1
|
Liu Y, Chen C, Zhao Y, Li M, Gao Y, Yan B, Jing Y, Zhang B, Li J. Transcriptional characteristics of human brain alterations in major depressive disorder: A systematic review. Psychoneuroendocrinology 2025; 177:107472. [PMID: 40288014 DOI: 10.1016/j.psyneuen.2025.107472] [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: 11/19/2024] [Revised: 03/05/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025]
Abstract
Many patients with major depressive disorder (MDD) experience limited treatment effectiveness due to an incomplete understanding of its neurobiological underpinnings. This review integrates neuroimaging and genetic data to examine structural and functional brain changes in MDD, alongside their genetic bases. A PRISMA-guided systematic review of imaging transcriptomics over the past decade was conducted using PubMed and Web of Science. Studies included MRI scans of both MDD patients and healthy controls, as well as brain-wide gene expression data, excluding those that were purely meta-analytical, lacked spatial correlations, or involved transdiagnostic analyses. Of the 206 studies reviewed, 20 met the inclusion criteria. Consistent patterns across studies reveal that key biological processes-such as synaptic signaling, calcium ion binding, neurodevelopment, immune regulation, and neurotransmitter transport-play a central role in brain alterations associated with MDD. Additionally, our findings suggest that electroconvulsive therapy (ECT) may alleviate symptoms by modulating these shared pathways. This review underscores the link between brain changes in MDD and specific gene expression profiles, offering insights that could inform more targeted therapeutic approaches.
Collapse
Affiliation(s)
- Yuan Liu
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Chengfeng Chen
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongping Zhao
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Meijuan Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Ying Gao
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Bo Yan
- Department of Geriatrics, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin 300052, China
| | - Yifan Jing
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Bin Zhang
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China.
| | - Jie Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin 300222, China.
| |
Collapse
|
2
|
Liang S, Gao Y, Palaniyappan L, Song XM, Zhang T, Han JF, Tan ZL, Li T. Transcriptional substrates of cortical thickness alterations in anhedonia of major depressive disorder. J Affect Disord 2025; 379:118-126. [PMID: 40044088 DOI: 10.1016/j.jad.2025.03.003] [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: 08/12/2024] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Anhedonia is a core symptom of major depressive disorder (MDD), which has been shown to be associated with abnormalities in cortical morphology. However, the correlation between cortical thickness (CT) changes with anhedonia in MDD and gene expression remains unclear. METHODS We investigated the link between brain-wide gene expression and CT correlates of anhedonia in individuals with MDD, using 7 Tesla neuroimaging and a publicly available transcriptomic dataset. The interest-activity score was used to evaluation MDD with high anhedonia (HA) and low anhedonia (LA). Nineteen patients with HA, nineteen patients with LA, and twenty healthy controls (HC) were enrolled. We investigated CT alterations of anhedonia subgroups relative to HC and related cortical gene expression, enrichment and specific cell types. We further used Neurosynth and von Economo-Koskinas atlas to assess the meta-analytic cognitive functions and cytoarchitectural variation associated with anhedonia-related cortical changes. RESULTS Both patient subgroups exhibited widespread CT reduction, with HA manifesting more pronounced changes. Gene expression related to anhedonia had significant spatial correlations with CT differences. Transcriptional signatures related to anhedonia-associated cortical thinning were connected to mitochondrial dysfunction and enriched in adipogenesis, oxidative phosphorylation, mTORC1 signaling pathways, involving neurons, astrocytes, and oligodendrocytes. These CT alterations were significantly correlated with meta-analytic terms involving somatosensory processing and pain perception. HA had reduced CT within the somatomotor and ventral attention networks, and in agranular cortical regions. LIMITATIONS These include measuring anhedonia using interest-activity score and employing a cross-sectional design. CONCLUSIONS This study sheds light on the molecular basis underlying gene expression associated with anhedonia in MDD, suggesting directions for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Sugai Liang
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, China
| | - Yuan Gao
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, China; Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Lena Palaniyappan
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Quebec H4H1R3, Canada.; Department of Psychiatry, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A5C1, Canada; Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A5K8, Canada
| | - Xue-Mei Song
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, China; Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Tian Zhang
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, China
| | - Jin-Fang Han
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, China
| | - Zhong-Lin Tan
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, China.
| | - Tao Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 310000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310063, China.
| |
Collapse
|
3
|
Ma Z, Zhang R, Yuan D, Yu C, Baranova A, Cao H, Zhang F. Association of branched-chain amino acids with major depressive disorder: A bidirectional Mendelian randomization study. J Affect Disord 2025; 379:467-472. [PMID: 40081595 DOI: 10.1016/j.jad.2025.03.032] [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: 11/30/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Recent studies have linked branched-chain amino acids (BCAAs) metabolism with the risk of major depressive disorder (MDD). However, it is unclear whether associations of plasma BCAA levels with MDD are causal or driven by reverse causality. METHODS Mendelian randomization (MR) was used to investigate the causal association of genetically determined BCAA levels with the risk of MDD. The large genome-wide association study (GWAS) datasets on plasma BCAA levels (n = 115,051) were obtained from the UK Biobank. The summary GWAS dataset for MDD was obtained from the Psychiatric Genomics Consortium (n = 1,035,760). We applied the inverse variance-weighted (IVW) method to explore the causal relationships between BCAA levels and MDD, followed by multiple pleiotropy and heterogeneity tests. RESULTS Our results demonstrated that genetically determined circulating total BCAAs (odds ratio (OR): 1.05, 95 % confidence interval (CI): 1.01-1.10, P = 0.016), leucine (OR: 1.06, 95 % CI: 1.02-1.11, P = 7.22 × 10-3), and isoleucine (OR: 1.08, 95 % CI: 1.01-1.16, P = 0.032) levels were associated with an increased risk of MDD. There was suggestive evidence supporting the causal effect of valine levels on MDD (OR: 1.04, 95 % CI: 1.00-1.08, P = 0.075). Bidirectional MR analysis did not provide evidence of reverse causality. CONCLUSIONS We report evidence supporting the causal role of BCAAs in the development of MDD. This study offers new insights into the mechanisms and treatment of MDD.
Collapse
Affiliation(s)
- Zhongxuan Ma
- Department of Pharmacy, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Ruyi Zhang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing 210000, Jiangsu Province, China
| | - Daorui Yuan
- Department of Geriatric Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Chuanyong Yu
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Ancha Baranova
- School of Systems Biology, George Mason University, Manassas, VA 20110, USA; Research Centre for Medical Genetics, Moscow 115478, Russia
| | - Hongbao Cao
- School of Systems Biology, George Mason University, Manassas, VA 20110, USA
| | - Fuquan Zhang
- Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing 210029, Jiangsu, China; Institute of Neuropsychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| |
Collapse
|
4
|
Liu C, Zhang C, Glatt SJ. Psychiatric Genomics 2025: State of the Art and the Path Forward. Psychiatr Clin North Am 2025; 48:217-240. [PMID: 40348414 DOI: 10.1016/j.psc.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Psychiatric genetics has evolved from candidate-gene studies to whole-genome sequencing efforts. With hundreds of disease-associated loci now identified, functional interpretation of the associated loci becomes the critical next step toward translational applications. The article discusses achievements, challenges, and opportunities in psychiatric genomics associated with complexity and heterogeneity. Brain expression quantitative trait loci, single-cell ribonucleic acid-sequence, and functional genomics technologies are highlighted. It also covers newly developed techniques with improved spatiotemporal resolution, quality and sensitivity, coupled with advanced analytical methods and artificial intelligence. The power of collaborative research and inclusion of diverse populations will ensure a bright future for precision psychiatry.
Collapse
Affiliation(s)
- Chunyu Liu
- Department of Psychiatry, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA.
| | - Chunling Zhang
- Department of Neuroscience & Physiology, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA
| | - Stephen J Glatt
- Department of Psychiatry, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA
| |
Collapse
|
5
|
Madeira MM, Hage Z, Kokkosis AG, Nnah K, Guzman R, Schappell LE, Koliatsis D, Resutov E, Nadkarni NA, Rahme GJ, Tsirka SE. Oligodendroglia Are Primed for Antigen Presentation in Response to Chronic Stress-Induced Microglial-Derived Inflammation. Glia 2025; 73:1130-1147. [PMID: 39719686 PMCID: PMC12014386 DOI: 10.1002/glia.24661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/26/2024]
Abstract
Chronic stress is a major contributor to the development of major depressive disorder, one of the leading causes of disability worldwide. Using a model of repeated social defeat stress in mice, we and others have reported that neuroinflammation plays a dynamic role in the development of behavioral deficits consistent with social avoidance and impaired reward responses. Animals susceptible to the model also exhibit hypomyelination in the medial prefrontal cortex, indicative of changes in the differentiation pathway of cells of the oligodendroglial lineage (OLN). We computationally confirmed the presence of immune oligodendrocytes, a population of OLN cells, which express immune markers and myelination deficits. In the current study, we report that microglia are necessary to induce expression of antigen presentation markers (and other immune markers) on oligodendroglia. We further associate the appearance of these markers with changes in the OLN and confirm that microglial changes precede OLN changes. Using co-cultures of microglia and OLN, we show that under inflammatory conditions the processes of phagocytosis and expression of MHCII are linked, suggesting potential priming for antigen presentation by OLN cells. Our findings provide insights into the nature of these OLN cells with immune capabilities, their obligatory interaction with microglia, and identify them as a potential cellular contributor to the pathological manifestations of psychosocial stress.
Collapse
Affiliation(s)
- Miguel M. Madeira
- Molecular and Cellular Pharmacology Program
- Scholars in Biomedical Sciences Program
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Zachary Hage
- Molecular and Cellular Pharmacology Program
- Scholars in Biomedical Sciences Program
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Alexandros G. Kokkosis
- Molecular and Cellular Pharmacology Program
- Scholars in Biomedical Sciences Program
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Kimberly Nnah
- Scholars in Biomedical Sciences Program
- Program in Neuroscience
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Ryan Guzman
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Laurel E. Schappell
- Molecular and Cellular Pharmacology Program
- Medical Scientist Training Program
- Department of Neurology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Dimitris Koliatsis
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Emran Resutov
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Neil A. Nadkarni
- Molecular and Cellular Pharmacology Program
- Department of Neurology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Gilbert J. Rahme
- Molecular and Cellular Pharmacology Program
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Stella E. Tsirka
- Molecular and Cellular Pharmacology Program
- Scholars in Biomedical Sciences Program
- Program in Neuroscience
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
6
|
Dong X, Zhang H, Chen M, Zhong W, Wang B. Genetically supported causality between gut microbiota, prefrontal cortex and depression: insights from multiple Mendelian randomization, multi-omics analysis, and network pharmacology. J Affect Disord 2025:119458. [PMID: 40419156 DOI: 10.1016/j.jad.2025.119458] [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: 08/12/2024] [Revised: 05/18/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND Gut microbiota (GM) alterations have been implicated in depression, potentially mediated via changes in the prefrontal cortex (PFC). However, the causal relationship and underlying biological mechanisms remain unclear. METHODS Summary statistics from genome-wide association studies (GWAS) of 473 GM (n = 5959), depression (43,280 cases, 329,192 controls), and 3935 brain imaging-derived phenotypes (IDPs) (n = 33,224) were utilized. Univariable Mendelian randomization (MR) analyses examined the associations between (i) GM and depression, (ii) depression and GM, (iii) brain IDPs and depression, and (iv) GM and PFC-related IDPs. Multivariable MR (MVMR) assessed whether PFC structural phenotypes mediated the effect of GM on depression risk. Summary-data-based MR (SMR) identified potential genes and therapeutic targets. Single-cell RNA sequencing and bulk RNA data from GEO database about PFC tissue of depression patients were analyzed for biomarker expression and regulatory mechanisms. Target genes were validated in mice model through behavioral tests, RT-qPCR, and immunofluorescence analyses. Drug prediction and molecular docking explored potential therapeutic targets and traditional Chinese medicine. RESULTS The analysis identified 15 positive and 16 negative causal effects between GM and depression. Reverse MR analysis revealed 36 GM traits with reverse causality to depression. After excluding results with heterogeneity and pleiotropy, 11 PFC-related IDPs exhibited causal relationships with depression. MVMR adjustment for genetically predicted GM levels showed that the effects of PFC-related IDPs remained significant, indicating mediation of GM's effect on depression. Integration of GWAS and eQTL data identified 14 genes passing the SMR test. Differential expression of these genes was observed across cell types and between patients with depression and healthy controls. FARS2 and SLC25A21 were identified as potential therapeutic targets for depression, with protective effects shown in MR analysis (FARS2: OR: 0.892, 95 % CI: 0.813-0.971, P = 0.005; SLC25A21: OR: 0.924, 95 % CI: 0.851-0.997, P = 0.035). Reduced expression of FARS2 and SLC25A21 was validated in CUMS induced mice, along with depressive-like behaviors confirmed through behavioral tests. Molecular docking predicted Betanidin, Aloenin B, Rehmannioside D, and Vicenin-2 as potential drugs targeting both FARS2 and SLC25A21. CONCLUSION This study supports causal relationships between specific GM and depression, with PFC structures potentially mediating these effects. The findings provide insights into personalized depression treatment strategies and identify promising candidate targets and drugs for future research.
Collapse
Affiliation(s)
- Xiaoqian Dong
- Nankai University, Tianjin 300071, China; Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Hao Zhang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Mengshi Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China.
| | - Bangmao Wang
- Nankai University, Tianjin 300071, China; Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China.
| |
Collapse
|
7
|
Teng T, Wu Q, Yin B, Zhang J, Li X, Zhang L, Zhou X, Xie P. Single-Nucleus Transcriptomics of the Nucleus Accumbens Reveals Cell-Type-Specific Dysregulation in Adolescent Macaques with Depressive-Like Behaviors. Neurosci Bull 2025:10.1007/s12264-025-01412-5. [PMID: 40399551 DOI: 10.1007/s12264-025-01412-5] [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: 09/04/2024] [Accepted: 01/08/2025] [Indexed: 05/23/2025] Open
Abstract
Adolescent depression is increasingly recognized as a serious mental health disorder with distinct clinical and molecular features. Using single-nucleus RNA sequencing, we identified cell-specific transcriptomic changes in the nucleus accumbens (NAc), particularly in astrocytes, of adolescent macaques exhibiting depressive-like behaviors. The level of diacylglycerol kinase beta was significantly reduced in neurons and glial cells of depressed macaques, while FKBP5 levels increased in glial cells. Disruption of GABAergic synapses and disruption of D-glutamine and D-glutamate metabolism were linked to depressive phenotypes in medium spiny neurons (MSNs) and subtypes of astrocytes. Communication pathways between astrocytes and D1/D2-MSNs were also disrupted, involving factors like bone morphogenetic protein-6 and Erb-B2 receptor tyrosine kinase-4. Bulk transcriptomic and proteomic analyses corroborated these findings, and FKBP5 upregulation was confirmed by qRT-PCR, western blotting, and immunofluorescence in the NAc of rats and macaques with chronic unpredictable mild stress. Our results highlight the specific roles of different cell types in adolescent depression in the NAc, offering potential targets for new antidepressant therapies.
Collapse
Affiliation(s)
- Teng Teng
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400014, China
- Department of Psychiatry, Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Qingyuan Wu
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400014, China
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, 404000, China
| | - Bangmin Yin
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400014, China
- Department of Psychiatry, Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Jushuang Zhang
- Department of Psychiatry, Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Xuemei Li
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400014, China
- Department of Psychiatry, Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Lige Zhang
- Department of Psychiatry, Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Xinyu Zhou
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Department of Psychiatry, Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China.
| | - Peng Xie
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
8
|
Niu RZ, Feng WQ, Chen L, Bao TH. Single-Cell Transcriptomic Profiling Reveals Regional Differences in the Prefrontal and Entorhinal Cortex of Alzheimer's Disease Brain. Int J Mol Sci 2025; 26:4841. [PMID: 40429980 DOI: 10.3390/ijms26104841] [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: 03/17/2025] [Revised: 05/02/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Previous studies have largely overlooked cellular differential alterations across differentially affected brain regions in both disease mechanisms and therapeutic development of Alzheimer's disease (AD). This study aimed to compare the differential cellular and transcriptional changes in the prefrontal cortex (PFC) and entorhinal cortex (EC) of AD patients through an integrated single-cell transcriptomic analysis. We integrated three single-cell RNA sequencing (scRNA-seq) datasets comprising PFC and EC samples from AD patients and age-matched healthy controls. A total of 124,658 nuclei and 31 cell clusters were obtained and classified into eight major cell types, with EC exhibiting much more pronounced transcriptional alterations than PFC. Through network analysis, we pinpointed hub regulatory genes that form interconnected networks driving AD pathogenesis, findings validated by RT-qPCR showing more pronounced expression changes in EC versus PFC of AD mice. Moreover, dysregulation of the LINC01099-associated regulatory networks in the PFC and EC, showing correlation with AD progression, may present new therapeutic targets for AD. Together, these results suggest that effective AD biomarkers and therapeutic strategies may require simultaneous, precise targeting of specific cell populations across multiple brain regions.
Collapse
Affiliation(s)
- Rui-Ze Niu
- Mental Health Center, Kunming Medical University, No. 733, Chuanjin Road, Panlong District, Kunming 650034, China
| | - Wan-Qing Feng
- Laboratory Zoology Department, Kunming Medical University, Kunming 650034, China
| | - Li Chen
- Mental Health Center, Kunming Medical University, No. 733, Chuanjin Road, Panlong District, Kunming 650034, China
| | - Tian-Hao Bao
- Mental Health Center, Kunming Medical University, No. 733, Chuanjin Road, Panlong District, Kunming 650034, China
| |
Collapse
|
9
|
Poggi G, Treccani G, von der Bey M, Tanti A, Schmeisser MJ, Müller M. Canonical and non-canonical roles of oligodendrocyte precursor cells in mental disorders. NPJ MENTAL HEALTH RESEARCH 2025; 4:19. [PMID: 40374740 DOI: 10.1038/s44184-025-00133-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/29/2025] [Indexed: 05/18/2025]
Abstract
Psychiatric research has shifted from a neuroncentric view to understanding mental disorders as disturbances of heterogeneous brain networks. Oligodendrocyte precursor cells (OPCs)- actively involved in the modulation of neuronal functions - are altered in psychiatric patients, but the extent and related consequences are unclear. This review explores canonical and non-canonical OPC-related pathways in schizophrenia, bipolar disorder, post-traumatic stress disorder, and depression in humans, highlighting potential mechanisms shared across diagnostic entities.
Collapse
Affiliation(s)
- Giulia Poggi
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| | - Giulia Treccani
- Department of Systemic Neuroscience Institute of Anatomy and Cell Biology, Philipps Universität Marburg, Marburg, Germany
| | - Martina von der Bey
- Molecular and Translational Neuroscience, Department of Neurology, University Hospital Ulm, Ulm, Germany
| | - Arnaud Tanti
- Inserm, UMR 1253, iBrain, Université de Tours, Tours, France
| | - Michael J Schmeisser
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Focus Program Translational Neurosciences, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Marianne Müller
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Leibniz Institute for Resilience Research, Mainz, Germany
| |
Collapse
|
10
|
Wan M, Wang W, He M, Yang S, Feng Y, Luo Y. Cubebin alleviates chronic stress-induced depression-like behavior in mice by regulating the gut microbiome. Eur J Pharmacol 2025; 994:177384. [PMID: 39956262 DOI: 10.1016/j.ejphar.2025.177384] [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: 10/14/2024] [Revised: 01/27/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
The gut-brain axis is dysregulated as a consequence of alterations in the gut microbiota. These alterations increase toxic microbial metabolites, endotoxemia, and the release of immune mediators and contribute to the development of depression. Cubebin is a dibenzyl butyrolactone lignan, and its stem is also known as Agaru in Tibetan areas, it is commonly used as a sedative and tranquilizing medicine. This study aimed to investigate the effects of cubebin on chronic stress-induced depression-like behavior in mice. Cubebin was observed to mitigate depressive-like behavior in chronic unpredictable mild stress (CUMS) mice, influence the restoration of their cerebral cortex and hippocampal tissue morphology, and enhance the abundance of relevant intestinal flora in depression model mice, particularly by decreasing the abundance of Clostridium, Dorea, and Ruminococcus. The final protein function expression was normalized by regulating depression-related metabolic pathways. Concomitantly, the concentrations of neurotransmitters serotonin (5-HT), norepinephrine (NE), and dopamine (DA) in the brains of mice in the model group were enhanced, and their depressive symptoms were mitigated. Our study findings suggest that cubebin may ameliorate CUMS-induced depression in mice by modulating the microbe-gut-brain axis, elucidating the key effect of gut metabolites on depressive symptoms.
Collapse
Affiliation(s)
- Mengqiang Wan
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
| | - Wei Wang
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
| | - Mingzhen He
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
| | - Shilin Yang
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
| | - Yulin Feng
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
| | - Yingying Luo
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
11
|
Nano PR, Fazzari E, Azizad D, Martija A, Nguyen CV, Wang S, Giang V, Kan RL, Yoo J, Wick B, Haeussler M, Bhaduri A. Integrated analysis of molecular atlases unveils modules driving developmental cell subtype specification in the human cortex. Nat Neurosci 2025; 28:949-963. [PMID: 40259073 PMCID: PMC12081304 DOI: 10.1038/s41593-025-01933-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/27/2025] [Indexed: 04/23/2025]
Abstract
Human brain development requires generating diverse cell types, a process explored by single-cell transcriptomics. Through parallel meta-analyses of the human cortex in development (seven datasets) and adulthood (16 datasets), we generated over 500 gene co-expression networks that can describe mechanisms of cortical development, centering on peak stages of neurogenesis. These meta-modules show dynamic cell subtype specificities throughout cortical development, with several developmental meta-modules displaying spatiotemporal expression patterns that allude to potential roles in cell fate specification. We validated the expression of these modules in primary human cortical tissues. These include meta-module 20, a module elevated in FEZF2+ deep layer neurons that includes TSHZ3, a transcription factor associated with neurodevelopmental disorders. Human cortical chimeroid experiments validated that both FEZF2 and TSHZ3 are required to drive module 20 activity and deep layer neuron specification but through distinct modalities. These studies demonstrate how meta-atlases can engender further mechanistic analyses of cortical fate specification.
Collapse
Grants
- UM1 MH130991 NIMH NIH HHS
- T32 NS048004 NINDS NIH HHS
- R01MH132689 U.S. Department of Health & Human Services | NIH | National Institute of Mental Health (NIMH)
- RF1 MH132662 NIMH NIH HHS
- T32 GM008243 NIGMS NIH HHS
- R00 NS111731 NINDS NIH HHS
- R00NS111731 U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- R01 MH132689 NIMH NIH HHS
- T32 GM145388 NIGMS NIH HHS
- U24 HG002371 NHGRI NIH HHS
- U.S. Department of Health & Human Services | NIH | National Institute of Mental Health (NIMH)
- U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- We would like to thank the members of the Bhaduri Lab for their insightful advice and comments on the study. We would like to thank the Broad Stem Cell Research Center Flow Cytometry core for their help in isolating cells for this project, Charina Julian for help with running sequencing, and Dr. Laurent Fasano for generously sharing the antibody against TSHZ3. The work performed in the manuscript was generously funded by R00NS111731 from the NIH (NINDS), R01MH132689 from the NIH (NIMH), the Young Investigator Award from the Brain & Behavior Research Foundation, the Alfred P. Sloan Foundation, the Rose Hills Foundation, and the Klingenstein-Simons Fellowship from the Esther A. & Joseph Klingenstein Fund and the Simons Foundation (to A.B.). Additional funding was provided to P.R.N. (UCLA Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research Training Program, UCLA Intercampus Medical Genetics Training Program (USHHS Ruth L. Kirschstein Institutional National Research Service Award # T32GM008243)), C.V.N. (T32 NS048004, Predoctoral Fellowship in association with the Training Grant in Neurobehavioral Genetics), and R.K. (T32 GM145388, Cell and Molecular Biology Training Program), and M.H. (NIMH BRAIN NIMH RF1MH132662, NHGRI U24HG002371, CIRM DISC0-14514 (with A.B.)).
Collapse
Affiliation(s)
- Patricia R Nano
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Elisa Fazzari
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Daria Azizad
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Antoni Martija
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Claudia V Nguyen
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sean Wang
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Vanna Giang
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ryan L Kan
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Juyoun Yoo
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Brittney Wick
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | | | - Aparna Bhaduri
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
12
|
Li C, Li W, Wei W, Chen Q, Gao H, Zhao Y, Zhang L, Ling L, Shen H, Shen Y, Shen Y. Gene expression profiles of endothelium, microglia and oligodendrocytes in hippocampus of post-stroke depression rat at single cell resolution. Mol Psychiatry 2025; 30:1995-2008. [PMID: 39521840 PMCID: PMC12015115 DOI: 10.1038/s41380-024-02810-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/20/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Post-stroke depression (PSD) is a common but severe mental complication after stroke. However, the cellular and molecular understanding of PSD is still yet to be illustrated. In current study, we prepared PSD rat model (MD) via unilateral middle cerebral artery occlusion (MCAO) and chronic stress stimulation (DEPR), and isolated hippocampal tissues for single cell sequencing of 10x Genomics Chromium. First, we determined the presence of the increased cell population of endothelium and microglia and the compromised oligodendrocytes in MD compared to NC, MCAO and DEPR. The enriched functions of highly variable genes (HVGs) of endothelium and microglia suggested a reinforced blood-brain barrier in MD. Next, cell clusters of endothelium, microglia and oligodendrocytes were individually analyzed, and the subtypes with distinct functions were identified. The presence of expression profiles, intercellular communications and signaling pathways of these three cell populations of PSD displayed a similar but more aggressive appearance with DEPR compared to MCAO and NC. Taken together, this study characterized the specific gene profile of endothelium, microglia and oligodendrocytes of hippocampal PSD by single cell sequencing, emphasizing the crosstalk among them to provide theoretical basis for the in-depth mechanism research and drug therapy of PSD.
Collapse
Affiliation(s)
- Cai Li
- Department of Neurology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Wentao Li
- Department of Neurology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Wenbin Wei
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, 200013, China
| | - Qili Chen
- School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning, China
| | - Han Gao
- Department of Neurology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Yanqing Zhao
- Department of Neurology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Lingling Zhang
- Department of Neurology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Li Ling
- Department of Neurology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Hao Shen
- Clinical laboratory, Suzhou Ninth People's Hospital, Suzhou, 215200, Jiangsu, China.
| | - Yifen Shen
- Central laboratory, Suzhou Ninth People's Hospital, Suzhou, 215200, Jiangsu, China.
| | - Yihang Shen
- Central laboratory, Suzhou Ninth People's Hospital, Suzhou, 215200, Jiangsu, China.
| |
Collapse
|
13
|
Maden SK, Huuki-Myers LA, Kwon SH, Collado-Torres L, Maynard KR, Hicks SC. lute: estimating the cell composition of heterogeneous tissue with varying cell sizes using gene expression. BMC Genomics 2025; 26:433. [PMID: 40312738 PMCID: PMC12045009 DOI: 10.1186/s12864-025-11508-x] [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] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/19/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Relative cell type fraction estimates in bulk RNA-sequencing data are important to control for cell composition differences across heterogenous tissue samples. While there exist algorithms to estimate the cell type proportions in tissues, a major challenge is the algorithms can show reduced performance if using tissues that have varying cell sizes, such as in brain tissue. In this way, without adjusting for differences in cell sizes, computational algorithms estimate the relative fraction of RNA attributable to each cell type, rather than the relative fraction of cell types, leading to potentially biased estimates in cellular composition. Furthermore, these tools were built on different frameworks with non-uniform input data formats while addressing different types of systematic errors or unwanted bias. RESULTS We present lute, a software tool to accurately deconvolute cell types with varying sizes. Our package lute wraps existing deconvolution algorithms in a flexible and extensible framework to enable easy benchmarking and comparison of existing deconvolution algorithms. Using simulated and real datasets, we demonstrate how lute adjusts for differences in cell sizes to improve the accuracy of cell composition. CONCLUSIONS Our software ( https://bioconductor.org/packages/lute ) can be used to enhance and improve existing deconvolution algorithms and can be used broadly for any type of tissue containing cell types with varying cell sizes.
Collapse
Affiliation(s)
- Sean K Maden
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Louise A Huuki-Myers
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Clinical Neurosciences, School of Clinical Medicine, The University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at The University of Cambridge, Cambridge, UK
| | - Sang Ho Kwon
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Leonardo Collado-Torres
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Stephanie C Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
14
|
Goes FS, Collado-Torres L, Zandi PP, Huuki-Myers L, Tao R, Jaffe AE, Pertea G, Shin JH, Weinberger DR, Kleinman JE, Hyde TM. Large-scale transcriptomic analyses of major depressive disorder reveal convergent dysregulation of synaptic pathways in excitatory neurons. Nat Commun 2025; 16:3981. [PMID: 40295477 PMCID: PMC12037741 DOI: 10.1038/s41467-025-59115-4] [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: 01/28/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Major Depressive Disorder (MDD) is a common, complex disorder that is a leading cause of disability worldwide and a significant risk factor for suicide. In this study, we have performed the largest molecular analysis of MDD in postmortem human brains (846 samples across 458 individuals) in the subgenual Anterior Cingulate Cortex (sACC) and the Amygdala, two regions central to mood regulation and the pathophysiology of MDD. We found extensive expression differences, particularly at the level of specific transcripts, with prominent enrichment for genes associated with the vesicular functioning, the postsynaptic density, GTPase signaling, and gene splicing. We find associated transcriptional features in 107 of 243 genome-wide significant loci for MDD and, through integrative analyses, highlight convergence of genetic risk, gene expression, and network-based analyses on dysregulated glutamatergic signaling and synaptic vesicular functioning. Together, these results provide an initial mechanistic understanding of MDD and highlight potential targets for novel drug discovery.
Collapse
Affiliation(s)
- Fernando S Goes
- Department of Psychiatry and Behavioral Sciences, Stanley and Elizabeth Star Precision Medicine Center of Excellence in Mood Disorders, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Leonardo Collado-Torres
- The Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Peter P Zandi
- Department of Psychiatry and Behavioral Sciences, Stanley and Elizabeth Star Precision Medicine Center of Excellence in Mood Disorders, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Ran Tao
- The Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Andrew E Jaffe
- The Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Geo Pertea
- The Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Joo Heon Shin
- The Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Daniel R Weinberger
- The Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joel E Kleinman
- Department of Psychiatry and Behavioral Sciences, Stanley and Elizabeth Star Precision Medicine Center of Excellence in Mood Disorders, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Thomas M Hyde
- The Lieber Institute for Brain Development, Baltimore, MD, USA.
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
15
|
Worf K, Matosin N, Gerstner N, Fröhlich AS, Koller AC, Degenhardt F, Thiele H, Rietschel M, Udawela M, Scarr E, Dean B, Theis FJ, Mueller NS, Knauer-Arloth J. Exon-variant interplay and multi-modal evidence identify endocrine dysregulation in severe psychiatric disorders impacting excitatory neurons. Transl Psychiatry 2025; 15:153. [PMID: 40253403 PMCID: PMC12009313 DOI: 10.1038/s41398-025-03366-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 03/17/2025] [Accepted: 03/31/2025] [Indexed: 04/21/2025] Open
Abstract
Bipolar disorder (BD), major depressive disorder (MDD), and schizophrenia share genetic architecture, yet their molecular mechanisms remain elusive. Both common and rare genetic variants contribute to neural dysfunction, impacting cognition and behavior. This study investigates the molecular effects of genetic variants on human cortical single-cell types using a single-exon analysis approach. Integrating exon-level eQTLs (common variants influencing exon expression) and joint exon eQT-Scores (combining polygenic risk scores with exon-level gene expression) from a postmortem psychiatric cohort (BD = 15, MDD = 24, schizophrenia = 68, controls = 62) with schizophrenia-focused rare variant data from the SCHEMA consortium, we identified 110 core genes enriched in pathways including circadian entrainment (FDR = 0.02), cortisol synthesis and secretion (FDR = 0.026), and dopaminergic synapse (FDR = 0.038). Additional enriched pathways included hormone signaling (FDRs < 0.0298, including insulin, GnRH, aldosterone, and growth hormone pathways) and, notably, adrenergic signaling in cardiomyocytes (FDR = 0.0028). These pathways highlight shared molecular mechanisms in the three disorders. Single-nuclei RNA sequencing data from three cortical regions revealed that these core set genes are predominantly expressed in excitatory neuron layers 2-6 of the dorsolateral prefrontal cortex, linking molecular changes to cell types involved in cognitive dysfunction. Our results demonstrate the power of integrating multimodal genetic and transcriptomic data at the exon level. This approach moves beyond symptom-based diagnoses toward molecular classifications, identifying potential therapeutic targets for psychiatric disorders.
Collapse
Affiliation(s)
- Karolina Worf
- Institute of Computational Biology, Helmholtz Center, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Natalie Matosin
- Department of Gene and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia
| | - Nathalie Gerstner
- Institute of Computational Biology, Helmholtz Center, Munich, Germany
- Department of Gene and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Anna S Fröhlich
- Department of Gene and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Anna C Koller
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Franziska Degenhardt
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Holger Thiele
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, University Medical Center Mannheim/University of Heidelberg, Mannheim, Germany
| | - Madhara Udawela
- The Molecular Psychiatry Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Elizabeth Scarr
- The Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia
| | - Brian Dean
- The Molecular Psychiatry Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- The Department of Florey, The University of Melbourne, Parkville, VIC, Australia
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Center, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Nikola S Mueller
- Institute of Computational Biology, Helmholtz Center, Munich, Germany
| | - Janine Knauer-Arloth
- Institute of Computational Biology, Helmholtz Center, Munich, Germany.
- Department of Gene and Environment, Max Planck Institute of Psychiatry, Munich, Germany.
| |
Collapse
|
16
|
Zhu W, Shi R, Li Y, Zhang G, Feng X, Cong J, He M, An Y, Ma R, Shi W, Cong B. Stress-Induced Cholesterol Metabolic Dysregulation and Differentiation Trajectory Shift in Oligodendrocytes Synergistically Drive Demyelination. Int J Mol Sci 2025; 26:3517. [PMID: 40332029 PMCID: PMC12026842 DOI: 10.3390/ijms26083517] [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: 03/14/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Stress-induced demyelination resulting from oligodendrocyte (OLG) dysfunction is one of the key pathological mechanisms of depression, yet its dynamic regulatory network remains unclear. This study integrates single-cell transcriptomics, lineage tracing, and functional interventions to uncover a temporally disordered OLG cholesterol metabolism in a restraint stress mouse model: After 3 days of stress, upregulation of efflux genes Abca1/Abcg1 triggers a compensatory response; however, by day 14, persistent suppression of transport genes (Apoe, Apod) and homeostatic regulatory genes (Dhcr24, Srebf2, etc.) leads to intracellular accumulation of "ineffective cholesterol", with compensatory activation of the AMPK pathway unable to restore cholesterol conversion into myelin. Pseudotime analysis further reveals that stress alters OLG differentiation trajectories, decreasing the proportion of mature OLGs and causing immature precursors to abnormally stall at the late pre-differentiation stage, resulting in myelin regeneration failure. Moreover, an immune OLG_C10 subpopulation expressing complement component C3 and P2ry12 is identified, indicating that OLGs may contribute to neuroinflammatory cascades through immune reprogramming. In summary, these findings reveal a novel mechanism from the dynamic perspective of OLGs, in which the interplay of "metabolic imbalance, differentiation blockade, and immune activation" collaboratively drives stress-induced demyelination, providing a theoretical foundation for depression treatment targeting OLG functional restoration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Weibo Shi
- Department of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China; (W.Z.); (R.S.); (Y.L.); (G.Z.); (X.F.); (J.C.); (M.H.); (Y.A.); (R.M.)
| | - Bin Cong
- Department of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China; (W.Z.); (R.S.); (Y.L.); (G.Z.); (X.F.); (J.C.); (M.H.); (Y.A.); (R.M.)
| |
Collapse
|
17
|
Fang C, Zhang X, Yang L, Sun L, Lu Y, Liu Y, Guo J, Wang M, Tan Y, Zhang J, Gao X, Zhu L, Liu G, Ren M, Xiao J, Zhang F, Ma S, Zhao R, Mei X, Qi D. Transcriptomic and morphologic vascular aberrations underlying FCDIIb etiology. Nat Commun 2025; 16:3320. [PMID: 40199880 PMCID: PMC11978774 DOI: 10.1038/s41467-025-58535-6] [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: 07/31/2023] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
Focal cortical dysplasia type II (FCDII) is a major cause of drug-resistant epilepsy, but genetic factors explain only some cases, suggesting other mechanisms. In this study, we conduct a molecular analysis of brain lesions and adjacent areas in FCDIIb patients. By analyzing over 217,506 single-nucleus transcriptional profiles from 15 individuals, we find significant changes in smooth muscle cells (SMCs) and astrocytes. We identify abnormal vascular malformations and a unique type of SMC that we call "Firework cells", which migrate from blood vessels into the brain parenchyma and associate with VIM+ cells. These abnormalities create localized ischemic-hypoxic (I/H) microenvironments, as confirmed by clinical data, further impairing astrocyte function, activating the HIF-1α/mTOR/S6 pathway, and causing neuronal loss. Using zebrafish models, we demonstrate that vascular abnormalities resulting in I/H environments promote seizures. Our results highlight vascular malformations as a factor in FCDIIb pathogenesis, suggesting potential therapeutic avenues.
Collapse
Affiliation(s)
- Chuantao Fang
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
- Shanghai Tenth People's Hospital, Institute for Infectious Diseases and Vaccine Development, Tongji University School of Medicine, Shanghai, China
| | - Xiaodan Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Lin Yang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Licheng Sun
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yujia Lu
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Liu
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA) - CITEXVI, Vigo, Spain
| | - Jingjing Guo
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Min Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Yanfeng Tan
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Jinsen Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Xin Gao
- Shanghai Universal Medical Imaging Diagnostic Center, Shanghai, China
| | - Li Zhu
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guoping Liu
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Maozhi Ren
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Jianbo Xiao
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA) - CITEXVI, Vigo, Spain
| | - Fayong Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Shaojie Ma
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui Zhao
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China.
- Department of Neurosurgery, Children's Hospital of Shanghai, Shanghai, China.
- Department of Neurosurgery, Hainan Women and Children's Medical Center, Haikou, China.
| | - Xinyu Mei
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Dashi Qi
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China.
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China.
| |
Collapse
|
18
|
Kim S, Yang S, Jung J, Choi J, Kang M, Joo J. Psychedelic Drugs in Mental Disorders: Current Clinical Scope and Deep Learning-Based Advanced Perspectives. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413786. [PMID: 40112231 PMCID: PMC12005819 DOI: 10.1002/advs.202413786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 02/13/2025] [Indexed: 03/22/2025]
Abstract
Mental disorders are a representative type of brain disorder, including anxiety, major depressive depression (MDD), and autism spectrum disorder (ASD), that are caused by multiple etiologies, including genetic heterogeneity, epigenetic dysregulation, and aberrant morphological and biochemical conditions. Psychedelic drugs such as psilocybin and lysergic acid diethylamide (LSD) have been renewed as fascinating treatment options and have gradually demonstrated potential therapeutic effects in mental disorders. However, the multifaceted conditions of psychiatric disorders resulting from individuality, complex genetic interplay, and intricate neural circuits impact the systemic pharmacology of psychedelics, which disturbs the integration of mechanisms that may result in dissimilar medicinal efficiency. The precise prescription of psychedelic drugs remains unclear, and advanced approaches are needed to optimize drug development. Here, recent studies demonstrating the diverse pharmacological effects of psychedelics in mental disorders are reviewed, and emerging perspectives on structural function, the microbiota-gut-brain axis, and the transcriptome are discussed. Moreover, the applicability of deep learning is highlighted for the development of drugs on the basis of big data. These approaches may provide insight into pharmacological mechanisms and interindividual factors to enhance drug discovery and development for advanced precision medicine.
Collapse
Affiliation(s)
- Sung‐Hyun Kim
- Department of PharmacyCollege of PharmacyHanyang UniversityAnsanGyeonggi‐do15588Republic of Korea
| | - Sumin Yang
- Department of PharmacyCollege of PharmacyHanyang UniversityAnsanGyeonggi‐do15588Republic of Korea
| | - Jeehye Jung
- Department of PharmacyCollege of PharmacyHanyang UniversityAnsanGyeonggi‐do15588Republic of Korea
| | - Jeonghyeon Choi
- Department of PharmacyCollege of PharmacyHanyang UniversityAnsanGyeonggi‐do15588Republic of Korea
| | - Mingon Kang
- Department of Computer ScienceUniversity of NevadaLas VegasNV89154USA
| | - Jae‐Yeol Joo
- Department of PharmacyCollege of PharmacyHanyang UniversityAnsanGyeonggi‐do15588Republic of Korea
| |
Collapse
|
19
|
Chen Y, Zhen C, Mo Y, Liu J, Zhang L. Multiscale Dissection of Spatial Heterogeneity by Integrating Multi-Slice Spatial and Single-Cell Transcriptomics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413124. [PMID: 39999288 PMCID: PMC12005799 DOI: 10.1002/advs.202413124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/05/2025] [Indexed: 02/27/2025]
Abstract
The spatial structure of cells is highly organized at multiscale levels from global spatial domains to local cell type heterogeneity. Existing methods for analyzing spatially resolved transcriptomics (SRT) are separately designed for either domain alignment across multiple slices or deconvoluting cell type compositions within a single slice. To this end, a novel deep learning method, SMILE, is proposed which combines graph contrastive autoencoder and multilayer perceptron with local constraints to learn multiscale and informative spot representations. By comparing SMILE with the state-of-the-art methods on simulation and real datasets, the superior performance of SMILE is demonstrated on spatial alignment, domain identification, and cell type deconvolution. The results show SMILE's capability not only in simultaneously dissecting spatial variations at different scales but also in unraveling altered cellular microenvironments in diseased conditions. Moreover, SMILE can utilize prior domain annotation information of one slice to further enhance the performance.
Collapse
Affiliation(s)
- Yuqi Chen
- School of Computer ScienceWuhan UniversityWuchang DistrictWuhanHubei430072China
| | - Caiwei Zhen
- School of Computer ScienceWuhan UniversityWuchang DistrictWuhanHubei430072China
| | - Yuanyuan Mo
- School of Computer ScienceWuhan UniversityWuchang DistrictWuhanHubei430072China
| | - Juan Liu
- School of Computer ScienceWuhan UniversityWuchang DistrictWuhanHubei430072China
| | - Lihua Zhang
- School of Computer ScienceWuhan UniversityWuchang DistrictWuhanHubei430072China
| |
Collapse
|
20
|
Chen J, Zhu X, Yang F, Liu Y, Ba H, Huang P, Wang H, Bian Y, Li C, Zhang S. Exploring Male-Specific Synaptic Plasticity in Major Depressive Disorder: A Single-Nucleus Transcriptomic Analysis Using Bioinformatics Methods. Int J Mol Sci 2025; 26:3135. [PMID: 40243907 PMCID: PMC11989135 DOI: 10.3390/ijms26073135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Major depressive disorder (MDD) is a complex psychiatric illness, with synaptic plasticity playing a key role in its pathology. Our study aims to investigate the molecular basis of MDD by analyzing synaptic plasticity-related gene expression at the single-cell level. Utilizing a published snRNA-seq dataset (GSE144136), we identified Excitatory.neurons_1 as the cell cluster most associated with MDD and synaptic plasticity through cell clustering, gene set enrichment analysis (GSEA), and pseudotime analysis. Integrating the bulk RNA-seq data (GSE38206), we identified CASKIN1 and CSTB as hub genes via differential expression analysis and machine learning methods. Further exploration of the relevant mechanisms was performed via cell-cell communication and ligand-receptor interaction analysis, functional enrichment analysis, and the construction of molecular regulatory networks, highlighting miR-21-5p as a key biomarker. We propose that elevated miR-21-5p in MDD downregulates CASKIN1 in Excitatory.neurons_1 cells, resulting in decreased neural connectivity and altered synaptic plasticity. As our analyzed snRNA-seq dataset consists solely of male samples, these findings may be male-specific. Our findings shed light on potential mechanisms underlying synaptic plasticity in MDD, offering novel insights into the disorder's cellular and molecular dynamics.
Collapse
Affiliation(s)
- Ji Chen
- Institute of Forensic Science, Fudan University, Shanghai 200032, China; (J.C.); (X.Z.); (P.H.)
| | - Xiumei Zhu
- Institute of Forensic Science, Fudan University, Shanghai 200032, China; (J.C.); (X.Z.); (P.H.)
- School of Forensic Medicine, China Medical University, Shenyang 110122, China
| | - Fan Yang
- Key Laboratory of Forensic Evidence and Science Technology, Institute of Forensic Science, Ministry of Public Security, Shanghai 200042, China; (F.Y.); (Y.L.)
| | - Yanan Liu
- Key Laboratory of Forensic Evidence and Science Technology, Institute of Forensic Science, Ministry of Public Security, Shanghai 200042, China; (F.Y.); (Y.L.)
| | - Huajie Ba
- DNA Laboratory, Public Security Bureau of Changzhou, Changzhou 213022, China;
| | - Ping Huang
- Institute of Forensic Science, Fudan University, Shanghai 200032, China; (J.C.); (X.Z.); (P.H.)
| | - Hongyan Wang
- Institute of Metabolism and Integrative Biology, Institute of Reproduction and Development, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China;
| | - Yingnan Bian
- Enlight Medical Technologies, Pudong New Area, Shanghai 201318, China;
| | - Chengtao Li
- Institute of Forensic Science, Fudan University, Shanghai 200032, China; (J.C.); (X.Z.); (P.H.)
| | - Suhua Zhang
- Institute of Forensic Science, Fudan University, Shanghai 200032, China; (J.C.); (X.Z.); (P.H.)
| |
Collapse
|
21
|
Han Y, Wang X, Cheng S, Yan P, Chen Y, Kang N, Zhou Z, Guo X, Lu Y, Wang Q, Li X, Su X, Shi H, Liu Q, Li W, Yang Y, Lv L. Cortical morphometric similarity gradient in schizophrenia and its association with transcriptional profiles and clinical phenotype. Psychol Med 2025; 55:e97. [PMID: 40143806 DOI: 10.1017/s0033291725000479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
BACKGROUND Recent studies have increasingly utilized gradient metrics to investigate the spatial transitions of brain organization, enabling the conversion of macroscale brain features into low-dimensional manifold representations. However, it remains unclear whether alterations exist in the cortical morphometric similarity (MS) network gradient in patients with schizophrenia (SCZ). This study aims to examine potential differences in the principal MS gradient between individuals with SCZ and healthy controls and to explore how these differences relate to transcriptional profiles and clinical phenomenology. METHODS MS network was constructed in this study, and its gradient of the network was computed in 203 patients with SCZ and 201 healthy controls, who shared the same demographics in terms of age and gender. To examine irregularities in the MS network gradient, between-group comparisons were carried out, and partial least squares regression analysis was used to study the relationships between the MS network gradient-based variations in SCZ, and gene expression patterns and clinical phenotype. RESULTS In contrast to healthy controls, the principal MS gradient of patients with SCZ was primarily significantly lower in sensorimotor areas, and higher in more areas. In addition, the aberrant gradient pattern was spatially linked with the genes enriched for neurobiologically significant pathways and preferential expression in various brain regions and cortical layers. Furthermore, there were strong positive connections between the principal MS gradient and the symptomatologic score in SCZ. CONCLUSIONS These findings showed changes in the principal MS network gradient in SCZ and offered potential molecular explanations for the structural changes underpinning SCZ.
Collapse
Affiliation(s)
- Yong Han
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, China
| | - Xiujuan Wang
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Shumin Cheng
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Pengyue Yan
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Yi Chen
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Ning Kang
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Zhilu Zhou
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Xiaoge Guo
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Yanli Lu
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Qi Wang
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Xue Li
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, China
| | - Xi Su
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Han Shi
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Qing Liu
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Wenqiang Li
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| | - Yongfeng Yang
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, China
| | - Luxian Lv
- Department of Psychiatry, Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
22
|
Liu H, Chen S, Xiang H, Xiao J, Zhao S, Zhang X, Shu Z, Zhang J, Ouyang J, Liu Q, Quan Q, Fan J, Gao P, Zheng X, Chen AF, Lu H. S1PR3 in hippocampal neurons improves synaptic plasticity and decreases depressive behavior via downregulation of RhoA/ROCK1. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111256. [PMID: 39828081 DOI: 10.1016/j.pnpbp.2025.111256] [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: 05/12/2024] [Revised: 10/30/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
The study investigates how Sphingosine-1-phosphate receptor 3 (S1PR3) and the Chronic Unpredictable Mild Stress (CUMS) affects depression-like behaviors. The S1P/S1PR3 signaling pathway is known to play a role in mood regulation, but it is not yet fully understood how it is connected to depression. This study looks to further explore this topic. To investigate the effect of CUMS on S1PR3 expression in hippocampus neurons and the synaptic plasticity, we observed animals' behavior with Sucrose Preference Test (SPT), Forced Swim Test (FST) and Open Field Test (OFT). Combining molecular and histological analysis, we investigated the S1PR3 expression, the change in synapse density, and synaptic structure change in the hippocampus. The CUMS caused a significant decrease in the S1PR3 expression, the density of the synaptic spine and synaptic ultrastructure change in mice. On the other hand, over-expression of S1PR3 by adeno-associated virus (AAV) in hippocampal neurons alleviated the depressive-like behaviors and synaptic deficits observed in stress-susceptible animals. Furthermore, the depressive-like phenotype and synaptic impairments were normalized by the expression of RhoA, implicating the RhoA/ROCK1 pathway in S1PR3 actions. Collectively, our findings provide strong evidence that S1PR3 plays a key role in hippocampal synaptic plasticity and depression and that modulation of S1PR3/RhoA/ROCK1 signaling may offer a novel therapeutic strategy for MDD. This study not only underscores the therapeutic potential of S1PR3 but also provides novel insights into the molecular mechanisms underlying depression.
Collapse
Affiliation(s)
- Huiqin Liu
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shuhua Chen
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, China
| | - Hong Xiang
- Center for Experimental Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jie Xiao
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shaoli Zhao
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiao Zhang
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, China
| | - Zhihao Shu
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jing Zhang
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jie Ouyang
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Quanjun Liu
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Qisheng Quan
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jianing Fan
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Peng Gao
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xinru Zheng
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Alex F Chen
- Center for Experimental Medicine, The Third Xiangya Hospital of Central South University, Changsha, China; Institute for Cardiovascular Development and Regenerative Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hongwei Lu
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
23
|
Lv H, Liu P, Wang Y, Huang J, Xie Y, Guan M, Cong J, Jiang Y, Xu Y. Identification of DIO2 as a Molecular Therapeutic Target for Depression in Chronic Rhinosinusitis: A Comprehensive Bioinformatics and Experimental Study. Biochem Genet 2025:10.1007/s10528-025-11085-4. [PMID: 40089956 DOI: 10.1007/s10528-025-11085-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/10/2025] [Indexed: 03/18/2025]
Abstract
Chronic rhinosinusitis (CRS) and depression are both common conditions with significant socioeconomic impact. The high co-occurrence of depression in CRS patients suggests a common pathophysiology, but the mechanisms are unclear. This study aimed to identify potential molecular links between the two conditions. We retrieved gene expression datasets for CRS and depression from the GEO database. Using differentially expressed genes (DEGs) analysis and weighted gene co-expression network analysis (WGCNA), we identified co-expression genes associated with CRS and depression. Enrichment analyses including GO, KEGG, and GSEA were performed to explore biological pathways. Machine learning algorithms including random forest and LASSO regression were engaged to screen for shared hub genes predictive of CRS and depression. Single-cell RNA sequencing (scRNA-seq) data were analyzed to delineate the expression profiles of the shared hub genes across different cell types. Animal experiments were employed to validate the role of core genes in CRS-related depression. We identified five shared hub genes: CHRDL1, DIO2, HSD17B6, PDE3A, and PLA2G5, with the TGF-β signaling, cytokine-cytokine interaction receptors, and cell adhesion as key biological pathways. DIO2, as identified by machine learning, is a promising diagnostic biomarker for CRS and depression. The scRNA-seq analysis showed that DIO2 is primarily expressed in neurons and astrocytes. Animal experiments showed that overexpression of DIO2 improved the depressive-like behaviors in CRS mice. This study sheds new light on the molecular basis of the comorbidity between CRS and depression. DIO2 is a potential diagnostic and therapeutic target for CRS patients with comorbid depression.
Collapse
Affiliation(s)
- Hao Lv
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China
| | - Peiqiang Liu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China
| | - Yunfei Wang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China
| | - Jingyu Huang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China
| | - Yulie Xie
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China
| | - Mengting Guan
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China
| | - Jianchao Cong
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China
| | - Yang Jiang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China.
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China.
| | - Yu Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China.
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China.
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Rd, Wuhan, Hubei, China.
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, Hubei, China.
| |
Collapse
|
24
|
Chi D, Zhang K, Zhang J, He Z, Zhou H, Huang W, Liu Y, Huang J, Zeng W, Bai X, Ou C, Ouyang H. Astrocytic pleiotrophin deficiency in the prefrontal cortex contributes to stress-induced depressive-like responses in male mice. Nat Commun 2025; 16:2528. [PMID: 40087317 PMCID: PMC11909280 DOI: 10.1038/s41467-025-57924-1] [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: 11/01/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Astrocytes are closely linked to depression, and the prefrontal cortex (PFC) is an important brain region involved in major depressive disorder (MDD). However, the underlying mechanism by which astrocytes within PFC contribute to MDD remains unclear. Using single-nucleus RNA sequencing analyses, we show a significant reduction in astrocytes and attenuated pleiotrophin-protein tyrosine phosphatase receptor type Z1 (PTN-PTPRZ1) signaling in astrocyte-to-excitatory neuron communication in the PFC of male MDD patients. We find reduced astrocytes and PTN in the dorsomedial PFC of male mice with depression induced by chronic restraint and social defeat stress. Knockdown of astrocytic PTN induces depression-related responses, which is reversed by exogenous PTN supplementation or overexpression of astrocytic PTN. The antidepressant effects exerted by astrocytic PTN require interaction with PTPRZ1 in excitatory neurons, and PTN-PTPRZ1 activates the AKT signaling pathway to regulate depression-related responses. Our findings indicate the PTN-PTPRZ1-AKT pathway may be a potential therapeutic target for MDD.
Collapse
Affiliation(s)
- Dongmei Chi
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Kun Zhang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jianxing Zhang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Zhaoli He
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Hongxia Zhou
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Wan Huang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yang Liu
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jingxiu Huang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Weian Zeng
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Xiaohui Bai
- Department of Anesthesiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation; Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Chaopeng Ou
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.
| | - Handong Ouyang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.
| |
Collapse
|
25
|
Zheng J, Wang H, Wu W, Wang L, Qin M, Zhu L, Liu Z, Chen Y, Yu Y. Role of FPR2 antagonism in alleviating social isolation-induced depression and protecting blood-brain barrier integrity. J Neuroinflammation 2025; 22:79. [PMID: 40083006 PMCID: PMC11907847 DOI: 10.1186/s12974-025-03408-4] [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: 10/21/2024] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
Social isolation (SI) is a prevalent issue in modern society, particularly exacerbated during the COVID-19 pandemic, and it is a significant contributor to depressive disorders. Inflammation-related markers are upregulated in patients with major depressive disorder (MDD) unresponsive to first-line selective serotonin reuptake inhibitor (SSRI) antidepressants. This study investigates the role of formyl peptide receptor 2 (FPR2), a G-protein coupled receptor expressed in central and peripheral immune cells, in SI-induced depression. We developed a mouse model of SI by housing mice individually for three weeks. SI mice exhibited increased capillary-associated microglia (CAMs) with upregulated FPR2 expression in the prefrontal cortex (PFC) and hippocampus compared to group-housed controls. Notably, subcutaneous administration of the FPR2 antagonist WRW4 alleviated depressive and anxiety-like behaviors in SI mice, reducing microglial activation and neuronal damage. WRW4 treatment decreased CAM numbers and their FPR2 expression. RNA sequencing revealed that SI primarily induced changes in genes associated with blood-brain barrier (BBB) function, followed by alterations in genes related to hormone activity, immune activation, and neuronal function. Transcriptomic changes in brain endothelial cells from SI mice resembled those observed in animal models of several neurological disorders and in MDD patients. WRW4 treatment partially reversed these transcriptomic alterations and restored compromised BBB integrity. Additionally, intracerebroventricular (ICV) injection of WRW4 also alleviated depressive and anxiety-like behaviors in SI mice. Finally, our analysis of public transcriptome databases indicates FPR2 upregulation in the orbital ventral PFC of MDD patients and peripheral blood mononuclear cells of those in severe depressive episodes. These findings suggest that the pharmacological targeting of FPR2 may rescue SI-induced pathology in mice by protecting BBB integrity.
Collapse
Affiliation(s)
- Jiayi Zheng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hanqi Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wanning Wu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Linlin Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Meizhen Qin
- School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, China
| | - Lingfeng Zhu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhen Liu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yijun Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yang Yu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
26
|
Gerstner N, Fröhlich AS, Matosin N, Gagliardi M, Cruceanu C, Ködel M, Rex-Haffner M, Tu X, Mostafavi S, Ziller MJ, Binder EB, Knauer-Arloth J. Contrasting genetic predisposition and diagnosis in psychiatric disorders: A multi-omic single-nucleus analysis of the human OFC. SCIENCE ADVANCES 2025; 11:eadq2290. [PMID: 40053590 PMCID: PMC11887846 DOI: 10.1126/sciadv.adq2290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025]
Abstract
Psychiatric disorders like schizophrenia, bipolar disorder, and major depressive disorder exhibit substantial genetic and clinical overlap. However, their molecular architecture remains elusive due to their polygenic nature and complex brain cell interactions. We integrated clinical data with genetic susceptibility to investigate gene expression and chromatin accessibility in the orbitofrontal cortex of 92 postmortem human brain samples at the single-nucleus (sn) level. Using snRNA-seq and snATAC-seq, we analyzed ~800,000 and 400,000 nuclei, respectively. We observed cell-type-specific dysregulation related to clinical diagnosis and genetic risk. Dysregulation in gene expression and chromatin accessibility associated with diagnosis was pronounced in excitatory neurons. Conversely, genetic risk predominantly affected glial and endothelial cells. Notably, INO80E and HCN2 genes exhibited dysregulation in excitatory neurons' superficial layers 2/3 influenced by schizophrenia polygenic risk. This study unveils the complex genetic and epigenetic landscape of psychiatric disorders, emphasizing the importance of cell-type-specific analyses in understanding their pathogenesis and contrasting genetic predisposition with clinical diagnosis.
Collapse
Affiliation(s)
- Nathalie Gerstner
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry, Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Anna S. Fröhlich
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Natalie Matosin
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Miriam Gagliardi
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Cristiana Cruceanu
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Maik Ködel
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Monika Rex-Haffner
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Xinming Tu
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - Sara Mostafavi
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | | | - Elisabeth B. Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Janine Knauer-Arloth
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
27
|
Lian K, Yang W, Ye J, Chen Y, Zhang L, Xu X. The role of senescence-related genes in major depressive disorder: insights from machine learning and single cell analysis. BMC Psychiatry 2025; 25:188. [PMID: 40033248 PMCID: PMC11874787 DOI: 10.1186/s12888-025-06542-8] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Evidence indicates that patients with Major Depressive Disorder (MDD) exhibit a senescence phenotype or an increased susceptibility to premature senescence. However, the relationship between senescence-related genes (SRGs) and MDD remains underexplored. METHODS We analyzed 144 MDD samples and 72 healthy controls from the GEO database to compare SRGs expression. Using Random Forest (RF) and Support Vector Machine-Recursive Feature Elimination (SVM-RFE), we identified five hub SRGs to construct a logistic regression model. Consensus cluster analysis, based on SRGs expression patterns, identified subclusters of MDD patients. Weighted Gene Co-expression Network Analysis (WGCNA) identified gene modules strongly linked to each cluster. Single-cell RNA sequencing was used to analyze MDD SRGs functions. RESULTS The five hub SRGs: ALOX15B, TNFSF13, MARCH 15, UBTD1, and MAPK14 showed differential expression between MDD patients and controls. Diagnostics models based on these hub genes demonstrated high accuracy. The hub SRGs correlated positively with neutrophils and negatively with T lymphocytes. SRGs expression pattern revealed two distinct MDD subclusters. WGCNA identified significant gene modules within these subclusters. Additionally, individual endothelial cells with high senescence scores were found to interact with astrocytes via the Notch signaling pathway, suggesting a specific role in MDD pathogenesis. CONCLUSION This comprehensive study elucidates the significant role of SRGs in MDD, highlighting the importance of the Notch signaling pathway in mediating senescence effects.
Collapse
Affiliation(s)
- Kun Lian
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, China
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, No.295, Xichang Road, Wuhua District, Kunming, Yunnan, 650000, China
| | - Wei Yang
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, No.295, Xichang Road, Wuhua District, Kunming, Yunnan, 650000, China
- Department of Psychiatry, The Second People's Hospital of Yuxi, No. 4, Xingyun Road, High-tech Development Zone, Yuxi, Yunnan, 653100, China
- Yuxi Hospital, Kunming University of Science and Technology, Yuxi, Yunnan, 653100, China
| | - Jing Ye
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, No.295, Xichang Road, Wuhua District, Kunming, Yunnan, 650000, China
| | - Yilan Chen
- Department of Psychiatry, The Second People's Hospital of Yuxi, No. 4, Xingyun Road, High-tech Development Zone, Yuxi, Yunnan, 653100, China
- Yuxi Hospital, Kunming University of Science and Technology, Yuxi, Yunnan, 653100, China
| | - Lei Zhang
- Department of Psychiatry, The Second People's Hospital of Yuxi, No. 4, Xingyun Road, High-tech Development Zone, Yuxi, Yunnan, 653100, China
- Yuxi Hospital, Kunming University of Science and Technology, Yuxi, Yunnan, 653100, China
| | - Xiufeng Xu
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, No.295, Xichang Road, Wuhua District, Kunming, Yunnan, 650000, China.
- Yunnan Clinical Research Center for Mental Disorders, Kunming, Yunnan, 650000, China.
| |
Collapse
|
28
|
Li XY, Rao Y, Li GH, He L, Wang Y, He W, Fang P, Pei C, Xi L, Xie H, Lu YR. Single-nucleus RNA sequencing uncovers metabolic dysregulation in the prefrontal cortex of major depressive disorder patients. Sci Rep 2025; 15:7418. [PMID: 40033004 PMCID: PMC11876315 DOI: 10.1038/s41598-025-92030-8] [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: 11/16/2024] [Accepted: 02/25/2025] [Indexed: 03/05/2025] Open
Abstract
Major depressive disorder (MDD) is a widespread psychiatric condition, recognized as the third leading cause of global disease burden in 2008. In the context of MDD, alterations in synaptic transmission within the prefrontal cortex (PFC) are associated with PFC hypoactivation, a key factor in cognitive function and mood regulation. Given the high energy demands of the central nervous system, these synaptic changes suggest a metabolic imbalance within the PFC of MDD patients. However, the cellular mechanisms underlying this metabolic dysregulation remain not fully elucidated. This study employs single-nucleus RNA sequencing (snRNA-seq) data to predict metabolic alterations in the dorsolateral PFC (DLPFC) of MDD patients. Our analysis revealed cell type-specific metabolic patterns, notably the disruption of oxidative phosphorylation and carbohydrate metabolism in the DLPFC of MDD patients. Gene set enrichment analysis based on human phenotype ontology predicted alterations in serum lactate levels in MDD patients, corroborated by the observed decrease in lactate levels in MDD patients compared to 47 age-matched healthy controls (HCs). This transcriptional analysis offers novel insights into the metabolic disturbances associated with MDD and the energy dynamics underlying DLPFC hypoactivation. These findings are instrumental for comprehending the pathophysiology of MDD and may guide the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Xiang-Yao Li
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain, Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Yingbo Rao
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Guo-Hao Li
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Luxi He
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yaohan Wang
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Wenli He
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Ping Fang
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Chenyu Pei
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain, Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Lun Xi
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Haiyan Xie
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yun-Rong Lu
- Department of Psychiatry, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
- Department of Psychiatry, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
29
|
Arbabi K, Newton DF, Oh H, Davie MC, Lewis DA, Wainberg M, Tripathy SJ, Sibille E. Transcriptomic pathology of neocortical microcircuit cell types across psychiatric disorders. Mol Psychiatry 2025; 30:1057-1068. [PMID: 39237723 DOI: 10.1038/s41380-024-02707-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 09/07/2024]
Abstract
Psychiatric disorders such as major depressive disorder (MDD), bipolar disorder (BD), and schizophrenia (SCZ) are characterized by altered cognition and mood, brain functions that depend on information processing by cortical microcircuits. We hypothesized that psychiatric disorders would display cell type-specific transcriptional alterations in neuronal subpopulations that make up cortical microcircuits: excitatory pyramidal (PYR) neurons and vasoactive intestinal peptide- (VIP), somatostatin- (SST), and parvalbumin- (PVALB) expressing inhibitory interneurons. Using laser capture microdissection followed by RNA sequencing (LCM-seq), we performed cell type-specific molecular profiling of subgenual anterior cingulate cortex, a region implicated in mood and cognitive control. We sequenced libraries from 130 whole cells pooled per neuronal subtype (VIP, SST, PVALB, superficial and deep PYR) in 76 subjects from the University of Pittsburgh Brain Tissue Donation Program, evenly split between MDD, BD and SCZ subjects and healthy controls (totaling 380 bulk transcriptomes from ~50,000 neurons). We identified hundreds of differentially expressed (DE) genes and biological pathways across disorders and neuronal subtypes, with the vast majority in interneurons, particularly PVALB. While DE genes were unique to each cell type, there was a partial overlap across disorders for genes involved in the formation and maintenance of neuronal circuits. We observed coordinated alterations in biological pathways between select pairs of microcircuit cell types, also partially shared across disorders. Finally, DE genes coincided with known risk variants from psychiatric genome-wide association studies, suggesting cell type-specific convergence between genetic and transcriptomic risk for psychiatric disorders. Our study suggests transdiagnostic cortical microcircuit pathology in SCZ, BD, and MDD and sets the stage for larger-scale studies investigating how cell circuit-based changes contribute to shared psychiatric risk.
Collapse
Affiliation(s)
- Keon Arbabi
- The Krembil Centre for Neuroinformatics, Centre for Addiction & Mental Health, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Dwight F Newton
- Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
| | - Hyunjung Oh
- Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
| | - Melanie C Davie
- The Krembil Centre for Neuroinformatics, Centre for Addiction & Mental Health, Toronto, ON, Canada
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael Wainberg
- The Krembil Centre for Neuroinformatics, Centre for Addiction & Mental Health, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Shreejoy J Tripathy
- The Krembil Centre for Neuroinformatics, Centre for Addiction & Mental Health, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Etienne Sibille
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
30
|
Warden AS, Salem NA, Brenner E, Sutherland GT, Stevens J, Kapoor M, Goate AM, Mayfield RD. Integrative Genomics Approach Identifies Glial Transcriptomic Dysregulation and Risk in the Cortex of Individuals With Alcohol Use Disorder. Biol Psychiatry 2025:S0006-3223(25)00994-1. [PMID: 40024496 DOI: 10.1016/j.biopsych.2025.02.895] [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: 09/09/2024] [Revised: 01/24/2025] [Accepted: 02/14/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Alcohol use disorder (AUD) is a prevalent neuropsychiatric disorder that is a major global health concern, affecting millions of people worldwide. Previous studies of AUD used underpowered single-cell analysis or bulk homogenates of postmortem brain tissue, which obscure gene expression changes in specific cell types. Therefore, we sought to conduct the largest-to-date single-nucleus RNA sequencing (snRNA-seq) postmortem brain study in AUD to elucidate transcriptomic pathology with cell type-specific resolution. METHODS Here, we performed snRNA-seq and high-dimensional network analysis of 73 postmortem samples from individuals with AUD (n = 36, nnuclei = 248,873) and neurotypical control individuals (n = 37, nnuclei = 210,573) in the dorsolateral prefrontal cortex from both male and female donors. Additionally, we performed analysis for cell type-specific enrichment of aggregate genetic risk for AUD as well as integration of the AUD proteome for secondary validation. RESULTS We identified 32 distinct cell clusters and found widespread cell type-specific transcriptomic changes across the cortex in AUD, particularly affecting glial populations. We found the greatest dysregulation in novel microglial and astrocytic subtypes that accounted for the majority of differential gene expression and coexpression modules linked to AUD. Differential gene expression was secondarily validated by integration of a publicly available AUD proteome. Finally, analysis for aggregate genetic risk for AUD identified subtypes of glia as potential key players not only affected by but also causally linked to the progression of AUD. CONCLUSIONS These results highlight the importance of cell type-specific molecular changes in AUD and offer opportunities to identify novel targets for treatment on the single-nucleus level.
Collapse
Affiliation(s)
- Anna S Warden
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas
| | - Nihal A Salem
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas; Institute for Neuroscience, University of Texas at Austin, Austin, Texas
| | - Eric Brenner
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas
| | - Greg T Sutherland
- School of Medical Sciences and Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Julia Stevens
- New South Wales Brain Tissue Resource Centre, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Manav Kapoor
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Alison M Goate
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, New York; Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas; Institute for Neuroscience, University of Texas at Austin, Austin, Texas.
| |
Collapse
|
31
|
Zhan Y, Zhang Y, Hu Z, Wang Y, Zhu Z, Du S, Yan X, Li X. LETSmix: a spatially informed and learning-based domain adaptation method for cell-type deconvolution in spatial transcriptomics. Genome Med 2025; 17:16. [PMID: 40022231 PMCID: PMC11869467 DOI: 10.1186/s13073-025-01442-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/17/2025] [Indexed: 03/03/2025] Open
Abstract
Spatial transcriptomics (ST) enables the study of gene expression in spatial context, but many ST technologies face challenges due to limited resolution, leading to cell mixtures at each spot. We present LETSmix to deconvolve cell types by integrating spatial correlations through a tailored LETS filter, which leverages layer annotations, expression similarities, image texture features, and spatial coordinates to refine ST data. Additionally, LETSmix employs a mixup-augmented domain adaptation strategy to address discrepancies between ST and reference single-cell RNA sequencing data. Comprehensive evaluations across diverse ST platforms and tissue types demonstrate its high accuracy in estimating cell-type proportions and spatial patterns, surpassing existing methods (URL: https://github.com/ZhanYangen/LETSmix ).
Collapse
Affiliation(s)
- Yangen Zhan
- Division of Information Science and Technology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518052, China
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Yongbing Zhang
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China.
| | - Zheqi Hu
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Yifeng Wang
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Zirui Zhu
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Sijing Du
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Xiangming Yan
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Xiu Li
- Division of Information Science and Technology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518052, China.
| |
Collapse
|
32
|
Nakatsuka N, Adler D, Jiang L, Hartman A, Cheng E, Klann E, Satija R. A Reproducibility Focused Meta-Analysis Method for Single-Cell Transcriptomic Case-Control Studies Uncovers Robust Differentially Expressed Genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.15.618577. [PMID: 39463993 PMCID: PMC11507907 DOI: 10.1101/2024.10.15.618577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
We assessed the reproducibility of differentially expressed genes (DEGs) in previously published Alzheimer's (AD), Parkinson's (PD), Schizophrenia (SCZ), and COVID-19 scRNA-seq studies. While transcriptional scores from DEGs of individual PD and COVID-19 datasets had moderate predictive power for case-control status of other datasets (AUC=0.77 and 0.75), genes from individual AD and SCZ datasets had poor predictive power (AUC=0.68 and 0.55). We developed a non-parametric meta-analysis method, SumRank, based on reproducibility of relative differential expression ranks across datasets, and found DEGs with improved predictive power (AUC=0.88, 0.91, 0.78, and 0.62). By multiple other metrics, specificity and sensitivity of these genes were substantially higher than those discovered by dataset merging and inverse variance weighted p-value aggregation methods. The DEGs revealed known and novel biological pathways, and we validate BCAT1 as down-regulated in AD mouse oligodendrocytes. Lastly, we evaluate factors influencing reproducibility of individual studies as a prospective guide for experimental design.
Collapse
|
33
|
Xue K, Liu F, Liang S, Guo L, Shan Y, Xu H, Xue J, Jiang Y, Zhang Y, Lu J. Brain connectivity and transcriptomic similarity inform abnormal morphometric similarity patterns in first-episode, treatment-naïve major depressive disorder. J Affect Disord 2025; 370:519-531. [PMID: 39522735 DOI: 10.1016/j.jad.2024.11.021] [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: 06/06/2024] [Revised: 10/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) is associated with disrupted brain structural integration. Morphometric similarity offers a means to capture the coordinated patterns of various structural features. However, it remains unknown whether MDD-related changes can be detected in cortical morphometric similarity through the Morphometric Inverse Divergence (MIND) network. Additionally, the role of brain connectivity in shaping these alterations, and their links to neuroreceptors and gene expression, have yet to be investigated. METHODS Using the T1-weighted MRI data from 71 patients with first-episode, treatment-naïve MDD and 69 healthy controls, we constructed the MIND network for all participants. We then performed between-group comparisons to investigate abnormalities in the network and spatial relationships between the observed patterns of MIND disruption and the patterns of neuroreceptors were estimated. Network-based spreading was utilized to explore the abnormalities constrained by brain connectivity based on structural, functional, and transcriptional connectome architecture and to further identify potential epicenters of MDD. In addition, partial least squares regression was conducted to examine the associations of gene expression profiles with MIND changes in MDD. RESULTS Patients with MDD showed significantly increased MIND in regions associated with sensation and cognition compared with healthy controls, with this altered pattern being influenced by a combination of transcriptional and structural connectivity, and potential epicenters of MDD were identified in the frontal, parietal, and paracentral cortices. Furthermore, the cortical map of case-control differences in MIND was spatially correlated with the cannabinoid CB1 receptor and the brain-wide expression of a weighted combination of genes. These genes were enriched for neurobiologically relevant pathways and preferentially expressed in different cell classes and cortical layers. CONCLUSION These results highlight the abnormal pattern of morphometric similarity observed in MDD, shedding light on the complex interplay between disrupted macroscale coordinated morphology and microscale molecular organization in MDD.
Collapse
Affiliation(s)
- Kaizhong Xue
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China; Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Feng Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Sixiang Liang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China; Tianjin Anding Hospital, Tianjin 300222, China
| | - Lining Guo
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yi Shan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China
| | - Huijuan Xu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China
| | - Jiao Xue
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China
| | - Yifan Jiang
- School of Nursing, Tianjin Medical University, Tianjin 300070, China
| | - Yong Zhang
- Tianjin Anding Hospital, Tianjin 300222, China.
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China.
| |
Collapse
|
34
|
Li M, Yang Y, Wu R, Gong H, Yuan Z, Wang J, Long E, Zhang X, Chen Y. SEE: A Method for Predicting the Dynamics of Chromatin Conformation Based on Single-Cell Gene Expression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406413. [PMID: 39778075 PMCID: PMC11848634 DOI: 10.1002/advs.202406413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 12/01/2024] [Indexed: 01/11/2025]
Abstract
The dynamics of chromatin conformation involve continuous and reversible changes within the nucleus of a cell, which participate in regulating processes such as gene expression, DNA replication, and damage repair. Here, SEE is introduced, an artificial intelligence (AI) method that utilizes autoencoder and transformer techniques to analyze chromatin dynamics using single-cell RNA sequencing data and a limited number of single-cell Hi-C maps. SEE is employed to investigate chromatin dynamics across different scales, enabling the detection of (i) rearrangements in topologically associating domains (TADs), and (ii) oscillations in chromatin interactions at gene loci. Additionally, SEE facilitates the interpretation of disease-associated single-nucleotide polymorphisms (SNPs) by leveraging the dynamic features of chromatin conformation. Overall, SEE offers a single-cell, high-resolution approach to analyzing chromatin dynamics in both developmental and disease contexts.
Collapse
Affiliation(s)
- Minghong Li
- State Key Laboratory of Common Mechanism Research for Major DiseasesDepartment of Biochemistry and Molecular BiologyInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100005China
- Department of Computer Science and TechnologyUniversity of Science and Technology BeijingBeijing100083China
| | - Yurong Yang
- State Key Laboratory of Common Mechanism Research for Major DiseasesDepartment of Biochemistry and Molecular BiologyInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100005China
| | - Rucheng Wu
- State Key Laboratory of Common Mechanism Research for Major DiseasesDepartment of Biochemistry and Molecular BiologyInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100005China
| | - Haiyan Gong
- Beijing Advanced Innovation Center for Materials Genome EngineeringUniversity of Science and Technology BeijingBeijing100083China
| | - Zan Yuan
- State Key Laboratory of Common Mechanism Research for Major DiseasesDepartment of Biochemistry and Molecular BiologyInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100005China
| | - Jixin Wang
- School of Basic MedicineTsinghua UniversityBeijing100084China
| | - Erping Long
- The State Key Laboratory of Respiratory Health and MultimorbidityInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100005China
| | - Xiaotong Zhang
- Department of Computer Science and TechnologyUniversity of Science and Technology BeijingBeijing100083China
- Shunde Innovation SchoolUniversity of Science and Technology BeijingFoshan528399China
| | - Yang Chen
- State Key Laboratory of Common Mechanism Research for Major DiseasesDepartment of Biochemistry and Molecular BiologyInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100005China
| |
Collapse
|
35
|
Joseph JF, Tural U, Joseph ND, Mendoza TE, Patel E, Reifer R, Deregnaucourt M. Understanding High-Functioning Depression in Adults. Cureus 2025; 17:e78891. [PMID: 39963293 PMCID: PMC11831407 DOI: 10.7759/cureus.78891] [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] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
INTRODUCTION High-functioning depression (HFD) is described as experiencing depressive symptoms such as fatigue, anhedonia, poor concentration, guilt, restlessness, sleep disturbances, and appetite changes without experiencing a lack of functioning or significant distress. The purpose of this study is to characterize the clinical correlates of HFD. METHODS This study entailed a descriptive, cross-sectional design based on interviews administered to120 English-speaking participants with HFD (aged 18-75). The interview involved administering a semi-structured HFD Analysis Questionnaire, the Joseph HFD Inventory, the HFD Trauma Inventory, and the Joseph HFD Anhedonia Scale in a single, 30-minute session for each participant. Big traumas, defined as extremely traumatic events, were analyzed by the trauma inventory. RESULTS Out of the 120 participants, 72 (60%) demonstrated HFD, and 17 (14%) demonstrated very HFD. A correlation was observed between symptoms of HFD, such as anhedonia and marital status, as post hoc tests showed that the average Anhedonia Scale score was higher for married or partnered participants than those who were single (p=0.038). As anticipated, the participants with higher Anhedonia Scale scores had higher HFD scores (p=0.003). These participants also experienced higher trauma inventory scores and big traumas. Furthermore, as participant education level increased, the number of big traumas reported decreased (p<0.001). Participants who were parents/caregivers of children also had the highest Anhedonia Scale and HFD scores (p=0.0126 and p=0.0210, respectively). CONCLUSION The results supported the hypothesis that individuals with HFD have increased levels of anhedonia and trauma. However, trauma scores were inversely associated with education level in HFD.
Collapse
Affiliation(s)
- Judith F Joseph
- Pediatric and Adult Psychiatry, Manhattan Behavioral Medicine, New York, USA
| | - Umit Tural
- Psychiatric Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, USA
| | - Nikeisha D Joseph
- Pediatric and Adult Psychiatry, Manhattan Behavioral Medicine, New York, USA
| | - Teresa E Mendoza
- Pediatric and Adult Psychiatry, Manhattan Behavioral Medicine, New York, USA
| | - Eshna Patel
- Pediatric and Adult Psychiatry, Manhattan Behavioral Medicine, New York, USA
| | - Rachel Reifer
- Pediatric and Adult Psychiatry, Manhattan Behavioral Medicine, New York, USA
| | | |
Collapse
|
36
|
Liao C, Dua AN, Wojtasiewicz C, Liston C, Kwan AC. Structural neural plasticity evoked by rapid-acting antidepressant interventions. Nat Rev Neurosci 2025; 26:101-114. [PMID: 39558048 PMCID: PMC11892022 DOI: 10.1038/s41583-024-00876-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 11/20/2024]
Abstract
A feature in the pathophysiology of major depressive disorder (MDD), a mood disorder, is the impairment of excitatory synapses in the prefrontal cortex. Intriguingly, different types of treatment with fairly rapid antidepressant effects (within days or a few weeks), such as ketamine, electroconvulsive therapy and non-invasive neurostimulation, seem to converge on enhancement of neural plasticity. However, the forms and mechanisms of plasticity that link antidepressant interventions to the restoration of excitatory synaptic function are still unknown. In this Review, we highlight preclinical research from the past 15 years showing that ketamine and psychedelic drugs can trigger the growth of dendritic spines in cortical pyramidal neurons. We compare the longitudinal effects of various psychoactive drugs on neuronal rewiring, and we highlight rapid onset and sustained time course as notable characteristics for putative rapid-acting antidepressant drugs. Furthermore, we consider gaps in the current understanding of drug-evoked in vivo structural plasticity. We also discuss the prospects of using synaptic remodelling to understand other antidepressant interventions, such as repetitive transcranial magnetic stimulation. Finally, we conclude that structural neural plasticity can provide unique insights into the neurobiological actions of psychoactive drugs and antidepressant interventions.
Collapse
Affiliation(s)
- Clara Liao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
| | - Alisha N Dua
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | | | - Conor Liston
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Alex C Kwan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
37
|
Liu A, Sun L, Meng W. Proteomics of neuropsychiatric disorders. Clin Chim Acta 2025; 567:120093. [PMID: 39681231 DOI: 10.1016/j.cca.2024.120093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 12/18/2024]
Abstract
The pathogenesis of neuropsychiatric disorders (NDs) remains largely unclear, hence there is a lack of objective and reliable biomarkers. Proteomics, as a powerful tool for disease biomarkers research, has been largely ignored in the field of NDs. This review summarizes recent research on the application of mass spectrometry-based proteomics in NDs. Proteins associated with NDs have been identified in various sample sources, including blood, urine, saliva, tear, cerebrospinal fluid, and brain tissue. These studies have preliminarily demonstrated the potential of proteomics in NDs and require comprehensive validation in multi-center, large-scale clinical cohorts. We also discuss the challenges and prospects of proteomics in the research of early diagnostic biomarkers for NDs. These findings may provide a foundation for developing proteomic-based diagnostics and advancing precision medicine in NDs.
Collapse
Affiliation(s)
- Afeng Liu
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China
| | - Lina Sun
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China
| | - Wenshu Meng
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China.
| |
Collapse
|
38
|
Ma Y, Jiang D, Li J, Zheng G, Deng Y, Gou X, Gao S, Chen C, Zhou Y, Zhang Y, Deng C, Yao Y, Han H, Su J. Systematic dissection of pleiotropic loci and critical regulons in excitatory neurons and microglia relevant to neuropsychiatric and ocular diseases. Transl Psychiatry 2025; 15:24. [PMID: 39856056 PMCID: PMC11760387 DOI: 10.1038/s41398-025-03243-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/08/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Advancements in single-cell multimodal techniques have greatly enhanced our understanding of disease-relevant loci identified through genome-wide association studies (GWASs). To investigate the biological connections between the eye and brain, we integrated bulk and single-cell multiomic profiles with GWAS summary statistics for eight neuropsychiatric and five ocular diseases. Our analysis uncovered five latent factors explaining 61.7% of the genetic variance across these 13 diseases, revealing diverse correlational patterns among them. We identified 45 pleiotropic loci with 91 candidate genes that contribute to disease risk. By integrating GWAS and single-cell profiles, we implicated excitatory neurons and microglia as key contributors in the eye-brain connections. Polygenic enrichment analysis further identified 15 pleiotropic regulons in excitatory neurons and 16 in microglia that were linked to comorbid conditions. Functionally, excitatory neuron-specific regulons were involved in axon guidance and synaptic activity, while microglia-specific regulons were associated with immune response and cell activation. In sum, these findings underscore the genetic link between psychiatric disorders and ocular diseases.
Collapse
Affiliation(s)
- Yunlong Ma
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Lifespan Brain Institute at Penn Med and the Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Dingping Jiang
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingjing Li
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gongwei Zheng
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yao Deng
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuanxuan Gou
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuaishuai Gao
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cheng Chen
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yijun Zhou
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yaru Zhang
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chunyu Deng
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yinghao Yao
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haijun Han
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Jianzhong Su
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
39
|
Jessa S, De Cola A, Chandarana B, McNicholas M, Hébert S, Ptack A, Faury D, Tsai JW, Korshunov A, Phoenix TN, Ellezam B, Jones DT, Taylor MD, Bandopadhayay P, Pathania M, Jabado N, Kleinman CL. FOXR2 Targets LHX6+/DLX+ Neural Lineages to Drive Central Nervous System Neuroblastoma. Cancer Res 2025; 85:231-250. [PMID: 39495206 PMCID: PMC11733536 DOI: 10.1158/0008-5472.can-24-2248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/17/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Central nervous system neuroblastoma with forkhead box R2 (FOXR2) activation (NB-FOXR2) is a high-grade tumor of the brain hemispheres and a newly identified molecular entity. Tumors express dual neuronal and glial markers, leading to frequent misdiagnoses, and limited information exists on the role of FOXR2 in their genesis. To identify their cellular origins, we profiled the transcriptomes of NB-FOXR2 tumors at the bulk and single-cell levels and integrated these profiles with large single-cell references of the normal brain. NB-FOXR2 tumors mapped to LHX6+/DLX+ lineages derived from the medial ganglionic eminence, a progenitor domain in the ventral telencephalon. In vivo prenatal Foxr2 targeting to the ganglionic eminences in mice induced postnatal cortical tumors recapitulating human NB-FOXR2-specific molecular signatures. Profiling of FOXR2 binding on chromatin in murine models revealed an association with ETS transcriptional networks, as well as direct binding of FOXR2 at key transcription factors that coordinate initiation of gliogenesis. These data indicate that NB-FOXR2 tumors originate from LHX6+/DLX+ interneuron lineages, a lineage of origin distinct from that of other FOXR2-driven brain tumors, highlight the susceptibility of ventral telencephalon-derived interneurons to FOXR2-driven oncogenesis, and suggest that FOXR2-induced activation of glial programs may explain the mixed neuronal and oligodendroglial features in these tumors. More broadly, this work underscores systematic profiling of brain development as an efficient approach to orient oncogenic targeting for in vivo modeling, critical for the study of rare tumors and development of therapeutics. Significance: Profiling the developing brain enabled rationally guided modeling of FOXR2-activated CNS neuroblastoma, providing a strategy to overcome the heterogeneous origins of pediatric brain tumors that hamper tumor modeling and therapy development. See related commentary by Orr, p. 195.
Collapse
Affiliation(s)
- Selin Jessa
- Lady Davis Research Institute, Jewish General Hospital, Montreal, Canada
- Quantitative Life Sciences, McGill University, Montreal, Canada
| | - Antonella De Cola
- Department of Oncology, Early Cancer Institute, Adrian Way, University of Cambridge, Cambridge, United Kingdom
- CRUK Children’s Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, United Kingdom
| | - Bhavyaa Chandarana
- Lady Davis Research Institute, Jewish General Hospital, Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
| | - Michael McNicholas
- Department of Oncology, Early Cancer Institute, Adrian Way, University of Cambridge, Cambridge, United Kingdom
- CRUK Children’s Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, United Kingdom
| | - Steven Hébert
- Lady Davis Research Institute, Jewish General Hospital, Montreal, Canada
| | - Adam Ptack
- Department of Experimental Medicine, McGill University, Montreal, Canada
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Damien Faury
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Jessica W. Tsai
- Department of Pediatrics, Keck School of Medicine of University of Southern California, Los Angeles, California
- Department of Pediatrics, Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, California
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, California
| | - Andrey Korshunov
- Clinical Cooperation Unit Neuropathology (B300), German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg University Hospital and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Timothy N. Phoenix
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio
| | - Benjamin Ellezam
- Department of Pathology, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, Canada
| | - David T.W. Jones
- Division of Pediatric Glioma Research, Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael D. Taylor
- Pediatric Neuro-Oncology Research Program, Texas Children’s Hospital, Houston, Texas
- Department of Pediatrics, Hematology/Oncology, Hematology/Oncology Section, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Pratiti Bandopadhayay
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Boston Children’s Cancer and Blood Disorder Center, Boston, Massachusetts
| | - Manav Pathania
- Department of Oncology, Early Cancer Institute, Adrian Way, University of Cambridge, Cambridge, United Kingdom
- CRUK Children’s Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, United Kingdom
| | - Nada Jabado
- Department of Human Genetics, McGill University, Montreal, Canada
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Pediatrics, McGill University, Montreal, Canada
| | - Claudia L. Kleinman
- Lady Davis Research Institute, Jewish General Hospital, Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
| |
Collapse
|
40
|
Lan T, Li Y, Chen X, Wang W, Wang C, Lou H, Chen S, Yu S. Exercise-Activated mPFC Tri-Synaptic Pathway Ameliorates Depression-Like Behaviors in Mouse. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408618. [PMID: 39574315 PMCID: PMC11744721 DOI: 10.1002/advs.202408618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/03/2024] [Indexed: 01/21/2025]
Abstract
Exercise is considered as playing a pivotal role in the modulation of emotional responses. However, a precise circuit that mediates the effects of exercise on depression have yet to be elucidated. Here, a molecularly defined tri-synaptic pathway circuit is identified that correlates motor inputs with antidepressant effects. With this pathway, initial inputs from neurons within the dorsal root ganglia (DRG) project to excitatory neurons in the gracile nucleus (GR), which in turn connect with 5-HTergic neurons in the dorsal raphe nucleus (DRN), eventually coursing to excitatory pyramidal neurons within the medial prefrontal cortex (mPFC). Exercise activates this pathway, with the result that depressive- and anxiety-like behaviors in mice are significantly reduced. In addition, it is found that exercise may exert antidepressant effects through regulating synaptic plasticity within this tri-synaptic pathway. These findings reveal a hindbrain-to-forebrain neuronal circuit that specifically modulates depression and provides a potential mechanism for the antidepressant effects of exercise.
Collapse
Affiliation(s)
- Tian Lan
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Ye Li
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Xiao Chen
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Wenjing Wang
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Changmin Wang
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Haiyan Lou
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PharmacologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Shihong Chen
- Department of Endocrinology and MetabolismThe Second Hospital of Shandong UniversityJinanShandong250033China
| | - Shuyan Yu
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
- Department of Medical Psychology and EthicsSchool of Basic Medical sciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| |
Collapse
|
41
|
Snijders GJLJ, Gigase FAJ. Neuroglia in mood disorders. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:287-302. [PMID: 40148049 DOI: 10.1016/b978-0-443-19102-2.00010-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Multiple lines of evidence indicate that mood disorders, such as major depressive and bipolar disorder, are associated with abnormalities in neuroglial cells. This chapter discusses the existing literature investigating the potential role of astrocytes, oligodendrocytes, and microglia in mood pathology. We will describe evidence from in vivo imaging, postmortem, animal models based on (stress) paradigms that mimic depressive-like behavior, and biomarker studies in blood and cerebrospinal fluid in patients with mood disorders. The effect of medication used in the treatment of mood disorders, such as antidepressants and lithium, on glial function is discussed. Lastly, we highlight the most relevant findings about potential deficiencies in glia-glia crosstalk in mood disorders. Overall, decreased astrocyte and oligodendrocyte density and expression and microglial changes in homeostatic functions have frequently been put forward in MDD pathology. Studies of BD report similar findings to some extent; however, the evidence is less well established. Together, these findings are suggestive of reduced glial cell function leading to potential white matter abnormalities, glutamate dysregulation, disrupted neuronal functioning, and neurotransmission. However, more research is required to better understand the exact mechanisms underlying glial cell contributions to mood disorder development.
Collapse
Affiliation(s)
- Gijsje J L J Snijders
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| | - Frederieke A J Gigase
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
42
|
Song X, Yan S, Lai S, Zhang Y, Wang Y, He J, Huang D, Zhang J, Lu X, Chen G, Chen P, Zhong Q, Zhang R, Wu Y, Yin J, Zhong S, Jia Y. Gender differences of neurometabolic and neuroendocrine alternations and its lateralization in adolescents with major depressive disorder. BMC Psychiatry 2024; 24:949. [PMID: 39731037 DOI: 10.1186/s12888-024-06428-1] [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: 11/29/2023] [Accepted: 12/19/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND The clinical characteristics of major depressive disorder (MDD) in adolescents show notable gender-related differences, but the cause of these differences is still not understood. The current research concentrates on the changes in neurometabolism and neuroendocrine function, aiming to identify differences in endocrine function and brain metabolism between male and female adolescents with MDD. METHODS A total of 121 teenagers diagnosed with MDD (43 males and 78 females) were enlisted as participants. Measurement was conducted on levels of endocrine hormones, which included free tri-iodothyronine (FT3), total tri-iodothyronine (TT3), free thyroxin (FT4), total thyroxin (TT4), thyroid-stimulating hormone (TSH), cortisol, and adrenocorticotropic hormone (ACTH). Obtained through 1H-MRS, the N-acetyl aspartate (NAA) and choline containing compounds (Cho) to creatine (Cr) ratios were acquired for the prefrontal whiter matter (PWM), anterior cingulate cortex (ACC), basal ganglia (BG), thalamus, and cerebellum. RESULT After adjusting for multiple comparisons, female adolescents with MDD showed lower ACTH levels compared to their male counterparts. An increased lateralization index (LI) was observed in female patients for both the thalamic Cho/Cr ratio and the basal ganglia NAA/Cr ratio. Additionally, an intriguing finding was that in male adolescent patients, TT4 levels were significantly correlated with the Cho/Cr ratio in the left cerebellum. However, no such correlation between hormones and brain metabolism was found in females. CONCLUSIONS Gender differences in endocrine and neurometabolic abnormalities may contribute to the gender-specific pathophysiology of MDD in adolescent patients. Metabolic abnormalities and lateralization changes are observed in different brain regions for male and female MDD patients.
Collapse
Affiliation(s)
- Xiaodong Song
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Shuya Yan
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Shunkai Lai
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Yiliang Zhang
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Ying Wang
- Medical Imaging Center, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Jiali He
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Dong Huang
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Jianzhao Zhang
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Xiaodan Lu
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Guanmao Chen
- Medical Imaging Center, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Pan Chen
- Medical Imaging Center, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Qilin Zhong
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Rongxu Zhang
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Yangyu Wu
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Jie Yin
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Shuming Zhong
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China.
| | - Yanbin Jia
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
43
|
Mitsuhashi H, Lin R, Chawla A, Mechawar N, Nagy C, Turecki G. Altered m6A RNA methylation profiles in depression implicate the dysregulation of discrete cellular functions in males and females. iScience 2024; 27:111316. [PMID: 39650737 PMCID: PMC11625292 DOI: 10.1016/j.isci.2024.111316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/03/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024] Open
Abstract
Adverse environmental stress represents a significant risk factor for major depressive disorder (MDD), often resulting in disrupted synaptic connectivity which is known to be partly regulated by epigenetic mechanisms. N6-methyladenosine (m6A), an epitranscriptomic modification, has emerged as a crucial regulator of activity-dependent gene regulation. In this study, we characterized m6A profiles in the ventromedial prefrontal cortex (vmPFC) of individuals with MDD. Using m6A sequencing, we identified a total of 30,279 high-confidence m6A peaks, exhibiting significant enrichment in genes related to neuronal and synaptic function. The m6A peaks between males and females with MDD that passed the significance threshold showed opposite m6A patterns, while the threshold-free m6A patterns were concordant. Distinct m6A profiles were found in MDD for each sex, with dysregulation associated with microtubule movement in males and neuronal projection in females. Our results suggest the potential roles of m6A as part of the dysregulated molecular network in MDD.
Collapse
Affiliation(s)
- Haruka Mitsuhashi
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| | - Rixing Lin
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544 USA, USA
| | - Anjali Chawla
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
44
|
Zhuo Z, Lu W, Zhang L, Zhang D, Cui Y, Wu X, Mei H, Chang L, Song Q. Transcriptomic analysis reveals potential crosstalk genes and immune relationship between triple-negative breast cancer and depression. Discov Oncol 2024; 15:762. [PMID: 39692924 DOI: 10.1007/s12672-024-01562-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 11/11/2024] [Indexed: 12/19/2024] Open
Abstract
TNBC, the most aggressive form of breast cancer, lacks accurate and effective therapeutic targets. Immunotherapy presents a promising approach for addressing TNBC. Anxiety and depression are frequently concurrent symptoms in TNBC patients. MDD affects the tumor immune microenvironment of TNBC, with its characteristic genes affecting the pathophysiology of MDD and potentially increasing the risk of TNBC recurrence and metastasis. This study reveals significant differences in T lymphocyte infiltration between high-risk and low-risk TNBC groups based on MDD feature genes. This finding aids in identifying TNBC patients who may benefit from immunotherapy, providing new insights for future TNBC immunotherapy strategies. Our aim is to identify MDD-related genes involved in the pathogenesis of TNBC and to provide predictive biomarkers for TNBC immunotherapy.
Collapse
Affiliation(s)
- Zhili Zhuo
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Wenping Lu
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China.
| | - Ling Zhang
- Department of pathology, China Academy of Chinese Medical Sciences Guang' anmen Hospital, Beijing, 100053, China
| | - Dongni Zhang
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Yongjia Cui
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Xiaoqing Wu
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Heting Mei
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Lei Chang
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Qingya Song
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China
| |
Collapse
|
45
|
Boldrini M, Xiao Y, Singh T, Zhu C, Jabbi M, Pantazopoulos H, Gürsoy G, Martinowich K, Punzi G, Vallender EJ, Zody M, Berretta S, Hyde TM, Kleinman JE, Marenco S, Roussos P, Lewis DA, Turecki G, Lehner T, Mann JJ. Omics Approaches to Investigate the Pathogenesis of Suicide. Biol Psychiatry 2024; 96:919-928. [PMID: 38821194 PMCID: PMC11563882 DOI: 10.1016/j.biopsych.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
Suicide is the second leading cause of death in U.S. adolescents and young adults and is generally associated with a psychiatric disorder. Suicidal behavior has a complex etiology and pathogenesis. Moderate heritability suggests genetic causes. Associations between childhood and recent life adversity indicate contributions from epigenetic factors. Genomic contributions to suicide pathogenesis remain largely unknown. This article is based on a workshop held to design strategies to identify molecular drivers of suicide neurobiology that would be putative new treatment targets. The panel determined that while bulk tissue studies provide comprehensive information, single-nucleus approaches that identify cell type-specific changes are needed. While single-nuclei techniques lack information on cytoplasm, processes, spines, and synapses, spatial multiomic technologies on intact tissue detect cell alterations specific to brain tissue layers and subregions. Because suicide has genetic and environmental drivers, multiomic approaches that combine cell type-specific epigenome, transcriptome, and proteome provide a more complete picture of pathogenesis. To determine the direction of effect of suicide risk gene variants on RNA and protein expression and how these interact with epigenetic marks, single-nuclei and spatial multiomics quantitative trait loci maps should be integrated with whole-genome sequencing and genome-wide association databases. The workshop concluded with a recommendation for the formation of an international suicide biology consortium that will bring together brain banks and investigators with expertise in cutting-edge omics technologies to delineate the biology of suicide and identify novel potential treatment targets to be tested in cellular and animal models for drug and biomarker discovery to guide suicide prevention.
Collapse
Affiliation(s)
- Maura Boldrini
- Department of Psychiatry, Columbia University, New York, New York; Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York.
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Tarjinder Singh
- Department of Psychiatry, Columbia University, New York, New York; Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York; New York Genome Center, New York, New York
| | - Chenxu Zhu
- New York Genome Center, New York, New York; Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York
| | - Mbemba Jabbi
- Department of Psychiatry and Behavioral Sciences, Mulva Clinics for the Neurosciences, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi
| | - Gamze Gürsoy
- New York Genome Center, New York, New York; Departments of Biomedical Informatics and Computer Science, Columbia University, New York, New York
| | - Keri Martinowich
- Lieber Institute for Brain Development, Department of Psychiatry and Behavioral Sciences, Baltimore, Maryland
| | - Giovanna Punzi
- Lieber Institute for Brain Development, Department of Psychiatry and Behavioral Sciences, Baltimore, Maryland
| | - Eric J Vallender
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi
| | | | - Sabina Berretta
- Department of Psychiatry, Harvard Brain Tissue Resource Center, Harvard Medical School, McLean Hospital, Belmont, Massachusetts
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Department of Psychiatry and Behavioral Sciences, Baltimore, Maryland
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Department of Psychiatry and Behavioral Sciences, Baltimore, Maryland
| | - Stefano Marenco
- Human Brain Collection Core, National Institute of Mental Health's (NIMH) Division of Intramural Research Programs, Bethesda, Maryland
| | - Panagiotis Roussos
- Center for Precision Medicine and Translational Therapeutics, Mental Illness Research Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, New York
| | - David A Lewis
- Departments of Psychiatry and Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Gustavo Turecki
- Department of Psychiatry, Douglas Institute, McGill University, Montréal, Québec, Canada
| | | | - J John Mann
- Department of Psychiatry, Columbia University, New York, New York; Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York
| |
Collapse
|
46
|
Wu S, Jiang Q, Wang J, Wu D, Ren Y. Immune-related gene characterization and biological mechanisms in major depressive disorder revealed based on transcriptomics and network pharmacology. Front Psychiatry 2024; 15:1485957. [PMID: 39713769 PMCID: PMC11659238 DOI: 10.3389/fpsyt.2024.1485957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/08/2024] [Indexed: 12/24/2024] Open
Abstract
Background Major depressive disorder (MDD) is a severe psychiatric disorder characterized by complex etiology, with genetic determinants that are not fully understood. The objective of this study was to investigate the pathogenesis of MDD and to explore its association with the immune system by identifying hub biomarkers using bioinformatics analyses and examining immune infiltrates in human autopsy samples. Methods Gene microarray data were obtained from the Gene Expression Omnibus (GEO) datasets GSE32280, GSE76826, GSE98793, and GSE39653. Our approach included differential expression analysis, weighted gene co-expression network analysis (WGCNA), and protein-protein interaction (PPI) network analysis to identify hub genes associated with MDD. Subsequently, gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Cytoscape plugin CluGO, and Gene Set Enrichment Analysis (GSEA) were utilized to identify immune-related genes. The final selection of immune-related hub genes was determined through the least absolute shrinkage and selection operator (Lasso) regression analysis and PPI analysis. Immune cell infiltration in MDD patients was analyzed using CIBERSORT, and correlation analysis was performed between key immune cells and genes. The diagnostic accuracy of the identified hub genes was evaluated using receiver operating characteristic (ROC) curve analysis. Furthermore, we conducted a study involving 10 MDD patients and 10 healthy controls (HCs) meeting specific criteria to assess the expression levels of these hub genes in their peripheral blood mononuclear cells (PBMCs). The Herbal Ingredient Target Database (HIT) was employed to screen for herbal components that target these genes, potentially identifying novel therapeutic agents. Results A total of 159 down-regulated and 51 up-regulated genes were identified for further analysis. WGCNA revealed 12 co-expression modules, with modules "darked", "darkurquoise" and "light yellow" showing significant positive associations with MDD. Functional enrichment pathway analysis indicated that these differential genes were associated with immune functions. Integration of differential and immune-related gene analysis identified 21 common genes. The Lasso algorithm confirmed 4 hub genes as potential biomarkers for MDD. GSEA analysis suggested that these genes may be involved in biological processes such as protein export, RNA degradation, and fc gamma r mediated cytotoxis. Pathway enrichment analysis identified three highly enriched immune-related pathways associated with the 4 hub genes. ROC curve analysis indicated that these hub genes possess good diagnostic value. Quantitative reverse transcription-polymerase chain reaction (RT-qPCR) demonstrated significant expression differences of these hub genes in PBMCs between MDD patients and HCs. Immune infiltration analysis revealed significant correlations between immune cells, including Mast cells resting, T cells CD8, NK cells resting, and Neutrophils, which were significantly correlated with the hub genes expression. HIT identified one herb target related to IL7R and 14 targets related to TLR2. Conclusions The study identified four immune-related hub genes (TLR2, RETN, HP, and IL7R) in MDD that may impact the diagnosis and treatment of the disorder. By leveraging the GEO database, our findings contribute to the understanding of the relationship between MDD and immunity, presenting potential therapeutic targets.
Collapse
Affiliation(s)
- Shasha Wu
- Department of Psychiatry, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Jiang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Jinhui Wang
- Department of Pharmacy, Shanxi Medical University, Taiyuan, China
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Daming Wu
- Department of Psychiatry, Xiaoyi City Central Hospital, Xiaoyi, China
| | - Yan Ren
- Department of Psychiatry, The Fifth Hospital of Shanxi Medical University, The Fifth Clinical Medical College of Shanxi Medical University, Shanxi Provincial People’s Hospital, Taiyuan, China
| |
Collapse
|
47
|
Adameyko I, Bakken T, Bhaduri A, Chhatbar C, Filbin MG, Gate D, Hochgerner H, Kim CN, Krull J, La Manno G, Li Q, Linnarsson S, Ma Q, Mayer C, Menon V, Nano P, Prinz M, Quake S, Walsh CA, Yang J, Bayraktar OA, Gokce O, Habib N, Konopka G, Liddelow SA, Nowakowski TJ. Applying single-cell and single-nucleus genomics to studies of cellular heterogeneity and cell fate transitions in the nervous system. Nat Neurosci 2024; 27:2278-2291. [PMID: 39627588 PMCID: PMC11949301 DOI: 10.1038/s41593-024-01827-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 10/22/2024] [Indexed: 12/13/2024]
Abstract
Single-cell and single-nucleus genomic approaches can provide unbiased and multimodal insights. Here, we discuss what constitutes a molecular cell atlas and how to leverage single-cell omics data to generate hypotheses and gain insights into cell transitions in development and disease of the nervous system. We share points of reflection on what to consider during study design and implementation as well as limitations and pitfalls.
Collapse
Affiliation(s)
- Igor Adameyko
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Aparna Bhaduri
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Chintan Chhatbar
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Mariella G Filbin
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston Children's Hospital, Boston, MA, USA
| | - David Gate
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hannah Hochgerner
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Chang Nam Kim
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | - Jordan Krull
- Department of Biomedical Informatics, College of Medicine, the Ohio State University, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology, the James Comprehensive Cancer Center, the Ohio State University, Columbus, OH, USA
| | - Gioele La Manno
- Laboratory of Neurodevelopmental Systems Biology, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Qingyun Li
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Sten Linnarsson
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, the Ohio State University, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology, the James Comprehensive Cancer Center, the Ohio State University, Columbus, OH, USA
| | - Christian Mayer
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Vilas Menon
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Patricia Nano
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Steve Quake
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Christopher A Walsh
- Division of Genetics and Genomics, Manton Center for Orphan Disease, Boston Children's Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
| | - Jin Yang
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | | | - Ozgun Gokce
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, Bonn, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| | - Naomi Habib
- The Edmond and Lily Safra Center for Brain Sciences, the Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA.
- Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY, USA.
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA.
| | - Tomasz J Nowakowski
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA.
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
48
|
Luo Y, Zhang Y, Feng Y, Zeng X, Zhu D, Yang Y, Hu H, Wang Q, Guo L, Zou L, Zhong X. Prenatal exposure to low doses of benzophenone-3 elicits disruption of cortical vasculature in fetuses through perturbations in Wnt/β-catenin signaling correlating with depression-like behavior in offspring mice. Toxicology 2024; 509:153960. [PMID: 39343157 DOI: 10.1016/j.tox.2024.153960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/12/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Benzophenone-3 (BP-3), commonly used in personal care products, is routinely detected in environmental and human matrices. Evidence delineates a correlation between gestational BP-3 exposure and emotional and social disorders in children and adolescents. However, sensitive target cells and the mode of action underlying the early responses to environmentally relevant level of BP-3 exposure remain unclear. In this study, 0.3 and 3 mg/kg of BP-3 were administered to pregnant mice. Compared with the control group, the cortical blood vessel development process manifested the highest susceptibility to BP-3 exposure using transcriptomic sequencing at embryonic day 14 (E14). Notably, the diminution in vascular density and tight junction proteins presence was observed in the fetal cortex at E14, concomitant with the suppressed transcriptional activity of genes essential to angiogenesis and barrier formation. Strikingly, the investigation revealed that BP-3 exposure impeded vascular sprouting in aortic ring explants and neuroendothelial migration, implicating the Wnt/β-catenin signaling pathway. Moreover, BP-3 exposure compromised perivascular neural stem cell differentiation. Cortical vascular injury correlated with the exhibition of depression-like behavior in four-week postnatal progeny. These insights underscore the cerebrovasculature as an early sensitive target for low doses of BP-3 exposure, fostering the development of biomarkers and the establishment of the adverse outcome pathway framework for BP-3 hazard evaluation.
Collapse
Affiliation(s)
- Yijun Luo
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yangjian Zhang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yang Feng
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China; The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan 410005, China
| | - Xiangyu Zeng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Dan Zhu
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China; The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan 410005, China
| | - Ying Yang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Haichen Hu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Qi Wang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Lan Guo
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Lianhong Zou
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China; The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan 410005, China; Geriatric Immunization Research Center of Hunan Provincial Geriatric Institute, Changsha, Hunan, China.
| | - Xiali Zhong
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
49
|
Zhong X, Chen Y, Chen W, Liu Y, Gui S, Pu J, Wang D, He Y, Chen X, Chen X, Qiao R, Xie P. Identification of Potential Biomarkers for Major Depressive Disorder: Based on Integrated Bioinformatics and Clinical Validation. Mol Neurobiol 2024; 61:10355-10364. [PMID: 38722514 DOI: 10.1007/s12035-024-04217-1] [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: 03/15/2024] [Accepted: 05/06/2024] [Indexed: 11/24/2024]
Abstract
Major depressive disorder (MDD) is a severe mental illness characterized by a lack of objective biomarkers. Mounting evidence suggests there are extensive transcriptional molecular changes in the prefrontal cortex (PFC) of individuals with MDD. However, it remains unclear whether there are specific genes that are consistently altered and possess diagnostic power. In this study, we conducted a systematic search of PFC datasets of MDD patients from the Gene Expression Omnibus database. We calculated the differential expression of genes (DEGs) and identified robust DEGs using the RRA and MetaDE methods. Furthermore, we validated the consistently altered genes and assessed their diagnostic power through enzyme-linked immunosorbent assay experiments in our clinical blood cohort. Additionally, we evaluated the diagnostic power of hub DEGs in independent public blood datasets. We obtained eight PFC datasets, comprising 158 MDD patients and 263 healthy controls, and identified a total of 1468 unique DEGs. Through integrated analysis, we identified 290 robustly altered DEGs. Among these, seven hub DEGs (SLC1A3, PON2, AQP1, EFEMP1, GJA1, CENPD, HSD11B1) were significantly down-regulated at the protein level in our clinical blood cohort. Moreover, these hub DEGs exhibited a negative correlation with the Hamilton Depression Scale score (P < 0.05). Furthermore, these hub DEGs formed a panel with promising diagnostic power in three independent public blood datasets (average AUCs of 0.85) and our clinical blood cohort (AUC of 0.92). The biomarker panel composed of these genes demonstrated promising diagnostic efficacy for MDD and serves as a useful tool for its diagnosis.
Collapse
Affiliation(s)
- Xiaogang Zhong
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, China
| | - Yue Chen
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Weiyi Chen
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiyun Liu
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, China
| | - Siwen Gui
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, China
| | - Juncai Pu
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Dongfang Wang
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, China
| | - Yong He
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, China
| | - Xiang Chen
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaopeng Chen
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Renjie Qiao
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Peng Xie
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China.
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- The Jin Feng Laboratory, Chongqing, 401329, China.
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
50
|
Zhang T, Zhang X, Wu Z, Ren J, Zhao Z, Zhang H, Wang G, Wang T. VGAE-CCI: variational graph autoencoder-based construction of 3D spatial cell-cell communication network. Brief Bioinform 2024; 26:bbae619. [PMID: 39581873 PMCID: PMC11586124 DOI: 10.1093/bib/bbae619] [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: 08/23/2024] [Revised: 10/04/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024] Open
Abstract
Cell-cell communication plays a critical role in maintaining normal biological functions, regulating development and differentiation, and controlling immune responses. The rapid development of single-cell RNA sequencing and spatial transcriptomics sequencing (ST-seq) technologies provides essential data support for in-depth and comprehensive analysis of cell-cell communication. However, ST-seq data often contain incomplete data and systematic biases, which may reduce the accuracy and reliability of predicting cell-cell communication. Furthermore, other methods for analyzing cell-cell communication mainly focus on individual tissue sections, neglecting cell-cell communication across multiple tissue layers, and fail to comprehensively elucidate cell-cell communication networks within three-dimensional tissues. To address the aforementioned issues, we propose VGAE-CCI, a deep learning framework based on the Variational Graph Autoencoder, capable of identifying cell-cell communication across multiple tissue layers. Additionally, this model can be applied to spatial transcriptomics data with missing or partially incomplete data and can clustered cells at single-cell resolution based on spatial encoding information within complex tissues, thereby enabling more accurate inference of cell-cell communication. Finally, we tested our method on six datasets and compared it with other state of art methods for predicting cell-cell communication. Our method outperformed other methods across multiple metrics, demonstrating its efficiency and reliability in predicting cell-cell communication.
Collapse
Affiliation(s)
- Tianjiao Zhang
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Xiang Zhang
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Zhenao Wu
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Jixiang Ren
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Zhongqian Zhao
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Hongfei Zhang
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Guohua Wang
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
| | - Tao Wang
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|