, Il-6, Cebpb and Arg1 expression in control OCCs as well as on IL-6 and IL-10 levels in MIA OCCs. Moreover, CD200Fc reduced the impact of the bacterial endotoxin on IL-6 production in MIA OCCs. Thus, our results demonstrated that quetiapine, as well as the stimulation of CD200R by CD200Fc, beneficially affected LPS-induced neuroimmunological changes, including microglia-related activation.
Collapse
Affiliation(s)
- Katarzyna Chamera
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Katarzyna Curzytek
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Kinga Kamińska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Ewa Trojan
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Monika Leśkiewicz
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Kinga Tylek
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Magdalena Regulska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| |
Collapse
|
13
|
Abdelfattah AM, Abuelezz SA, Hendawy N, Negm EA, Nawishy SAEK, Khalil AMM. Sonic hedgehog pathway as a new target of atypical antipsychotics: Revisiting of amisulpride and aripiprazole effects in a rat model of schizophrenia. Life Sci 2023; 316:121366. [PMID: 36649751 DOI: 10.1016/j.lfs.2022.121366] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/22/2022] [Accepted: 12/31/2022] [Indexed: 01/16/2023]
Abstract
OBJECTIVES Schizophrenia is a chronic mental illness presented by cognitive deficits that precede its positive and negative symptoms. Sonic hedgehog (Shh)-pathway contributes to its pathophysiology. Shh has a role in neurogenesis as it regulates proliferation and survival of neural cells. In this study, effects of the anti-psychotics Amisulpride and/or Aripiprazole on the Shh-pathway and its relation to cognitive functions and neurogenesis in a rat model of schizophrenia were tested. METHODS 60 male Wistar rats were allocated into the following groups: control, socially isolated, amisulpride and/or aripiprazole-treated groups. Rats were then subjected to behavioral, biochemical, and histopathological tests to assess the impact of these drugs on Shh-pathway. KEY FINDINGS Cognitive-dysfunction was evidenced in socially isolated group in novel object, three-chamber, and Morris water maze tests, associated by disorganised Shh-pathway proteins levels concentrations, increased glial fibrillary acidic protein (GFAP)-stained astrocytes. Treated groups favorably reversed these changes evidenced by increased Shh, transmembrane patched-1 and smoothened, glioma-associated-oncogene (GLI)-1 levels, dopamine-1 receptors and brain derived neurotrophic factor, and decreased GLI-3 protein, GFAP immune reaction in astrocytes and inflammatory markers compared to socially isolated group. CONCLUSION Amisulpride and/or aripiprazole have a favorable role in turning on Shh-pathway with subsequent beneficial cognitive and neurogenesis effects.
Collapse
Affiliation(s)
- Ahmed M Abdelfattah
- Clinical Pharmacology Department, Faculty of Medicine, Port Said University, Cairo, Egypt.
| | - Sally A Abuelezz
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nevien Hendawy
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Eman A Negm
- Histology and Cell Biology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | |
Collapse
|
14
|
Aroke EN, Hobson JM, Ptacek T, Jackson P, Goodin BR. Genome-wide DNA methylation study identifies significant epigenomic changes associated with internalized stigma in adults with non-specific chronic low back pain. FRONTIERS IN PAIN RESEARCH 2022; 3:1021963. [PMID: 36518098 PMCID: PMC9742283 DOI: 10.3389/fpain.2022.1021963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/26/2022] [Indexed: 11/29/2022] Open
Abstract
Non-specific chronic low back pain (cLBP) represents a common musculoskeletal condition with no identifiable cause. It cannot be diagnosed with conventional neuroimaging techniques such as computerized tomography (CT). The diagnostic uncertainty that characterizes non-specific cLBP can lead to stigmatizing responses from others that can become internalized Among individuals with non-specific cLBP, internalized stigma is associated with greater pain intensity and disability. Yet, no study has examined the biological mechanism linking high internalized stigma to worse outcomes in individuals with non-specific cLBP. We aimed to identify differentially methylated loci (DML), enrichment pathways, and associated network interactions among individuals with non-specific cLBP experiencing low vs. high internalized stigma. We examined DNA methylation in whole blood samples from 48 adults, ages 19-85, using reduced representation bisulfite sequencing (RRBS). After controlling for age, sex, race, and multiple testing, differentially methylated loci (DML) differed in adults with low vs. high internalized stigma by at least 10% and q < 0.01 in 3,665 CpG sites: 2,280 hypomethylated and 1,385 hypermethylated. Gene ontology (GO) analyses of the annotated genes from these sites revealed significant enrichment of 274 biological processes, 29 cellular components, and 24 molecular functions (adjusted p < 0.05). The top enriched molecular functions regulate protein binding and DNA binding of transcription factor activity. Pathway analyses indicated that many functional genomic pathways, including Hippo Signaling, Melanogenesis, and Pathways in Cancer, were enriched with differentially methylated genes. Also, there was a significant interaction between relevance pathways such as P53, mTOR, PI3K-Akt, and Wnt signaling pathways. These pathways have previously been associated with neuroinflammation, neurodegeneration, and stress-related conditions. Thus, findings point to possible stress-induced DNAm changes as the link between high levels of internalized stigma and worse outcomes in adults with non-specific cLBP.
Collapse
Affiliation(s)
- Edwin N. Aroke
- School of Nursing, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Joanna M. Hobson
- Biobehavioral Pain Lab, Department of Psychology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Travis Ptacek
- Center for Clinical and Translational Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Pamela Jackson
- School of Nursing, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Burel R. Goodin
- Biobehavioral Pain Lab, Department of Psychology, University of Alabama at Birmingham, Birmingham, AL, United States
- Center for Addiction and Pain Prevention and Intervention (CAPPI), University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
15
|
Casas BS, Arancibia-Altamirano D, Acevedo-La Rosa F, Garrido-Jara D, Maksaev V, Pérez-Monje D, Palma V. It takes two to tango: Widening our understanding of the onset of schizophrenia from a neuro-angiogenic perspective. Front Cell Dev Biol 2022; 10:946706. [PMID: 36092733 PMCID: PMC9448889 DOI: 10.3389/fcell.2022.946706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Schizophrenia is a chronic debilitating mental disorder characterized by perturbations in thinking, perception, and behavior, along with brain connectivity deficiencies, neurotransmitter dysfunctions, and loss of gray brain matter. To date, schizophrenia has no cure and pharmacological treatments are only partially efficacious, with about 30% of patients describing little to no improvement after treatment. As in most neurological disorders, the main descriptions of schizophrenia physiopathology have been focused on neural network deficiencies. However, to sustain proper neural activity in the brain, another, no less important network is operating: the vast, complex and fascinating vascular network. Increasing research has characterized schizophrenia as a systemic disease where vascular involvement is important. Several neuro-angiogenic pathway disturbances have been related to schizophrenia. Alterations, ranging from genetic polymorphisms, mRNA, and protein alterations to microRNA and abnormal metabolite processing, have been evaluated in plasma, post-mortem brain, animal models, and patient-derived induced pluripotent stem cell (hiPSC) models. During embryonic brain development, the coordinated formation of blood vessels parallels neuro/gliogenesis and results in the structuration of the neurovascular niche, which brings together physical and molecular signals from both systems conforming to the Blood-Brain barrier. In this review, we offer an upfront perspective on distinctive angiogenic and neurogenic signaling pathways that might be involved in the biological causality of schizophrenia. We analyze the role of pivotal angiogenic-related pathways such as Vascular Endothelial Growth Factor and HIF signaling related to hypoxia and oxidative stress events; classic developmental pathways such as the NOTCH pathway, metabolic pathways such as the mTOR/AKT cascade; emerging neuroinflammation, and neurodegenerative processes such as UPR, and also discuss non-canonic angiogenic/axonal guidance factor signaling. Considering that all of the mentioned above pathways converge at the Blood-Brain barrier, reported neurovascular alterations could have deleterious repercussions on overall brain functioning in schizophrenia.
Collapse
|
16
|
Integrative Analyses of Transcriptomes to Explore Common Molecular Effects of Antipsychotic Drugs. Int J Mol Sci 2022; 23:ijms23147508. [PMID: 35886854 PMCID: PMC9325239 DOI: 10.3390/ijms23147508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/04/2022] [Accepted: 07/04/2022] [Indexed: 01/27/2023] Open
Abstract
There is little understanding of the underlying molecular mechanism(s) involved in the clinical efficacy of antipsychotics for schizophrenia. This study integrated schizophrenia-associated transcriptional perturbations with antipsychotic-induced gene expression profiles to detect potentially relevant therapeutic targets shared by multiple antipsychotics. Human neuronal-like cells (NT2-N) were treated for 24 h with one of the following antipsychotic drugs: amisulpride, aripiprazole, clozapine, risperidone, or vehicle controls. Drug-induced gene expression patterns were compared to schizophrenia-associated transcriptional data in post-mortem brain tissues. Genes regulated by each of four antipsychotic drugs in the reverse direction to schizophrenia were identified as potential therapeutic-relevant genes. A total of 886 genes were reversely expressed between at least one drug treatment (versus vehicle) and schizophrenia (versus healthy control), in which 218 genes were commonly regulated by all four antipsychotic drugs. The most enriched biological pathways include Wnt signaling and action potential regulation. The protein-protein interaction (PPI) networks found two main clusters having schizophrenia expression quantitative trait loci (eQTL) genes such as PDCD10, ANK2, and AKT3, suggesting further investigation on these genes as potential novel treatment targets.
Collapse
|
17
|
Liu ZSJ, Truong TTT, Bortolasci CC, Spolding B, Panizzutti B, Swinton C, Kim JH, Kidnapillai S, Richardson MF, Gray L, Dean OM, McGee SL, Berk M, Walder K. Effects of Psychotropic Drugs on Ribosomal Genes and Protein Synthesis. Int J Mol Sci 2022; 23:ijms23137180. [PMID: 35806181 PMCID: PMC9266764 DOI: 10.3390/ijms23137180] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/26/2022] [Accepted: 06/26/2022] [Indexed: 02/04/2023] Open
Abstract
Altered protein synthesis has been implicated in the pathophysiology of several neuropsychiatric disorders, particularly schizophrenia. Ribosomes are the machinery responsible for protein synthesis. However, there remains little information on whether current psychotropic drugs affect ribosomes and contribute to their therapeutic effects. We treated human neuronal-like (NT2-N) cells with amisulpride (10 µM), aripiprazole (0.1 µM), clozapine (10 µM), lamotrigine (50 µM), lithium (2.5 mM), quetiapine (50 µM), risperidone (0.1 µM), valproate (0.5 mM) or vehicle control for 24 h. Transcriptomic and gene set enrichment analysis (GSEA) identified that the ribosomal pathway was altered by these drugs. We found that three of the eight drugs tested significantly decreased ribosomal gene expression, whilst one increased it. Most changes were observed in the components of cytosolic ribosomes and not mitochondrial ribosomes. Protein synthesis assays revealed that aripiprazole, clozapine and lithium all decreased protein synthesis. Several currently prescribed psychotropic drugs seem to impact ribosomal gene expression and protein synthesis. This suggests the possibility of using protein synthesis inhibitors as novel therapeutic agents for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Zoe S. J. Liu
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong 3220, Australia; (Z.S.J.L.); (T.T.T.T.); (C.C.B.); (B.S.); (B.P.); (C.S.); (J.H.K.); (S.K.); (L.G.); (O.M.D.); (S.L.M.); (M.B.)
| | - Trang T. T. Truong
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong 3220, Australia; (Z.S.J.L.); (T.T.T.T.); (C.C.B.); (B.S.); (B.P.); (C.S.); (J.H.K.); (S.K.); (L.G.); (O.M.D.); (S.L.M.); (M.B.)
| | - Chiara C. Bortolasci
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong 3220, Australia; (Z.S.J.L.); (T.T.T.T.); (C.C.B.); (B.S.); (B.P.); (C.S.); (J.H.K.); (S.K.); (L.G.); (O.M.D.); (S.L.M.); (M.B.)
| | - Briana Spolding
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong 3220, Australia; (Z.S.J.L.); (T.T.T.T.); (C.C.B.); (B.S.); (B.P.); (C.S.); (J.H.K.); (S.K.); (L.G.); (O.M.D.); (S.L.M.); (M.B.)
| | - Bruna Panizzutti
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong 3220, Australia; (Z.S.J.L.); (T.T.T.T.); (C.C.B.); (B.S.); (B.P.); (C.S.); (J.H.K.); (S.K.); (L.G.); (O.M.D.); (S.L.M.); (M.B.)
| | - Courtney Swinton
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong 3220, Australia; (Z.S.J.L.); (T.T.T.T.); (C.C.B.); (B.S.); (B.P.); (C.S.); (J.H.K.); (S.K.); (L.G.); (O.M.D.); (S.L.M.); (M.B.)
| | - Jee Hyun Kim
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong 3220, Australia; (Z.S.J.L.); (T.T.T.T.); (C.C.B.); (B.S.); (B.P.); (C.S.); (J.H.K.); (S.K.); (L.G.); (O.M.D.); (S.L.M.); (M.B.)
- Florey Institute of Neuroscience and Mental Health, Parkville 3010, Australia
| | - Srisaiyini Kidnapillai
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong 3220, Australia; (Z.S.J.L.); (T.T.T.T.); (C.C.B.); (B.S.); (B.P.); (C.S.); (J.H.K.); (S.K.); (L.G.); (O.M.D.); (S.L.M.); (M.B.)
| | - Mark F. Richardson
- Genomics Centre, School of Life and Environmental Sciences, Deakin University, Burwood 3125, Australia;
| | - Laura Gray
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong 3220, Australia; (Z.S.J.L.); (T.T.T.T.); (C.C.B.); (B.S.); (B.P.); (C.S.); (J.H.K.); (S.K.); (L.G.); (O.M.D.); (S.L.M.); (M.B.)
- Florey Institute of Neuroscience and Mental Health, Parkville 3010, Australia
| | - Olivia M. Dean
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong 3220, Australia; (Z.S.J.L.); (T.T.T.T.); (C.C.B.); (B.S.); (B.P.); (C.S.); (J.H.K.); (S.K.); (L.G.); (O.M.D.); (S.L.M.); (M.B.)
- Florey Institute of Neuroscience and Mental Health, Parkville 3010, Australia
| | - Sean L. McGee
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong 3220, Australia; (Z.S.J.L.); (T.T.T.T.); (C.C.B.); (B.S.); (B.P.); (C.S.); (J.H.K.); (S.K.); (L.G.); (O.M.D.); (S.L.M.); (M.B.)
| | - Michael Berk
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong 3220, Australia; (Z.S.J.L.); (T.T.T.T.); (C.C.B.); (B.S.); (B.P.); (C.S.); (J.H.K.); (S.K.); (L.G.); (O.M.D.); (S.L.M.); (M.B.)
- Florey Institute of Neuroscience and Mental Health, Parkville 3010, Australia
| | - Ken Walder
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong 3220, Australia; (Z.S.J.L.); (T.T.T.T.); (C.C.B.); (B.S.); (B.P.); (C.S.); (J.H.K.); (S.K.); (L.G.); (O.M.D.); (S.L.M.); (M.B.)
- Correspondence:
| |
Collapse
|
18
|
Truong TTT, Bortolasci CC, Spolding B, Panizzutti B, Liu ZSJ, Kidnapillai S, Richardson M, Gray L, Smith CM, Dean OM, Kim JH, Berk M, Walder K. Co-Expression Networks Unveiled Long Non-Coding RNAs as Molecular Targets of Drugs Used to Treat Bipolar Disorder. Front Pharmacol 2022; 13:873271. [PMID: 35462908 PMCID: PMC9024411 DOI: 10.3389/fphar.2022.873271] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/24/2022] [Indexed: 12/13/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) may play a role in psychiatric diseases including bipolar disorder (BD). We investigated mRNA-lncRNA co-expression patterns in neuronal-like cells treated with widely prescribed BD medications. The aim was to unveil insights into the complex mechanisms of BD medications and highlight potential targets for new drug development. Human neuronal-like (NT2-N) cells were treated with either lamotrigine, lithium, quetiapine, valproate or vehicle for 24 h. Genome-wide mRNA expression was quantified for weighted gene co-expression network analysis (WGCNA) to correlate the expression levels of mRNAs with lncRNAs. Functional enrichment analysis and hub lncRNA identification was conducted on key co-expressed modules associated with the drug response. We constructed lncRNA-mRNA co-expression networks and identified key modules underlying these treatments, as well as their enriched biological functions. Processes enriched in key modules included synaptic vesicle cycle, endoplasmic reticulum-related functions and neurodevelopment. Several lncRNAs such as GAS6-AS1 and MIR100HG were highlighted as driver genes of key modules. Our study demonstrates the key role of lncRNAs in the mechanism(s) of action of BD drugs. Several lncRNAs have been suggested as major regulators of medication effects and are worthy of further investigation as novel drug targets to treat BD.
Collapse
Affiliation(s)
- Trang TT. Truong
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Chiara C. Bortolasci
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Briana Spolding
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Bruna Panizzutti
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Zoe SJ. Liu
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Srisaiyini Kidnapillai
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Mark Richardson
- Genomics Centre, School of Life and Environmental Sciences, Deakin University, Burwood, VIC, Australia
| | - Laura Gray
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Craig M. Smith
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Olivia M. Dean
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental health and Clinical Translation, Deakin University, Geelong, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Jee Hyun Kim
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental health and Clinical Translation, Deakin University, Geelong, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Michael Berk
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental health and Clinical Translation, Deakin University, Geelong, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| | - Ken Walder
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
19
|
Panizzutti B, Bortolasci CC, Spolding B, Kidnapillai S, Connor T, Richardson MF, Truong TTT, Liu ZSJ, Gray L, Kim JH, Dean OM, Berk M, Walder K. Biological Mechanism(s) Underpinning the Association between Antipsychotic Drugs and Weight Gain. J Clin Med 2021; 10:4095. [PMID: 34575210 PMCID: PMC8467356 DOI: 10.3390/jcm10184095] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022] Open
Abstract
Weight gain and consequent metabolic alterations are common side-effects of many antipsychotic drugs. Interestingly, several studies have suggested that improvement in symptoms and adverse metabolic effects are correlated. We used next generation sequencing data from NT-2 (human neuronal) cells treated with aripiprazole, amisulpride, risperidone, quetiapine, clozapine, or vehicle control, and compared with the Pillinger P-score (ranked from 0 to 1, indicating greater increase in weight gain and related metabolic parameters) to identify the genes most associated with the drugs' propensity to cause weight gain. The top 500 genes ranked for their correlation with the drugs' propensity to cause weight gain were subjected to pathway analysis using DAVID (NIH). We further investigated transcription factors (TFs) that are more likely to regulate the genes involved in these processes using the prediction tool of key TFs from TRRUST. The results suggest an enrichment for genes involved in lipid biosynthesis and metabolism, which are of interest for mechanisms underpinning weight-gain. The list of genes involved in the lipid pathways that correlated with weight gain was enriched for genes transcriptionally regulated by SREBF1 and SREBF2. Furthermore, quetiapine significantly increased the expression of SREBF1 and SREBF2 in NT-2 cells. Our results suggest that the effects of these antipsychotic drugs on lipid metabolism may be mediated, at least in part, via regulation of SREBF1/SREBF2 expression, with evidence of a direct effect of quetiapine on the expression of SREBF1/2. The effects of antipsychotic drugs on lipid metabolism may influence white matter structure (therapeutic effect) and the risk of weight gain, lipid disturbances, and, consequently, metabolic syndrome (adverse effects). Understanding the different molecular effects of these drugs could inform a personalized medicine approach in treating patients with schizophrenia.
Collapse
Affiliation(s)
- Bruna Panizzutti
- Institute for Innovation in Physical and Mental Health and Clinical Translation, IMPACT, School of Medicine, Deakin University, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Chiara C. Bortolasci
- Institute for Innovation in Physical and Mental Health and Clinical Translation, IMPACT, School of Medicine, Deakin University, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Briana Spolding
- Institute for Innovation in Physical and Mental Health and Clinical Translation, IMPACT, School of Medicine, Deakin University, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Srisaiyini Kidnapillai
- Institute for Innovation in Physical and Mental Health and Clinical Translation, IMPACT, School of Medicine, Deakin University, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Timothy Connor
- Institute for Innovation in Physical and Mental Health and Clinical Translation, IMPACT, School of Medicine, Deakin University, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Mark F. Richardson
- Genomics Centre, School of Life and Environmental Sciences, Deakin University, Geelong 3220, Australia;
| | - Trang T. T. Truong
- Institute for Innovation in Physical and Mental Health and Clinical Translation, IMPACT, School of Medicine, Deakin University, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Zoe S. J. Liu
- Institute for Innovation in Physical and Mental Health and Clinical Translation, IMPACT, School of Medicine, Deakin University, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Laura Gray
- Institute for Innovation in Physical and Mental Health and Clinical Translation, IMPACT, School of Medicine, Deakin University, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Australia
| | - Jee Hyun Kim
- Institute for Innovation in Physical and Mental Health and Clinical Translation, IMPACT, School of Medicine, Deakin University, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Australia
| | - Olivia M. Dean
- Institute for Innovation in Physical and Mental Health and Clinical Translation, IMPACT, School of Medicine, Deakin University, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Australia
| | - Michael Berk
- Institute for Innovation in Physical and Mental Health and Clinical Translation, IMPACT, School of Medicine, Deakin University, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Australia
- Department of Psychiatry, Royal Melbourne Hospital, University of Melbourne, Parkville 3052, Australia
- Centre of Youth Mental Health, University of Melbourne, Parkville 3052, Australia
- Orygen Youth Health Research Centre, Parkville 3052, Australia
| | - Ken Walder
- Institute for Innovation in Physical and Mental Health and Clinical Translation, IMPACT, School of Medicine, Deakin University, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| |
Collapse
|