1
|
Qu W, Yan G, Du Y, Zhou X, Huang C, Li B, Zhou J, Li Q. Crosstalk Between Mitochondrial DNA and Immune Response: Focus on Autism Spectrum Disorder. Mol Neurobiol 2025; 62:5629-5639. [PMID: 39589631 DOI: 10.1007/s12035-024-04637-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/16/2024] [Indexed: 11/27/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by multiple dysfunctions in behavior, the nervous system, and the immune system. Increasing evidence suggests that mitochondrial DNA (mtDNA) plays a crucial role in the pathology of ASD. In clinical practice, altered mtDNA levels have been observed in various tissues of individuals with ASD. Mutation or oxidation of mtDNA is also closely related to the immune response associated with the pathology of autism. With mtDNA identified as a causal factor, much interest has focused on how its production affects neurodevelopment and neurophysiology. Here, we review how mtDNA leads to dysfunction of cellular mitochondria and immune response. We also illustrate the relationship between mtDNA alterations and the pathology of autism. Finally, we discuss the existing evidence on cell-free mtDNA associated with ASD and look forward to its application in clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Wenxuan Qu
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Ge Yan
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Yajuan Du
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Xinyang Zhou
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Chutian Huang
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Bei Li
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Junmei Zhou
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Qian Li
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China.
| |
Collapse
|
2
|
Wang B, Zhang X, Wang S, Lin N, Li Y, Yu Y, Zhang Y, Yang J, Wu X, He Y, Wang S, Wan T, Chen R, Li G, Deng Y, Qi X, Wang Z, Shang D. Topology optimization of random memristors for input-aware dynamic SNN. SCIENCE ADVANCES 2025; 11:eads5340. [PMID: 40238875 PMCID: PMC12002125 DOI: 10.1126/sciadv.ads5340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 03/12/2025] [Indexed: 04/18/2025]
Abstract
Machine learning has advanced unprecedentedly, exemplified by GPT-4 and SORA. However, they cannot parallel human brains in efficiency and adaptability due to differences in signal representation, optimization, runtime reconfigurability, and hardware architecture. To address these challenges, we introduce pruning optimization for input-aware dynamic memristive spiking neural network (PRIME). PRIME uses spiking neurons to emulate brain's spiking mechanisms and optimizes the topology of random memristive SNNs inspired by structural plasticity, effectively mitigating memristor programming stochasticity. It also uses the input-aware early-stop policy to reduce latency and leverages memristive in-memory computing to mitigate von Neumann bottleneck. Validated on a 40-nm, 256-K memristor-based macro, PRIME achieves comparable classification accuracy and inception score to software baselines, with energy efficiency improvements of 37.8× and 62.5×. In addition, it reduces computational loads by 77 and 12.5% with minimal performance degradation and demonstrates robustness to stochastic memristor noise. PRIME paves the way for brain-inspired neuromorphic computing.
Collapse
Affiliation(s)
- Bo Wang
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
- ACCESS – AI Chip Center for Emerging Smart Systems, InnoHK Centers, Hong Kong Science Park, Hong Kong, China
| | - Xinyuan Zhang
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
- ACCESS – AI Chip Center for Emerging Smart Systems, InnoHK Centers, Hong Kong Science Park, Hong Kong, China
| | - Shaocong Wang
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
- ACCESS – AI Chip Center for Emerging Smart Systems, InnoHK Centers, Hong Kong Science Park, Hong Kong, China
| | - Ning Lin
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
- ACCESS – AI Chip Center for Emerging Smart Systems, InnoHK Centers, Hong Kong Science Park, Hong Kong, China
| | - Yi Li
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
- State Key Lab of Fabrication Technologies for Integrated Circuits, Institute of Microelectronics, Chinese Academy of Sciences, Beijing 100029, China
- Laboratory of Microelectronic Devices & Integrated Technology, Institute of Microelectronics, Chinese Academy of Sciences, Beijing 100029, China
| | - Yifei Yu
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
- ACCESS – AI Chip Center for Emerging Smart Systems, InnoHK Centers, Hong Kong Science Park, Hong Kong, China
| | - Yue Zhang
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
- ACCESS – AI Chip Center for Emerging Smart Systems, InnoHK Centers, Hong Kong Science Park, Hong Kong, China
| | - Jichang Yang
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
- ACCESS – AI Chip Center for Emerging Smart Systems, InnoHK Centers, Hong Kong Science Park, Hong Kong, China
| | - Xiaoshan Wu
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
- ACCESS – AI Chip Center for Emerging Smart Systems, InnoHK Centers, Hong Kong Science Park, Hong Kong, China
| | - Yangu He
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
- ACCESS – AI Chip Center for Emerging Smart Systems, InnoHK Centers, Hong Kong Science Park, Hong Kong, China
| | - Songqi Wang
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
- ACCESS – AI Chip Center for Emerging Smart Systems, InnoHK Centers, Hong Kong Science Park, Hong Kong, China
| | - Tao Wan
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Rui Chen
- State Key Lab of Fabrication Technologies for Integrated Circuits, Institute of Microelectronics, Chinese Academy of Sciences, Beijing 100029, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guoqi Li
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Yue Deng
- School of Artificial Intelligence, Beihang University, Beijing 100191, China
- School of Astronautics, Beihang University, Beijing 100191, China
| | - Xiaojuan Qi
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Zhongrui Wang
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
- ACCESS – AI Chip Center for Emerging Smart Systems, InnoHK Centers, Hong Kong Science Park, Hong Kong, China
| | - Dashan Shang
- State Key Lab of Fabrication Technologies for Integrated Circuits, Institute of Microelectronics, Chinese Academy of Sciences, Beijing 100029, China
- Laboratory of Microelectronic Devices & Integrated Technology, Institute of Microelectronics, Chinese Academy of Sciences, Beijing 100029, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
3
|
Nishi Y, Toritsuka M, Takada R, Ishikawa M, Ishida R, Kayashima Y, Yamauchi T, Okumura K, Takeda T, Yamamuro K, Ikehara M, Noriyama Y, Kamikawa K, Murayama S, Ichikawa O, Nagata H, Okano H, Iwata N, Makinodan M. Impaired synaptosome phagocytosis in macrophages of individuals with autism spectrum disorder. Mol Psychiatry 2025:10.1038/s41380-025-03002-3. [PMID: 40185900 DOI: 10.1038/s41380-025-03002-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 03/04/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
Dendritic spine abnormalities are believed to be one of the critical etiologies of autism spectrum disorder (ASD). Over the past decade, the importance of microglia in brain development, particularly in synaptic elimination, has become evident. Thus, microglial abnormalities may lead to synaptic dysfunction, which may underlie the pathogenesis of ASD. Several human studies have demonstrated aberrant microglial activation in the brains of individuals with ASD, and studies in animal models of ASD have also shown a relationship between microglial dysfunction and synaptic abnormalities. However, there are very few methods available to directly assess whether phagocytosis by human microglia is abnormal. Microglia are tissue-resident macrophages with phenotypic similarities to monocyte-derived macrophages, both of which consistently exhibit pathological phenotypes in individuals with ASD. Therefore, in this study, we examined the phagocytosis capacity of human macrophages derived from peripheral blood monocytes. These macrophages were polarized into two types: those induced by granulocyte-macrophage colony-stimulating factor (GM-CSF MΦ, traditionally referred to as "M1 MΦ") and those induced by macrophage colony-stimulating factor (M-CSF MΦ, traditionally referred to as "M2 MΦ"). Synaptosomes purified from human induced pluripotent stem cell-derived neuron were used to assess phagocytosis capacity. Our results revealed that M-CSF MΦ exhibited higher phagocytosis capacity compared to GM-CSF MΦ, whereas ASD-M-CSF MΦ showed a marked impairment in phagocytosis. Additionally, we found a positive correlation between phagocytosis capacity and cluster of differentiation 209 expression. This research contributes to a deeper understanding of the pathobiology of ASD and offers new insights into potential therapeutic targets for the disorder.
Collapse
Affiliation(s)
- Yuki Nishi
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Nara, 634-8522, Japan
| | - Michihiro Toritsuka
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Nara, 634-8522, Japan.
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan.
- Division of Transformative Psychiatry and Synergistic Research, International Center for Brain Science (ICBS), Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan.
| | - Ryohei Takada
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Nara, 634-8522, Japan
| | - Mitsuru Ishikawa
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
- Division of CNS Regeneration and Drug Discovery, International Center for Brain Science (ICBS), Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan
| | - Rio Ishida
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Nara, 634-8522, Japan
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan
- Division of Transformative Psychiatry and Synergistic Research, International Center for Brain Science (ICBS), Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan
| | - Yoshinori Kayashima
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Nara, 634-8522, Japan
| | - Takahira Yamauchi
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Nara, 634-8522, Japan
| | - Kazuki Okumura
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Nara, 634-8522, Japan
| | - Tsutomu Takeda
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Nara, 634-8522, Japan
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan
- Division of Transformative Psychiatry and Synergistic Research, International Center for Brain Science (ICBS), Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan
| | - Kazuhiko Yamamuro
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Nara, 634-8522, Japan
- Center for Health Control, Nara Medical University School of Medicine, Kashihara, Nara, 634-8522, Japan
| | - Minobu Ikehara
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Nara, 634-8522, Japan
| | - Yuki Noriyama
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Nara, 634-8522, Japan
| | - Kohei Kamikawa
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Nara, 634-8522, Japan
| | - Shuhei Murayama
- Research and Development, Sumitomo Pharma Co., Ltd., Osaka city, Osaka, 554-0022, Japan
| | - Osamu Ichikawa
- Research and Development, Sumitomo Pharma Co., Ltd., Osaka city, Osaka, 554-0022, Japan
| | - Hidetaka Nagata
- Research and Development, Sumitomo Pharma Co., Ltd., Osaka city, Osaka, 554-0022, Japan
| | - Hideyuki Okano
- Division of CNS Regeneration and Drug Discovery, International Center for Brain Science (ICBS), Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan
- Keio University Regenerative Medicine Research Center Research Gate Building TONOMACHI 2-4F, 3-25-10 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-0821, Japan
| | - Nakao Iwata
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan
| | - Manabu Makinodan
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, Nara, 634-8522, Japan.
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan.
- Division of Transformative Psychiatry and Synergistic Research, International Center for Brain Science (ICBS), Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan.
| |
Collapse
|
4
|
Xie W, Luo Z, Xiao J, Zhang X, Zhang C, Yang P, Li L. Identification of biomarkers related to propionate metabolism in schizophrenia. Front Psychiatry 2025; 16:1504699. [PMID: 40242178 PMCID: PMC12000038 DOI: 10.3389/fpsyt.2025.1504699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/04/2025] [Indexed: 04/18/2025] Open
Abstract
Purpose Schizophrenia (SCZ) is a severe mental disorder with complex etiology. Research shows propionate metabolism is crucial for neurological function and health. This suggests abnormalities in propionate metabolism may link to SCZ. Therefore, identifying biomarkers associated with propionate metabolism might be beneficial for the diagnosis and treatment of SCZ patients. Methods SCZ datasets and propionate metabolism-related genes (PMRGs) from public databases were obtained. DE-PMRGs were identified through differential and correlation analysis of PMRGs. Machine learning was used to screen for key genes and validate expression levels, aiming to identify potential biomarkers. Gene Set Enrichment Analysis (GSEA) and immune infiltration analysis were performed on the biomarkers. An upstream regulatory network was constructed, and potential drugs targeting these biomarkers were explored. Finally, real-time fluorescence quantitative PCR (qPCR) was used to verify biomarker expression levels. Result A total of 11 DE-PMRGs were identified, and machine learning technology was employed to further screen for 5 key genes. Among these, LY96 and TMEM123 emerged as potential biomarkers through expression verification. A diagnostic model was developed, achieving an area under the curve (AUC) greater than 0.7, which indicates strong diagnostic performance. Additionally, nomograms based on these biomarkers demonstrated promising predictive capabilities in assessing the risk of SCZ. To explore gene functions and regulatory mechanisms at a deeper level, a competitive endogenous RNA (ceRNA) regulatory network was constructed, including 2 biomarkers, 72 microRNAs, and 202 long non-coding RNAs. In addition, a regulatory network containing 2 biomarkers and 104 transcription factors (TFs) was also established to investigate the transcription factors interacting with the biomarkers. Potential biomarker-targeted drugs were identified by exploring the DrugBank database; notably, LY96 exhibited higher binding affinities for four drugs, with docking scores consistently below-5 kcal/mol. The qPCR results indicated that the expression levels of LY96 and TMEM123 in the whole blood of SCZ patients were significantly higher than those in the healthy control group, which was consistent with the results in the GSE38484 and GSE27383 datasets. Conclusion This study identified disease diagnostic biomarkers associated with propionate metabolism in SCZ, specifically LY96 and TMEM123. These findings offer novel perspectives for the diagnosis and management of SCZ.
Collapse
Affiliation(s)
| | | | | | | | | | - Ping Yang
- School of Clinical Medicine, Hunan Brain Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Liang Li
- School of Clinical Medicine, Hunan Brain Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
5
|
Orhan F, Malwade S, Khanlarkhani N, Gkogka A, Langeder A, Jungholm O, Koskuvi M, Lehtonen Š, Schwieler L, Jardemark K, Tiihonen J, Koistinaho J, Erhardt S, Engberg G, Samudyata S, Sellgren CM. Kynurenic Acid and Promotion of Activity-Dependent Synapse Elimination in Schizophrenia. Am J Psychiatry 2025; 182:389-400. [PMID: 40165559 DOI: 10.1176/appi.ajp.20240048] [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: 04/02/2025]
Abstract
OBJECTIVE Schizophrenia is a neurodevelopmental disorder characterized by an excessive loss of synapses. Kynurenic acid (KYNA), a neuroactive metabolite of tryptophan along the kynurenine pathway, can induce schizophrenia-related phenotypes in rodents, and clinical studies have revealed elevated KYNA levels in the CNS of individuals with schizophrenia. However, the factors that cause elevated KYNA levels in schizophrenia, and the mechanisms by which KYNA contributes to pathophysiology, remain largely elusive. The authors used patient-derived cellular modeling to test the hypothesis that KYNA can induce microglia-mediated synapse engulfment by reducing neuronal activity. METHODS Patient-derived induced pluripotent stem cells were used to generate 2D cultures of neurons and microglia-like cells, as well as forebrain organoids with innately developing microglia, to study how KYNA influences synaptic activity and microglial uptake of synaptic structures. To verify the experimental data in a clinical context, large-scale developmental postmortem brain tissue and genetic datasets were used to study coexpression networks for the KYNA-producing kynurenine aminotransferases (KATs) regarding enrichment for common schizophrenia genetic risk variants and functional annotations. RESULTS In these patient-derived experimental models, KYNA induced uptake of synaptic structures in microglia, and inhibition of the endogenous KYNA production led to a decrease in the internalization of synapses in microglia. The integrated large-scale transcriptomic and genetic datasets showed that KYNA-producing KATs enriched for genes governing synaptic activity and genetic risk variants for schizophrenia. CONCLUSIONS Together, these results link genetic risk variants for schizophrenia to elevated production of KYNA and excessive and activity-dependent internalization of synaptic material in microglia, while implicating pharmacological inhibition of KATs as a strategy to avoid synapse loss in schizophrenia.
Collapse
Affiliation(s)
- Funda Orhan
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| | - Susmita Malwade
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| | - Neda Khanlarkhani
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| | - Asimenia Gkogka
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| | - Angelika Langeder
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| | - Oscar Jungholm
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| | - Marja Koskuvi
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| | - Šárka Lehtonen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| | - Lilly Schwieler
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| | - Kent Jardemark
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| | - Jari Tiihonen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| | - Jari Koistinaho
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| | - Sophie Erhardt
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| | - Göran Engberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| | - Samudyata Samudyata
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Orhan, Malwade, Khanlarkhani, Gkogka, Jungholm, Koskuvi, Schwieler, Jardemark, Erhardt, Engberg, Samudyata, Sellgren); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (Langeder); Neuroscience Center, HiLIFE, University of Helsinki, Helsinki (Koskuvi, Lehtonen, Tiihonen, Koistinaho); A.I. Virtanen Institute for Molecular Sciences (Lehtonen) and Department of Forensic Psychiatry (Tiihonen), University of Eastern Finland, Kuopio; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Stockholm County Council, Stockholm (Tiihonen, Sellgren); Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania (Engberg)
| |
Collapse
|
6
|
Shen J, Wang Y, Liu Y, Lan J, Long S, Li Y, Chen D, Yu P, Zhao J, Wang Y, Wang S, Yang F. Behavioral Abnormalities, Cognitive Impairments, Synaptic Deficits, and Gene Replacement Therapy in a CRISPR Engineered Rat Model of 5p15.2 Deletion Associated With Cri du Chat Syndrome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415224. [PMID: 39965128 PMCID: PMC11984882 DOI: 10.1002/advs.202415224] [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: 11/19/2024] [Revised: 01/29/2025] [Indexed: 02/20/2025]
Abstract
The Cri du Chat Syndrome (CdCS), a devastating genetic disorder caused by a deletion on chromosome 5p, faces challenges in finding effective treatments and accurate animal models. Using CRISPR-Cas9, a novel CdCS rat model with a 2q22 deletion is developed, mirroring a common genetic alteration in CdCS patients. This model exhibits pronounced deficits in social behavior, cognition, and anxiety, accompanied by neuronal abnormalities and immune dysregulation in key brain regions such as the hippocampus and medial prefrontal cortex (mPFC). The immunostaining and RNA-seq analyses provide new insights into CdCS pathogenesis, revealing inflammatory and immune processes. Importantly, it is demonstrated that early gene replacement therapy with AAV-Ctnnd2 alleviates cognitive impairments in CdCS rats, highlighting the potential for early intervention. However, the effectiveness of this therapy is confined to the early developmental stages and does not fully restore all CdCS symptoms. The findings deepen the understanding of CdCS pathogenesis and suggest promising therapeutic directions.
Collapse
Affiliation(s)
- Jingjing Shen
- Institute of NeuroscienceSchool of Basic MedicineChongqing Medical UniversityChongqing400016China
| | - Yan Wang
- Institute of NeuroscienceSchool of Basic MedicineChongqing Medical UniversityChongqing400016China
| | - Yang Liu
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
| | - Junying Lan
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
- Advanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijing100070China
- Laboratory of Cognitive and Behavioral DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijing100069China
| | - Shuang Long
- Institute of NeuroscienceSchool of Basic MedicineChongqing Medical UniversityChongqing400016China
| | - Yingbo Li
- Institute of NeuroscienceSchool of Basic MedicineChongqing Medical UniversityChongqing400016China
| | - Di Chen
- Institute of NeuroscienceSchool of Basic MedicineChongqing Medical UniversityChongqing400016China
| | - Peng Yu
- Chinese Institutes for Medical ResearchCapital Medical UniversityBeijing100069China
| | - Jing Zhao
- Institute of NeuroscienceSchool of Basic MedicineChongqing Medical UniversityChongqing400016China
| | - Yongjun Wang
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
- Department of NeurologyBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
- Clinical Center for Precision Medicine in StrokeCapital Medical UniversityBeijing100070China
- Center of Excellence for Omics Research (CORe)Beijing Tiantan HospitalCapital Medical UniversityBeijing100070China
| | - Shali Wang
- Institute of NeuroscienceSchool of Basic MedicineChongqing Medical UniversityChongqing400016China
| | - Feng Yang
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
- Advanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijing100070China
- Laboratory of Cognitive and Behavioral DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijing100069China
| |
Collapse
|
7
|
Kalin NH. Psychotic Disorders and Schizophrenia: Treatment, Risk, Brain Alterations, and Mechanisms. Am J Psychiatry 2025; 182:313-315. [PMID: 40165557 DOI: 10.1176/appi.ajp.20250133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Affiliation(s)
- Ned H Kalin
- Department of Psychiatry, University of Wisconsin-Madison School of Medicine and Public Health
| |
Collapse
|
8
|
Hassani Nia F, Wittamer V. Zebrafish in neurodevelopmental disorders studies: Genetic models and pathological involvement of microglia. Dev Med Child Neurol 2025. [PMID: 40156170 DOI: 10.1111/dmcn.16317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 04/01/2025]
Abstract
Neurodevelopmental disorders (NDDs) are a group of brain disorders with a neonatal or early childhood onset and are lifelong. Various factors including genetics, and environmental and immune-related risk factors have been associated with NDDs. Given the complex nature of these disorders, multiple animal models have been used to investigate their aetiology and underlying cellular and molecular mechanisms. Recently, zebrafish have attracted great attention as an emerging model for studying NDDs. In addition to their easy maintenance, short developmental cycle, ex utero embryonic evolution, and optical clarity, zebrafish have successfully recapitulated phenotypes seen in human genetic disorders. This review explores the growing role of zebrafish in NDD research, by summarizing recently developed zebrafish genetic models for autism spectrum disorder, schizophrenia, and cerebral palsy. We then explore the potential of zebrafish as a model for studying NDDs linked to immune system dysfunction.
Collapse
Affiliation(s)
- Fatemeh Hassani Nia
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire Jacques E. Dumont, Brussels, Belgium
- ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Valerie Wittamer
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire Jacques E. Dumont, Brussels, Belgium
- ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
9
|
Hu W, Chen M, Lin Y, Zhang H, Sun L, Shao W, Ye Y, Cheng Y, Zhou S, Hu P, Wu X, Xu Y, Wang K. Neuronal CD47 induces behavioral alterations and ameliorates microglial synaptic pruning in wild-type and Alzheimer's mouse models. Cell Biosci 2025; 15:38. [PMID: 40140948 PMCID: PMC11948738 DOI: 10.1186/s13578-025-01378-x] [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: 01/15/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Microglia are brain-resident macrophages that play a crucial role in synapse pruning during the development and progression of various neuropsychiatric disorders, including autism spectrum disorder (ASD) and Alzheimer's disease (AD). Mechanistically, CD47 protein acts as a potent 'do not eat me' signal, protecting synapses from phagocytosis by microglia. However, the functional role of the upregulated neuronal CD47 signal under both physiological and pathological conditions remains unclear. RESULTS We utilized an adeno-associated virus gene expression system to induce neuron-specific overexpression of CD47 in wild-type and 5xFAD mice, assessing its effects on microglial synaptic phagocytosis and mouse behaviors. Our results indicate that neuronal CD47 induces ASD-like behaviors and synaptic pruning defects, while promoting behavioral disinhibition and improving memory in wild-type mice. Single-nucleus RNA sequencing was employed to profile gene expression patterns in subpopulations of neurons and microglia. Notably, neuronal CD47 enhances synaptic pathways in neurons and particularly shifts microglial subpopulations from a disease-associated state to a homeostatic state. Additionally, neuronal CD47 reduces excessive microglial synaptic phagocytosis induced by Aβ pathology in 5xFAD mice. CONCLUSION Our study provides evidence that neuronal CD47 overexpression results in synaptic pruning defects and is involved in the pathogenesis of ASD, while also playing a beneficial role in mitigating excessive synaptic loss in Alzheimer's disease.
Collapse
Affiliation(s)
- Wenjie Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Mengting Chen
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Yuxue Lin
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Hui Zhang
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Li Sun
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Wei Shao
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Yuping Ye
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Yujie Cheng
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Shanshan Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
| | - Panpan Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
| | - Xingqi Wu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
| | - Yin Xu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China.
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China.
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China.
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China.
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China.
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China.
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China.
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China.
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China.
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China.
- Anhui Institute of Translational Medicine, Hefei, 230032, China.
| |
Collapse
|
10
|
Huang Z, Ren Z, Wang S, Xiao L, Ling Y, Xie Y, Wang G, Zhou B. Urolithin A alleviates schizophrenic-like behaviors and cognitive impairment in rats through modulation of neuroinflammation, neurogenesis, and synaptic plasticity. Sci Rep 2025; 15:10477. [PMID: 40140679 PMCID: PMC11947095 DOI: 10.1038/s41598-025-93554-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Cognitive impairment in schizophrenia occurs in the early stages of the disease and is closely associated with prognosis. Alleviation of cognitive impairment in schizophrenia faces major challenges owing to the lack of preventive and therapeutic drugs that are novel and effective. Urolithin A (UA) is a gut microbial metabolite of ellagic acid that has demonstrated neuroprotective effects in multiple neurological disease models. Nonetheless, the neuromodulatory role of UA in schizophrenia is yet to be elucidated. Wistar rat pups were separated from their mothers for 24 h on postnatal days (PNDs) 9-10 to establish an early-life stress model. The pups were pretreated with UA at different administration times (2, 4, and 6 weeks) and doses (50, 100, and 150 mg/kg) from adolescence (PND29). Behavioral tests were performed after the end of the administration. Subsequently, hippocampal samples were collected for histopathological and molecular evaluations. Male offspring rats subjected to maternal separation exhibited increased sensitivity to prepulse inhibition and cognitive impairment, accompanied by severe neuroinflammation and impaired neurogenesis. However, UA attenuated maternal separation-induced prepulse inhibition deficits and cognitive impairments and restored hippocampal neurogenesis in a dose-dependent manner. Furthermore, UA pretreatment preserved dendritic spine density, synapses, and presynaptic vesicles. In addition, it exerted anti-inflammatory effects by inhibiting microglial activation and expression of the proinflammatory cytokines tumor necrosis factor-α, interleukin-6, and interleukin-1β. Potential mechanisms included upregulation of brain-derived neurotrophic factor protein expression and activation of the extracellular signal-regulated kinase signaling pathway. This study is the first preclinical evaluation of the effects of UA on cognitive impairment in schizophrenia. The findings suggest that changes in cognitive function linked to schizophrenia are driven by the interaction among neuroinflammation, neurogenesis, and synaptic plasticity and that UA has the potential to reverse these processes. These observations provide evidence for future clinical trials of UA as a dietary supplement for preventing schizophrenia.
Collapse
Affiliation(s)
- Zhengyuan Huang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China
- Department of Pharmacy, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China
| | - Zhongyu Ren
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China
| | - Sanwang Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China
| | - Ling Xiao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China
- Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yipeng Ling
- Department of Pharmacy, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China
| | - Yinping Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China
- Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China.
- Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Donghu Road 115#, Wuhan, 430071, China.
| | - Benhong Zhou
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China.
- Department of Pharmacy, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan, 430060, China.
| |
Collapse
|
11
|
Yang S, Wang J, Cao Y, Zhang Y, Sun Z, Wan P, Pi M, Xiong Q, Shu X, Wang X, Xia Y. Clec7a Signaling in Microglia Promotes Synapse Loss Associated with Tauopathy. Int J Mol Sci 2025; 26:2888. [PMID: 40243488 PMCID: PMC11988799 DOI: 10.3390/ijms26072888] [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: 02/21/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Alzheimer's disease (AD) pathogenesis involves progressive synaptic degeneration, a process potentially driven by maladaptive microglial pruning activity. While synaptic loss is a hallmark of AD, the molecular signals triggering pathological microglia-mediated synaptic engulfment remain elusive. Clec7a-a key marker of disease-associated microglia (DAM)-is known to activate spleen tyrosine kinase (SYK) signaling, enhancing Aβ phagocytosis and neuroprotective functions in 5×FAD models. However, its role in regulating synapse-microglia interactions under tauopathic conditions remains undefined. Our analysis revealed a progressive activation of the Clec7a-SYK signaling axis in the hippocampus of PS19 tauopathy mice, correlating with disease progression. Spatial mapping demonstrated a significant co-localization of Clec7a with hippocampal microglia, suggesting cell-autonomous signaling. The pharmacological inhibition of Clec7a achieved multimodal therapeutic effects by attenuating microglial hyperreactivity, suppressing neuroinflammatory cytokine release, and restoring physiological synaptic turnover. Mechanistically, we identified MD2 as a synaptic "eat-me" signal on tauopathy-related synapses, recruiting Clec7a+ microglia to drive aberrant synaptic elimination in PS19 mice. Strikingly, Clec7a blockade rescued hippocampal-dependent memory deficits in behavioral tests. These findings position Clec7a as a context-dependent therapeutic target, with inhibition strategies showing particular promise for tauopathy-related synaptic degeneration.
Collapse
Affiliation(s)
- Shubing Yang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan 430056, China
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Ji Wang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan 430056, China
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Yongkang Cao
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan 430056, China
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Yibo Zhang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan 430056, China
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Zhuoran Sun
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan 430056, China
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Pin Wan
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan 430056, China
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Mingshan Pi
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan 430056, China
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Qi Xiong
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan 430056, China
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Xiji Shu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan 430056, China
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Xiaochuan Wang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan 430056, China
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Yiyuan Xia
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan 430056, China
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan 430056, China
| |
Collapse
|
12
|
Byrne RAJ, Nimmo J, Torvell M, Carpanini SM, Daskoulidou N, Hughes TR, Noble LV, Veteleanu A, Watkins LM, Zelek WM, O'Donovan MC, Morgan BP. The schizophrenia-associated gene CSMD1 encodes a complement classical pathway inhibitor predominantly expressed by astrocytes and at synapses in mice and humans. Brain Behav Immun 2025; 127:287-302. [PMID: 40112933 DOI: 10.1016/j.bbi.2025.03.026] [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: 11/08/2024] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025] Open
Abstract
CUB and sushi multiple domains 1 (CSMD1) is predominantly expressed in brain and robustly associated with schizophrenia risk; however, understanding of which cells express CSMD1 in brain and how it impacts risk is lacking. CSMD1 encodes a large transmembrane protein including fifteen tandem short consensus repeats (SCRs), resembling complement C3 convertase regulators. CSMD1 complement regulatory activity has been reported and mapped to SCR17-21. We expressed two SCR domains of CSMD1, SCR17-21 and SCR23-26, and characterised their complement regulatory activity using a panel of functional assays testing convertase and terminal pathway inhibition. Both domains inhibited the classical pathway C3 convertase by acting as factor I cofactors; neither domain caused any inhibition in alternative or terminal pathway assays. Novel anti-CSMD1 monoclonal antibodies cross-reactive with human and mouse CSMD1 were generated that detected endogenous CSMD1 in human and rodent brain; immunostaining showed predominantly astrocyte and synaptic localisation of CSMD1, the latter confirmed using isolated synapses. Using iPSC-derived cells, astrocyte expression was confirmed and expression on cortical neurons demonstrated. We show that CSMD1 is a classical pathway-specific complement regulator expressed predominantly on astrocytes, neurons, and synapses in human and mouse brain. These findings will help reveal the mechanism by which CSMD1 impacts schizophrenia risk.
Collapse
Affiliation(s)
- Robert A J Byrne
- Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK.
| | - Jacqui Nimmo
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Megan Torvell
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Sarah M Carpanini
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Nikoleta Daskoulidou
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Timothy R Hughes
- Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Lucy V Noble
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Aurora Veteleanu
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Lewis M Watkins
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Wioleta M Zelek
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Michael C O'Donovan
- Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK
| | - Bryan Paul Morgan
- Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK.
| |
Collapse
|
13
|
Faustmann TJ, Corvace F, Faustmann PM, Ismail FS. Influence of antipsychotic drugs on microglia-mediated neuroinflammation in schizophrenia: perspectives in an astrocyte-microglia co-culture model. Front Psychiatry 2025; 16:1522128. [PMID: 40171306 PMCID: PMC11959008 DOI: 10.3389/fpsyt.2025.1522128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/12/2025] [Indexed: 04/03/2025] Open
Abstract
Schizophrenia is a severe mental disorder with a strong lifetime impact on patients' health and wellbeing. Usually, symptomatic treatment includes typical or atypical antipsychotics. Study findings show an involvement of low-grade inflammation (blood, brain parenchyma, and cerebrospinal fluid) in schizophrenia. Moreover, experimental and neuropathological evidence suggests that reactive microglia, which are the main resident immune cells of the central nervous system (CNS), have a negative impact on the differentiation and function of oligodendrocytes, glial progenitor cells, and astrocytes, which results in the disruption of neuronal networks and dysregulated synaptic transmission, contributing to the pathophysiology of schizophrenia. Here, the role of microglial cells related to neuroinflammation in schizophrenia was discussed to be essential. This review aims to summarize the evidence for the influence of antipsychotics on microglial inflammatory mechanisms in schizophrenia. Furthermore, we propose an established astrocyte-microglia co-culture model for testing regulatory mechanisms and examining the effects of antipsychotics on glia-mediated neuroinflammation. This could lead to a better understanding of how typical and atypical antipsychotics can be used to address positive and negative symptoms in schizophrenia and comorbidities like inflammatory diseases or the status of low-grade inflammation.
Collapse
Affiliation(s)
- Timo Jendrik Faustmann
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Franco Corvace
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Pedro M. Faustmann
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Fatme Seval Ismail
- Department of Neurology, Klinikum Vest, Academic Teaching Hospital of the Ruhr University Bochum, Recklinghausen, Germany
| |
Collapse
|
14
|
Cuní-López C, Stewart R, Okano S, Redlich GL, Appleby MW, White AR, Quek H. Exploring a patient-specific in vitro pipeline for stratification and drug response prediction of microglia-based therapeutics. Sci Rep 2025; 15:8296. [PMID: 40064964 PMCID: PMC11894103 DOI: 10.1038/s41598-025-92593-6] [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: 03/31/2024] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
The scarcity of effective biomarkers and therapeutic strategies for predicting disease onset and progression in neurodegenerative diseases such as Alzheimer's disease (AD) is a major challenge. Conventional drug discovery approaches have been unsuccessful in providing efficient interventions due to their 'one-size-fits-all' nature. As an alternative, personalised drug development holds promise to pre-select responders and identifying suitable indicators of drug efficacy. In this exploratory study, we have established a pipeline with the potential to guide patient stratification studies before clinical trials. This pipeline uses 2D and 3D in vitro models of monocyte-derived microglia-like cells (MDMi) from AD and mild cognitive impairment (MCI) patients, and matched healthy control (HC) individuals. By profiling cytokine responses in these models using multidimensional analyses, we have observed that the 3D model offers a more defined separation of profiles between individuals based on disease status. While this pilot study focuses on AD and MCI, future investigations incorporating other neurodegenerative disorders will be necessary to validate the pipeline's findings and demonstrate its broader applicability.
Collapse
Affiliation(s)
- Carla Cuní-López
- Brain and Mental Health, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, QLD, 4006, Australia.
- Faculty of Medicine, The University of Queensland, 20 Weightman St, Herston, QLD, 4006, Australia.
| | - Romal Stewart
- Brain and Mental Health, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, QLD, 4006, Australia
- UQ Centre for Clinical Research, the University of Queensland, Building 71/918 RBWH , Herston, Brisbane City, QLD, 4029, Australia
| | - Satomi Okano
- Statistics, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, QLD, 4006, Australia
| | - Garry L Redlich
- Implicit Bioscience, 523 Broadway E, Seattle, WA, 98102, USA
- Implicit Bioscience, 32 Logan Rd, Woolloongabba, QLD, 4102, Australia
| | - Mark W Appleby
- Implicit Bioscience, 523 Broadway E, Seattle, WA, 98102, USA
- Implicit Bioscience, 32 Logan Rd, Woolloongabba, QLD, 4102, Australia
| | - Anthony R White
- Brain and Mental Health, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, QLD, 4006, Australia.
- School of Biomedical Sciences, The University of Queensland, Chancellors Pl, St Lucia, QLD, 4072, Australia.
| | - Hazel Quek
- Brain and Mental Health, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, QLD, 4006, Australia.
- UQ Centre for Clinical Research, the University of Queensland, Building 71/918 RBWH , Herston, Brisbane City, QLD, 4029, Australia.
- School of Biomedical Sciences, The University of Queensland, Chancellors Pl, St Lucia, QLD, 4072, Australia.
- School of Biomedical Sciences, Queensland University of Technology, 2 George St, Brisbane City, QLD, 4000, Australia.
| |
Collapse
|
15
|
Zhao G, Lai B. SC-VAR: a computational tool for interpreting polygenic disease risks using single-cell epigenomic data. Brief Bioinform 2025; 26:bbaf123. [PMID: 40127183 PMCID: PMC11932087 DOI: 10.1093/bib/bbaf123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/11/2025] [Accepted: 03/01/2025] [Indexed: 03/26/2025] Open
Abstract
MOTIVATION One major challenge of interpreting variants from genome-wide association studies (GWAS) of complex traits or diseases is how to efficiently annotate noncoding variants. These variants influence gene expression by disrupting cis-regulatory elements (CREs), whose spatial and cell-type specificity are not adequately captured by conventional tools like multi-marker analysis of genomic annotation. Current methods either rely on linear proximity to genes or quantitative trait locus (QTL) data yet fail to integrate single-cell epigenomic information for a comprehensive annotation. RESULTS We present SC-VAR, a novel computational tool designed to enhance the interpretation of disease-associated risks from GWAS using single-cell epigenomic data. SC-VAR leverages single-cell epigenomic data to predict functional outcomes including risk genes, pathways, and cell types for both coding and noncoding disease-associated variants. We demonstrate that SC-VAR outperforms state-of-the-art methods by predicting more validated disease-related genes and pathways for multiple diseases. Additionally, SC-VAR identifies cell types that are susceptible to disease, along with their specific CREs and target genes linked to risk. By capturing a broad range of disease risks across human tissues at distinct developmental stages, SC-VAR could enhance our understanding of disease mechanisms in complex tissues across different life stages.
Collapse
Affiliation(s)
- Gefei Zhao
- Institute of Medical Technology, Peking University Health Science Center, 38 Xueyuan Rd, Hai Dian Qu, Beijing 100191, China
- Biomedical Engineering Department, Institute of Advanced Clinical Medicine, Peking University, 5 Yiheyuan Rd, Haidian District, Beijing 100191, China
| | - Binbin Lai
- Institute of Medical Technology, Peking University Health Science Center, 38 Xueyuan Rd, Hai Dian Qu, Beijing 100191, China
- Biomedical Engineering Department, Institute of Advanced Clinical Medicine, Peking University, 5 Yiheyuan Rd, Haidian District, Beijing 100191, China
- Department of Dermatology and Venerology, Peking University First Hospital, 8 Xishiku Ave, Xicheng Distric, Beijing 100191, China
- State Key Laboratory of Molecular Oncology, Peking University International Cancer Institute, 5 Yiheyuan Rd, Haidian District, Beijing 100191, China
| |
Collapse
|
16
|
Moran ER, Gabriele ML. Microglial Engulfment of Multisensory Terminals in the Midbrain Inferior Colliculus During an Early Critical Period. J Comp Neurol 2025; 533:e70033. [PMID: 40023818 PMCID: PMC11879250 DOI: 10.1002/cne.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/20/2025] [Accepted: 02/10/2025] [Indexed: 03/04/2025]
Abstract
The lateral cortex of the inferior colliculus (LCIC) receives multisensory input arrays that preferentially target its compartmental organization. Inputs of somatosensory origin end within modular zones, while auditory inputs terminate throughout an encompassing matrix. Such discrete mapping emerges during an early postnatal critical period (birth to postnatal day 12, P12) via a process of segregation. Similar to other primitive brain maps, it appears an initial excess of connections may be pruned through a refinement process. Microglial cells (MGCs) are involved in a variety of systems in the selective removal and degradation of unnecessary contacts. Aberrations in map plasticity during early critical periods have been associated with certain neurodevelopmental conditions, including autism spectrum disorders (ASD). Despite evidence linking multisensory integration deficits with cognitive/behavioral disturbances associated with ASD, mechanisms that govern multimodal network modifications remain poorly understood. Thus, the present study combines novel tract-tracing approaches in living brain preparations and immunocytochemistry in CX3CR1-GFP knock-in mice to determine: (1) if fractalkine signaling (CX3CL1-CX3CR1) influences MGC engulfment of auditory afferents, (2) whether individual MGCs phagocytose endings of multisensory origin (auditory and somatosensory), and (3) whether consumed product is degraded via the MGC's lysosomal pathway. We demonstrate active MGC pruning of auditory endings at peak LCIC stages for projection shaping (P4, P8) that significantly decreases coincident with its critical period closure (P12). While developmentally regulated, auditory engulfment appears fractalkine signaling-independent. We also provide evidence that individual LCIC microglia engulf both auditory and somatosensory terminals that co-localize with the lysosomal marker, CD68. These results suggest a prominent role for microglia in the remodeling of early multisensory midbrain maps.
Collapse
Affiliation(s)
- Emily R. Moran
- Dept. of Biology, James Madison Univ, Harrisonburg, VA, USA
| | | |
Collapse
|
17
|
Doyle AE, Bearden CE, Gur RE, Ledbetter DH, Martin CL, McCoy TH, Pasaniuc B, Perlis RH, Smoller JW, Davis LK. Advancing Mental Health Research Through Strategic Integration of Transdiagnostic Dimensions and Genomics. Biol Psychiatry 2025; 97:450-460. [PMID: 39424167 DOI: 10.1016/j.biopsych.2024.10.006] [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: 12/18/2023] [Revised: 09/11/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
Genome-wide studies are yielding a growing catalog of common and rare variants that confer risk for psychopathology. However, despite representing unprecedented progress, emerging data also indicate that the full promise of psychiatric genetics-including understanding pathophysiology and improving personalized care-will not be fully realized by targeting traditional dichotomous diagnostic categories. The current article provides reflections on themes that emerged from a 2021 National Institute of Mental Health-sponsored conference convened to address strategies for the evolving field of psychiatric genetics. As anticipated by the National Institute of Mental Health's Research Domain Criteria framework, multilevel investigations of dimensional and transdiagnostic phenotypes, particularly when integrated with biobanks and big data, will be critical to advancing knowledge. The path forward will also require more diverse representation in source studies. Additionally, progress will be catalyzed by a range of converging approaches, including capitalizing on computational methods, pursuing biological insights, working within a developmental framework, and engaging health care systems and patient communities.
Collapse
Affiliation(s)
- Alysa E Doyle
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| | - Carrie E Bearden
- Departments of Psychiatry and Biobehavioral Sciences & Psychology, University of California at Los Angeles, Los Angeles, California
| | - Raquel E Gur
- Departments of Psychiatry, Neurology and Radiology, Perelman School of Medicine, University of Pennsylvania, and the Lifespan Brain Institute of Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, Pennsylvania
| | - David H Ledbetter
- Departments of Pediatrics and Psychiatry, University of Florida College of Medicine, Jacksonville, Florida
| | - Christa L Martin
- Geisinger Autism & Developmental Medicine Institute, Lewisburg, Pennsylvania
| | - Thomas H McCoy
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bogdan Pasaniuc
- Departments of Computational Medicine, Pathology and Laboratory Medicine, and Human Genetics, University of California at Los Angeles, Los Angeles, California
| | - Roy H Perlis
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jordan W Smoller
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Lea K Davis
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
18
|
Akkouh IA, Osete JR, Szabo A, Andreassen OA, Djurovic S. Neurobiological Perturbations in Bipolar Disorder Compared With Schizophrenia: Evidence From Cell Cultures and Brain Organoids. Biol Psychiatry 2025:S0006-3223(25)00110-6. [PMID: 39983953 DOI: 10.1016/j.biopsych.2025.02.012] [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: 10/16/2024] [Revised: 01/06/2025] [Accepted: 02/13/2025] [Indexed: 02/23/2025]
Abstract
Bipolar disorder (BD) and schizophrenia (SCZ) are uniquely human disorders with a complex pathophysiology that involves adverse neuropathological events in brain development. High disease polygenicity and limited access to live human brain tissue make these disorders exceedingly challenging to study mechanistically. Cellular cultures and brain organoids generated from human-derived pluripotent stem cells preserve the genetic background of the donor cells and recapitulate some of the defining characteristics of human brain architecture and early spatiotemporal development. These model systems have already proven successful in deciphering some of the neuropathological perturbations in BD and SCZ, and methodological advancements, such as the functional integration of 2 or more region-specific organoids and organoid transplantation in animals, promise to deliver increasingly refined insights. Here, we review a selection of recent discoveries achieved by stem cell-based models, with a particular focus on patterns of cellular and molecular convergence and divergence between BD and SCZ. First, we provide a brief overview of the evidence from glial and neuronal cell cultures and brain organoids, centering our discussion on several key functional domains, including neuroinflammation, neuronal excitability, and mitochondrial function. Then, we review recent findings demonstrating the power of integrating stem cell-based systems with gene editing technologies to elucidate the functional consequences of risk variants identified through genetic association studies. We end with a discussion of current challenges and some promising avenues for future research.
Collapse
Affiliation(s)
- Ibrahim A Akkouh
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Jordi Requena Osete
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Attila Szabo
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
19
|
Zhang L, Zhang J, Wang N, Liu C, Wang S, Dong X, Yang L, Bao X, Nie X, Li J. Bioinformatics-based identification of CTSS, DOK2, and ENTPD1 as potential blood biomarkers of schizophrenia. BMC Psychiatry 2025; 25:157. [PMID: 39972407 PMCID: PMC11841330 DOI: 10.1186/s12888-025-06512-0] [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: 01/22/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Although schizophrenia is a severe mental disorder that significantly impacts patients and society, there are currently no reliable blood-based biomarkers to assist in its diagnosis. The diagnosis primarily relies on clinical assessment and patient history, a method that is inherently subjective and prone to errors, potentially leading to diagnostic delays. In this study, we aim to utilize bioinformatics approaches to explore potential blood-based biomarkers for the diagnosis of schizophrenia. By employing advanced bioinformatics techniques, we hope to identify key genes and construct an effective diagnostic model, providing the clinic with a more objective and accurate diagnostic tool. METHODS In this research, we employed bioinformatics techniques to identify potential blood-based biomarkers for the diagnosis of schizophrenia. Initially, we selected schizophrenia-associated differentially expressed genes (DEGs) from the Gene Expression Omnibus (GEO) database through the datasets GSE27383, GSE38484, and GSE38481. Subsequently, we performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses on these DEGs to elucidate their biological functions and related pathways. Furthermore, we constructed a protein-protein interaction (PPI) network of the differentially expressed genes to identify key genes and matched them with their target microRNAs (miRNAs). In addition, we assessed the diagnostic potential of these key genes through immune infiltration analysis. The aim of this study is to reveal the roles of these hub genes in the pathogenesis of schizophrenia. RESULTS Through bioinformatics analysis, we have identified three potential hub genes associated with the pathogenesis of schizophrenia: CTSS, DOK2, and ENTPD1. These genes are significantly correlated with the development of schizophrenia and may serve as promising diagnostic biomarkers for the condition. CONCLUSION In this study, we have identified three pivotal genes-CTSS, DOK2, and ENTPD1-that are intimately associated with the pathogenesis of schizophrenia. The discovery of these genes not only enhances the precision of diagnostic efforts for schizophrenia but also provides a robust scientific foundation for the development of innovative treatment approaches for schizophrenia and related disorders. The identification of these biomarkers offers a tangible basis for early, accurate diagnosis, treatment, prognostic assessment, and rehabilitation evaluation in schizophrenia, potentially improving patients' quality of life and supporting the development of personalized therapeutics and antipsychotic medications.
Collapse
Affiliation(s)
- Lei Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Jiale Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Na Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Chenwei Liu
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Shuting Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Xiaotao Dong
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Lu Yang
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Xiaohong Bao
- Department of Cardiothoracic Surgery, School of Medicine, Taizhou Central Hospital, Taizhou University, Taizhou, 318000, China
| | - Xiaobo Nie
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Jicheng Li
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
20
|
Nelson N, Miller V, Broadie K. Neuron-to-glia and glia-to-glia signaling directs critical period experience-dependent synapse pruning. Front Cell Dev Biol 2025; 13:1540052. [PMID: 40040788 PMCID: PMC11876149 DOI: 10.3389/fcell.2025.1540052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/31/2025] [Indexed: 03/06/2025] Open
Abstract
Experience-dependent glial synapse pruning plays a pivotal role in sculpting brain circuit connectivity during early-life critical periods of development. Recent advances suggest a layered cascade of intercellular communication between neurons and glial phagocytes orchestrates this precise, targeted synapse elimination. We focus here on studies from the powerful Drosophila forward genetic model, with reference to complementary findings from mouse work. We present both neuron-to-glia and glia-to-glia intercellular signaling pathways directing experience-dependent glial synapse pruning. We discuss a putative hierarchy of secreted long-distance cues and cell surface short-distance cues that act to sequentially orchestrate glia activation, infiltration, target recognition, engulfment, and then phagocytosis for synapse pruning. Ligand-receptor partners mediating these stages in different contexts are discussed from recent Drosophila and mouse studies. Signaling cues include phospholipids, small neurotransmitters, insulin-like peptides, and proteins. Conserved receptors for these ligands are discussed, together with mechanisms where the receptor identity remains unknown. Potential mechanisms are proposed for the tight temporal-restriction of heightened experience-dependent glial synapse elimination during early-life critical periods, as well as potential means to re-open such plasticity at maturity.
Collapse
Affiliation(s)
- Nichalas Nelson
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, United States
| | - Vanessa Miller
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, United States
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, United States
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University and Medical Center, Nashville, TN, United States
- Kennedy Center for Research on Human Development, Vanderbilt University and Medical Center, Nashville, TN, United States
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN, United States
| |
Collapse
|
21
|
Yassin LK, Nakhal MM, Alderei A, Almehairbi A, Mydeen AB, Akour A, Hamad MIK. Exploring the microbiota-gut-brain axis: impact on brain structure and function. Front Neuroanat 2025; 19:1504065. [PMID: 40012737 PMCID: PMC11860919 DOI: 10.3389/fnana.2025.1504065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/30/2025] [Indexed: 02/28/2025] Open
Abstract
The microbiota-gut-brain axis (MGBA) plays a significant role in the maintenance of brain structure and function. The MGBA serves as a conduit between the CNS and the ENS, facilitating communication between the emotional and cognitive centers of the brain via diverse pathways. In the initial stages of this review, we will examine the way how MGBA affects neurogenesis, neuronal dendritic morphology, axonal myelination, microglia structure, brain blood barrier (BBB) structure and permeability, and synaptic structure. Furthermore, we will review the potential mechanistic pathways of neuroplasticity through MGBA influence. The short-chain fatty acids (SCFAs) play a pivotal role in the MGBA, where they can modify the BBB. We will therefore discuss how SCFAs can influence microglia, neuronal, and astrocyte function, as well as their role in brain disorders such as Alzheimer's disease (AD), and Parkinson's disease (PD). Subsequently, we will examine the technical strategies employed to study MGBA interactions, including using germ-free (GF) animals, probiotics, fecal microbiota transplantation (FMT), and antibiotics-induced dysbiosis. Finally, we will examine how particular bacterial strains can affect brain structure and function. By gaining a deeper understanding of the MGBA, it may be possible to facilitate research into microbial-based pharmacological interventions and therapeutic strategies for neurological diseases.
Collapse
Affiliation(s)
- Lidya K. Yassin
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammed M. Nakhal
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Alreem Alderei
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Afra Almehairbi
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ayishal B. Mydeen
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Amal Akour
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
22
|
Knight SR, Abbasova L, Zeighami Y, Hansen JY, Martins D, Zelaya F, Dipasquale O, Liu T, Shin D, Bossong M, Azis M, Antoniades M, Howes OD, Bonoldi I, Egerton A, Allen P, O'Daly O, McGuire P, Modinos G. Transcriptional and Neurochemical Signatures of Cerebral Blood Flow Alterations in Individuals With Schizophrenia or at Clinical High Risk for Psychosis. Biol Psychiatry 2025:S0006-3223(25)00076-9. [PMID: 39923816 DOI: 10.1016/j.biopsych.2025.01.028] [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: 07/19/2024] [Revised: 01/24/2025] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND The brain integrates multiple scales of description, from the level of cells and molecules to large-scale networks and behavior. Understanding relationships across these scales may be fundamental to advancing understanding of brain function in health and disease. Recent neuroimaging research has shown that functional brain alterations that are associated with schizophrenia spectrum disorders (SSDs) are already present in young adults at clinical high risk for psychosis (CHR-P), but the cellular and molecular determinants of these alterations remain unclear. METHODS Here, we used regional cerebral blood flow (rCBF) data from 425 individuals (122 with an SSD compared with 116 healthy control participants [HCs] and 129 individuals at CHR-P compared with 58 HCs) and applied a novel pipeline to integrate brainwide rCBF case-control maps with publicly available transcriptomic data (17,205 gene maps) and neurotransmitter atlases (19 maps) from 1074 healthy volunteers. RESULTS We identified significant correlations between astrocyte, oligodendrocyte, oligodendrocyte precursor cell, and vascular leptomeningeal cell gene modules for both SSD and CHR-P rCBF phenotypes. Additionally, endothelial cell genes were correlated in SSD, and microglia in CHR-P. Receptor distribution significantly predicted case-control rCBF differences, with dominance analysis highlighting dopamine (D1, D2, dopamine transporter), acetylcholine (VAChT, M1), gamma-aminobutyric acid A (GABAA), and glutamate (NMDA) receptors as key predictors for SSD (R2adjusted = 0.58, false discovery rate [FDR]-corrected p < .05) and CHR-P (R2adjusted = 0.6, pFDR < .05) rCBF phenotypes. These associations were primarily localized in subcortical regions and implicate cell types involved in stress response and inflammation, alongside specific neuroreceptor systems, in shared and distinct rCBF phenotypes in psychosis. CONCLUSIONS Our findings underscore the value of integrating multiscale data as a promising hypothesis-generating approach toward decoding biological pathways involved in neuroimaging-based psychosis phenotypes, potentially guiding novel interventions.
Collapse
Affiliation(s)
- Samuel R Knight
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
| | - Leyla Abbasova
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Yashar Zeighami
- Douglas Research Centre, Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Justine Y Hansen
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Fernando Zelaya
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Ottavia Dipasquale
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Olea Medical, La Ciotat, France
| | - Thomas Liu
- Centre for Functional MRI, University of California San Diego, San Diego, California
| | - David Shin
- Global MR Applications and Workflow, GE Healthcare, Menlo Park, California
| | - Matthijs Bossong
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Department of Psychiatry, Brain Center Rudoph Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Matilda Azis
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Mathilde Antoniades
- Center for AI and Data Science for Integrated Diagnostics and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Oliver D Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Ilaria Bonoldi
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Paul Allen
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Owen O'Daly
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Philip McGuire
- Department of Psychiatry, Oxford University, Oxford, United Kingdom
| | - Gemma Modinos
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| |
Collapse
|
23
|
Hemati H, Blanton MB, True HE, Koura J, Khadka R, Grant KA, Messaoudi I. Phenotypic and Functional Alterations in Peripheral Blood Mononuclear Cell-Derived Microglia in a Primate Model of Chronic Alcohol Consumption. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636708. [PMID: 39975066 PMCID: PMC11839131 DOI: 10.1101/2025.02.05.636708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Alcohol-induced dysregulation of microglial activity is associated with neuroinflammation, cognitive decline, heightened risk for neurodegenerative diseases, alcohol dependence, and escalation of alcohol drinking. Given the challenge of longitudinally sampling primary microglia, we optimized an in vitro method to differentiate peripheral blood mononuclear cells (PBMC) from non-human primates (NHP) into microglia-like cells (induced-microglia; iMGL). The iMGLs displayed transcriptional profiles distinct from those of monocyte progenitors and closely resembling those of primary microglia. Notably, morphological features showed that differentiated iMGLs derived from NHPs with chronic alcohol consumption (CAC) possessed a more mature-like microglial morphology. Additionally, dysregulation in key inflammatory and regulatory markers alongside increased baseline phagocytic activity was observed in CAC-derived IMGLs in the resting state. Phenotypic and functional assessments following LPS stimulation indicated the presence of an immune-tolerant phenotype and enrichment of a CD86+ hyper-inflammatory subpopulation in iMGLs derived from ethanol-exposed animals. Collectively, these findings demonstrate that in vitro differentiation of PBMC offers a minimally invasive approach to studying the impact of CAC on microglial function revealing that CAC reshapes both functional and transcriptional profiles of microglia.
Collapse
Affiliation(s)
- Hami Hemati
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Madison B Blanton
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Heather E True
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Jude Koura
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Rupak Khadka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, OR, United States
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, OR, United States
| | - Ilhem Messaoudi
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
24
|
Blasco MB, Nisha Aji K, Ramos-Jiménez C, Leppert IR, Tardif CL, Cohen J, Rusjan PM, Mizrahi R. Synaptic Density in Early Stages of Psychosis and Clinical High Risk. JAMA Psychiatry 2025; 82:171-180. [PMID: 39535765 PMCID: PMC11561726 DOI: 10.1001/jamapsychiatry.2024.3608] [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: 05/03/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024]
Abstract
Importance Synaptic dysfunction is involved in schizophrenia pathophysiology. However, whether in vivo synaptic density is reduced in early stages of psychosis, including its high-risk states, remains unclear. Objective To investigate whether synaptic density (synaptic vesicle glycoprotein 2A [SV2A] binding potential) is reduced in first-episode psychosis (FEP) and in clinical high risk (CHR) and investigate the effect of cannabis use on synaptic density and examine its relationship with psychotic symptoms and gray matter microstructure across groups. Design, Setting, and Participants This cross-sectional study was performed in a tertiary care psychiatric hospital from July 2021 to October 2023. Participants were patients with antipsychotic-free or minimally exposed FEP or CHR and healthy controls with a clean urine drug screen (except cannabis). Main Outcomes and Measures Synaptic density was quantified with dynamic 90-minute [18F]SynVesT-1 positron emission tomography (PET) scans across prioritized brain regions of interest (ROIs) delineated in individual magnetic resonance images (MRIs). Cannabis use was confirmed with urine drug screens. Gray matter microstructure was assessed using diffusion-weighted MRI to estimate neurite density. Results A total of 49 participants were included, including 16 patients with FEP (mean [SD] age, 26.1 [4.6] years; 9 males and 7 females), 17 patients at CHR (mean [SD] age, 21.2 [3.5] years; 8 males and 9 females), and 16 healthy controls (mean [SD] age, 23.4 [3.6] years; 7 males and 9 females). Synaptic density was significantly different between groups (F2,273 = 4.02, P = .02, Cohen F = 0.17; ROI: F5,273 = 360.18, P < .01, Cohen F = 2.55) with a group × ROI interaction (F10,273 = 2.67, P < .01, Cohen F = 0.32). Synaptic density was lower in cannabis users (F1,272 = 5.31, P = .02, Cohen F = 0.14). Lower synaptic density across groups was associated with more negative symptoms (Positive and Negative Syndrome Scale negative scores: F1,81 = 4.31, P = .04, Cohen F = 0.23; Scale of Psychosis-Risk Symptoms negative scores: F1,90 = 4.12, P = .04, Cohen F = 0.21). SV2A binding potential was significantly associated with neurite density index (F1,138 = 6.76, P = .01, Cohen F = 0.22). Conclusions and Relevance This study found that synaptic density reductions were present during the early stages of psychosis and its risk states and associated with negative symptoms. The implications of SV2A for negative symptoms in psychosis and CHR warrant further investigation. Future studies should investigate the impact of cannabis use on synaptic density in CHR longitudinally.
Collapse
Affiliation(s)
- M. Belen Blasco
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
| | - Kankana Nisha Aji
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
| | - Christian Ramos-Jiménez
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
| | - Ilana Ruth Leppert
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Christine Lucas Tardif
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| | - Johan Cohen
- Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
| | - Pablo M. Rusjan
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Romina Mizrahi
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
25
|
Sun H, Wang H, Wu C, Liu G, He M, Zhang H, Hou F, Liao H. Enhancing Neuron Activity Promotes Functional Recovery by Inhibiting Microglia-Mediated Synapse Elimination After Stroke. Stroke 2025; 56:505-516. [PMID: 39772780 DOI: 10.1161/strokeaha.124.049265] [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: 04/25/2024] [Revised: 11/05/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Activating glutamatergic neurons in the ipsilesional motor cortex can promote functional recovery after stroke. However, the underlying molecular mechanisms remain unclear. Clarifying key molecular mechanisms involved in recovery could help understand the development of neuromodulation strategies after stroke. METHODS Adeno-associated virus 2/9-CamKIIa-hM3Dq-mCherry was injected into ipsilesional motor cortex by stereotaxic in the photothrombotic stroke model. Starting from the third day after the stroke, male mice were injected intraperitoneally with clozapine-N-oxide every day to activate excitatory neurons. C1q-blocking antibody and annexin V were used to inhibit C1q and exposed phosphatidylserine (EPS), respectively. The cylinder test and grid-walking test were performed to evaluate functional recovery. The potential molecular mechanisms of excitatory neuronal activation on microglia-mediated synaptic pruning after stroke by immunofluorescence, real-time polymerase chain reaction, Western blotting, and RNA sequencing. RESULTS Activating excitatory neurons significantly promoted functional recovery and inhibited microglia-mediated synaptic pruning after stroke. Furthermore, it decreased EPS and C1q levels in synapses. On the contrary, inhibiting excitatory neurons aggravated functional defects, promoted microglia-mediated synaptic pruning, and increased EPS and C1q levels in synapses. Selective blocking of EPS repressed C1q tagging of synapses and microglia-mediated synaptic pruning and improved functional recovery. Meanwhile, blocking EPS markedly rescued synaptic density, and motor function deteriorated by chemogenetic inhibition. In addition, C1q-blocking antibody prevented phosphatidylserine engulfment by microglia. CONCLUSIONS Together, these data provide mechanistic insight into microglia-mediated synapse pruning after neuronal activation after stroke and identify the role of C1q binding to EPS in stroke treatment during the repair phase.
Collapse
Affiliation(s)
- Hao Sun
- New Drug Screening Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (H.S., H.W., C.W., G.L., M.H., H.Z., F.H., H.L.)
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, China (H.S., H.L.)
| | - Heng Wang
- New Drug Screening Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (H.S., H.W., C.W., G.L., M.H., H.Z., F.H., H.L.)
| | - Chaoran Wu
- New Drug Screening Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (H.S., H.W., C.W., G.L., M.H., H.Z., F.H., H.L.)
| | - Gang Liu
- New Drug Screening Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (H.S., H.W., C.W., G.L., M.H., H.Z., F.H., H.L.)
| | - Meijun He
- New Drug Screening Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (H.S., H.W., C.W., G.L., M.H., H.Z., F.H., H.L.)
| | - Hao Zhang
- New Drug Screening Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (H.S., H.W., C.W., G.L., M.H., H.Z., F.H., H.L.)
| | - Fengsheng Hou
- New Drug Screening Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (H.S., H.W., C.W., G.L., M.H., H.Z., F.H., H.L.)
| | - Hong Liao
- New Drug Screening Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (H.S., H.W., C.W., G.L., M.H., H.Z., F.H., H.L.)
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, China (H.S., H.L.)
| |
Collapse
|
26
|
Schepanski S, Ngoumou GB, Buss C, Seifert G. Assessing in-vitro models for microglial development and fetal programming: a critical review. Front Immunol 2025; 16:1538920. [PMID: 39944696 PMCID: PMC11814449 DOI: 10.3389/fimmu.2025.1538920] [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: 12/03/2024] [Accepted: 01/08/2025] [Indexed: 05/09/2025] Open
Abstract
This review evaluates in-vitro models for studying how maternal influences during pregnancy impact the development of offspring microglia, the immune cells of the central nervous system. The models examined include primary microglia cultures, microglia cell lines, iPSC-derived microglia, PBMC-induced microglia-like cells, 3D brain organoids derived from iPSCs, and Hofbauer cells. Each model is assessed for its ability to replicate the in-vivo environment of the developing brain, with a focus on their strengths, limitations, and practical challenges. Key factors such as scalability, genetic and epigenetic fidelity, and physiological relevance are highlighted. Microglia cell lines are highly scalable but lack genetic and epigenetic fidelity. iPSC-derived microglia provide moderate physiological relevance and patient-specific genetic insights but face operational and epigenetic challenges inherent to reprogramming. 3D brain organoids, derived from iPSCs, offer an advanced platform for studying complex neurodevelopmental processes but require extensive resources and technical expertise. Hofbauer cells, which are fetal macrophages located in the placenta and share a common developmental origin with microglia, are uniquely exposed to prenatal maternal factors and, depending on fetal barrier maturation, exhibit variable epigenetic fidelity. This makes them particularly useful for exploring the impact of maternal influences on fetal programming of microglial development. The review concludes that no single model comprehensively captures all aspects of maternal influences on microglial development, but it offers guidance on selecting the most appropriate model based on specific research objectives and experimental constraints.
Collapse
Affiliation(s)
- Steven Schepanski
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité Competence Center for Traditional and Integrative Medicine (CCCTIM), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pediatrics, Division of Oncology and Hematology, Berlin, Germany
| | - Gonza B. Ngoumou
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité Competence Center for Traditional and Integrative Medicine (CCCTIM), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pediatrics, Division of Oncology and Hematology, Berlin, Germany
| | - Claudia Buss
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Medical Psychology, Berlin, Germany
- University of California, Irvine, Development, Health and Disease Research Program, Irvine, CA, United States
- German Center for Child and Adolescent Health (DZKJ), Partner Site Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Mental Health (DZPG), Partner Site Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Georg Seifert
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité Competence Center for Traditional and Integrative Medicine (CCCTIM), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pediatrics, Division of Oncology and Hematology, Berlin, Germany
| |
Collapse
|
27
|
Zheng T, Long K, Wang S, Rui M. Glial-derived TNF/Eiger signaling promotes somatosensory neurite sculpting. Cell Mol Life Sci 2025; 82:47. [PMID: 39833565 PMCID: PMC11747020 DOI: 10.1007/s00018-024-05560-1] [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: 08/08/2024] [Revised: 11/11/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025]
Abstract
The selective elimination of inappropriate projections is essential for sculpting neural circuits during development. The class IV dendritic arborization (C4da) sensory neurons of Drosophila remodel the dendritic branches during metamorphosis. Glial cells in the central nervous system (CNS), are required for programmed axonal pruning of mushroom body (MB) γ neurons during metamorphosis in Drosophila. However, it is entirely unknown whether the glial cells are involved in controlling the neurite pruning of C4da sensory neurons. Here, we show that glial deletion of Eiger (Egr), orthologous to mammalian tumor necrosis factor TNF superfamily ligand, results in dendrite remodeling deficiency of Drosophila C4da sensory neurons. Moreover, the attenuation of neuronal Wengen (Wgn) and Grindelwald (Grnd), the receptors for TNF ligands, is also examined for defects in dendrite remodeling. We further discover that Wgn and Grnd facilitate dendrite elimination through the JNK Signaling. Overall, our findings demonstrate that glial-derived Egr signal links to the neuronal receptor Wgn/Grnd, activating the JNK signaling pathway and promoting developmental neuronal remodeling. Remarkably, our findings reveal a crucial role of peripheral glia in dendritic pruning of C4da neurons.
Collapse
Affiliation(s)
- Ting Zheng
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Keyao Long
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Su Wang
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Menglong Rui
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| |
Collapse
|
28
|
Göteson A, Holmén-Larsson J, Celik H, Pelanis A, Sellgren CM, Sparding T, Pålsson E, Zetterberg H, Blennow K, Jonsson L, Gobom J, Landén M. Mapping the Cerebrospinal Fluid Proteome in Bipolar Disorder. Biol Psychiatry 2025:S0006-3223(25)00029-0. [PMID: 39827936 DOI: 10.1016/j.biopsych.2025.01.007] [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: 07/18/2024] [Revised: 01/04/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Bipolar disorder (BD) is a severe psychiatric condition with unclear etiology and no established biomarkers. Here, we aimed to characterize the cerebrospinal fluid (CSF) proteome in euthymic individuals with BD to identify potential protein biomarkers. METHODS We used nano-flow liquid chromatography coupled to high-resolution mass spectrometry to quantify over 2000 CSF proteins in 374 individuals from two independent clinical cohorts (n = 164 cases + 89 controls and 66 cases + 55 controls, respectively). A subset of the cases was followed longitudinally and reexamined after a median of 6.5 years. RESULTS Differential abundance analysis revealed 41 proteins with robust case-control association in both cohorts. These included lower levels of synaptic proteins (e.g., APP, CLSTN1, NPTX2, NRXN1) and axon guidance and cell adhesion molecules (e.g., NEO1, NCAM1, SEMA7A) and higher levels of blood-brain barrier integrity proteins (e.g., VTN, SERPIN3) and complement components (e.g., C1RL, C3, C5). The findings were consistently driven by the BD type 1 subtype. Comparing BD type 1 participants with control participants increased discoverability, revealing 86 replicated associations despite a loss of statistical power. Moreover, longitudinal analyses of coexpression modules revealed dynamic changes in the CSF proteome composition that correlated with clinical outcomes, including disease severity, future manic episodes, and symptom improvement. Finally, we conducted association analyses of CSF proteins with genetic risk loci for BD and schizophrenia. CONCLUSIONS This study represents the first large-scale untargeted profiling of the CSF proteome in BD, unveiling potential biomarkers and providing in vivo support for altered synaptic and brain connectivity processes, impaired neurovascular integrity, and complement activation in the pathology of BD.
Collapse
Affiliation(s)
- Andreas Göteson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| | - Jessica Holmén-Larsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Hatice Celik
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Aurimantas Pelanis
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Region Stockholm, Sweden
| | - Timea Sparding
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Erik Pålsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, Dementia Research Centre, University College London Institute of Neurology, London, United Kingdom; UK Dementia Research Institute, University College London, London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute of Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of University of Science and Technology, Hefei, China
| | - Lina Jonsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Mikael Landén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
29
|
Lendemeijer B, de Vrij FMS. In vitro models for human neuroglia. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:213-227. [PMID: 40122626 DOI: 10.1016/b978-0-443-19104-6.00015-2] [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: 03/25/2025]
Abstract
Neuroglia are a heterogenous population of cells in the nervous system. In the central nervous system, this group is classified into astrocytes, oligodendrocytes, and microglia. Neuroglia in the peripheral nervous system are divided into Schwann cells and enteric glia. These groups of cells display considerable differences in their developmental origin, morphology, function, and regional abundance. Compared to animal models, human neuroglia differ in their transcriptomic profile, morphology, and function. Investigating the physiology of healthy or diseased human neuroglia in vivo is challenging due to the inaccessibility of the tissue. Therefore, researchers have developed numerous in vitro models attempting to replicate the natural tissue environment. Earlier models made use of postmortem, postsurgical, or fetal tissue to establish human neuroglial cells in vitro, either as a pure population of the desired cell type or as organotypic slice cultures. Advancements in human stem cell differentiation techniques have greatly enhanced the possibilities for creating in vitro models of human neuroglia. This chapter provides an overview of the current models used to study the functioning and development of human neuroglia in vitro, both in health and disease.
Collapse
Affiliation(s)
- Bas Lendemeijer
- Department of Psychiatry, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Psychiatry, Columbia University Medical Center, New York, NY, United States
| | - Femke M S de Vrij
- Department of Psychiatry, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
30
|
Llaves-López A, Micoli E, Belmonte-Mateos C, Aguilar G, Alba C, Marsal A, Pulido-Salgado M, Rabaneda-Lombarte N, Solà C, Serratosa J, Vidal-Taboada JM, Saura J. Human Microglia-Like Cells Differentiated from Monocytes with GM-CSF and IL-34 Show Phagocytosis of α-Synuclein Aggregates and C/EBPβ-Dependent Proinflammatory Activation. Mol Neurobiol 2025; 62:756-772. [PMID: 38900366 PMCID: PMC11711251 DOI: 10.1007/s12035-024-04289-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 06/02/2024] [Indexed: 06/21/2024]
Abstract
Microglia, the main resident immune cells in the central nervous system, are implicated in the pathogenesis of various neurological disorders. Much of our knowledge on microglial biology was obtained using rodent microglial cultures. To understand the role of microglia in human disease, reliable in vitro models of human microglia are necessary. Monocyte-derived microglia-like cells (MDMi) are a promising approach. This study aimed to characterize MDMi cells generated from adult human monocytes using granulocyte-macrophage colony-stimulating factor and interleukin-34. To this end, 49 independent cultures of MDMI were prepared, and various methodological and functional studies were performed. We show that with this protocol, adult human monocytes develop into microglia-like cells, a coating is unnecessary, and high cell density seeding is preferable. When compared to monocytes, MDMi upregulate the expression of many, but not all, microglial markers, indicating that, although these cells display a microglia-like phenotype, they cannot be considered bona fide human microglia. At the functional level, MDMi phagocytose α-synuclein aggregates and responds to lipopolysaccharide (LPS) by nuclear translocation of the transcription factor nuclear factor-kappaB (NFkappaB) and the upregulation of proinflammatory genes. Finally, a long-lasting silencing of the transcription factor CCAAT/enhancer protein β (C/EBPβ) was achieved by small interfering RNA, resulting in the subsequent downregulation of proinflammatory genes. This supports the hypothesis that C/EBPβ plays a key role in proinflammatory gene program activation in human microglia. Altogether, this study sheds new light on the properties of MDMi cells and supports these cells as a promising in vitro model for studying adult human microglia-like cells.
Collapse
Affiliation(s)
- Andrea Llaves-López
- Biochemistry and Molecular Biology Unit, Department of Biomedical Sciences, School of Medicine, University of Barcelona, IDIBAPS, Casanova 143, 08036, Barcelona, Catalonia, Spain
| | - Elia Micoli
- Biochemistry and Molecular Biology Unit, Department of Biomedical Sciences, School of Medicine, University of Barcelona, IDIBAPS, Casanova 143, 08036, Barcelona, Catalonia, Spain
| | - Carla Belmonte-Mateos
- Biochemistry and Molecular Biology Unit, Department of Biomedical Sciences, School of Medicine, University of Barcelona, IDIBAPS, Casanova 143, 08036, Barcelona, Catalonia, Spain
| | - Gerard Aguilar
- Biochemistry and Molecular Biology Unit, Department of Biomedical Sciences, School of Medicine, University of Barcelona, IDIBAPS, Casanova 143, 08036, Barcelona, Catalonia, Spain
| | - Clara Alba
- Biochemistry and Molecular Biology Unit, Department of Biomedical Sciences, School of Medicine, University of Barcelona, IDIBAPS, Casanova 143, 08036, Barcelona, Catalonia, Spain
| | - Anais Marsal
- Biochemistry and Molecular Biology Unit, Department of Biomedical Sciences, School of Medicine, University of Barcelona, IDIBAPS, Casanova 143, 08036, Barcelona, Catalonia, Spain
| | - Marta Pulido-Salgado
- Biochemistry and Molecular Biology Unit, Department of Biomedical Sciences, School of Medicine, University of Barcelona, IDIBAPS, Casanova 143, 08036, Barcelona, Catalonia, Spain
| | - Neus Rabaneda-Lombarte
- Department of Neuroscience and Experimental Therapeutics, IIBB, CSIC, IDIBAPS, Barcelona, Catalonia, Spain
| | - Carme Solà
- Department of Neuroscience and Experimental Therapeutics, IIBB, CSIC, IDIBAPS, Barcelona, Catalonia, Spain
| | - Joan Serratosa
- Department of Neuroscience and Experimental Therapeutics, IIBB, CSIC, IDIBAPS, Barcelona, Catalonia, Spain
| | - Jose M Vidal-Taboada
- Peripheral Nervous System, Neuroscience Department, VHIR, Vall d'Hebron Research Institute, Barcelona, Catalonia, Spain
| | - Josep Saura
- Biochemistry and Molecular Biology Unit, Department of Biomedical Sciences, School of Medicine, University of Barcelona, IDIBAPS, Casanova 143, 08036, Barcelona, Catalonia, Spain.
- Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia, Spain.
| |
Collapse
|
31
|
Campos-Sánchez JC, Meseguer J, Guardiola FA. Fish microglia: Beyond the resident macrophages of the central nervous system - A review of their morphofunctional characteristics. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 162:105274. [PMID: 39341477 DOI: 10.1016/j.dci.2024.105274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
From classical to modern literature on microglia, the importance of the potential and variability of these immune cells in vertebrates has been pointed out. Recent aspects such as relationships and interactions between microglia and neurons in both normal and injured neural tissues, as well as their nexus with other organs and with the microbiota, or how these cells are modulated during development and adulthood are current topics of major interest. State-of-the-art research methodologies, including microscopy and potent in vivo imaging techniques, genomic and proteomic methods, current culture conditions together with the easy maintenance and manipulation of some fish embryos and adult specimens such as zebrafish (Danio rerio), have emerged and adapted to the phylogenetic position of some fish species. Furthermore, these advancements have facilitated the development of successful protocols aimed at addressing significant hypotheses and unresolved questions regarding vertebrate glia. The present review aims to analyse the available information on fish microglia, mainly the most recent one concerning teleosts, to establish an overview of their structural and immune functional features as a basis for their potentialities, heterogeneity, diversification, involvement, and relationships with neurons under normal and pathological conditions.
Collapse
Affiliation(s)
- Jose Carlos Campos-Sánchez
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - José Meseguer
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - Francisco A Guardiola
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain.
| |
Collapse
|
32
|
Lieberman JA, Mendelsohn A, Goldberg TE, Emsley R. Preventing disease progression in schizophrenia: What are we waiting for. J Psychiatr Res 2025; 181:716-727. [PMID: 39754992 DOI: 10.1016/j.jpsychires.2024.12.042] [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: 09/06/2023] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 01/06/2025]
Abstract
Despite research advances and progress in health care, schizophrenia remains a debilitating and costly disease. Onset occurs typically during youth and can lead to a relapsing and ultimately chronic course with persistent symptoms and functional impairment if not promptly and properly treated. Consequently, over time, schizophrenia causes substantial distress and disability for patients, their families and accrues to a collective burden to society. Recent research has revealed much about the pathophysiology that underlies the progressive nature of schizophrenia. Additionally, treatment strategies for disease management have been developed that have the potential to not just control psychotic symptoms but limit the cumulative morbidity of the illness. Given the evidence for their effectiveness and feasibility for their application, it is perplexing that this model of care has not yet become the standard of care and widely implemented to reduce the burden of illness on patients and society. This begs the question of whether the failure of implementation of a potentially disease-modifying strategy is due to the lack of evidence of efficacy (or belief in it) and readiness for implementation, or whether it's the lack of motivation and political will to support their utilization. To address this question, we reviewed and summarized the literature describing the natural history, pathophysiology and therapeutic strategies that can alleviate symptoms, prevent relapse, and potentially modify the course of schizophrenia. We conclude that, while we await further advances in mental health care from research, we must fully appreciate and take advantage of the effectiveness of existing treatments and overcome the attitudinal, policy, and infrastructural barriers to providing optimal mental health care capable of providing a disease-modifying treatment to patients with schizophrenia.
Collapse
Affiliation(s)
- Jeffrey A Lieberman
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| | - Alana Mendelsohn
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Terry E Goldberg
- Division of Geriatric Psychiatry, Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Robin Emsley
- Department of Psychiatry, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
33
|
Sokolova D, Ghansah SA, Puletti F, Georgiades T, De Schepper S, Zheng Y, Crowley G, Wu L, Rueda-Carrasco J, Koutsiouroumpa A, Muckett P, Freeman OJ, Khakh BS, Hong S. Astrocyte-derived MFG-E8 facilitates microglial synapse elimination in Alzheimer's disease mouse models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.31.606944. [PMID: 39257734 PMCID: PMC11383703 DOI: 10.1101/2024.08.31.606944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Region-specific synapse loss is an early pathological hallmark in Alzheimer's disease (AD). Emerging data in mice and humans highlight microglia, the brain-resident macrophages, as cellular mediators of synapse loss; however, the upstream modulators of microglia-synapse engulfment remain elusive. Here, we report a distinct subset of astrocytes, which are glial cells essential for maintaining synapse homeostasis, appearing in a region-specific manner with age and amyloidosis at onset of synapse loss. These astrocytes are distinguished by their peri-synaptic processes which are 'bulbous' in morphology, contain accumulated p62-immunoreactive bodies, and have reduced territorial domains, resulting in a decrease of astrocyte-synapse coverage. Using integrated in vitro and in vivo approaches, we show that astrocytes upregulate and secrete phagocytic modulator, milk fat globule-EGF factor 8 (MFG-E8), which is sufficient and necessary for promoting microglia-synapse engulfment in their local milieu. Finally, we show that knocking down Mfge8 specifically from astrocytes using a viral CRISPR-saCas9 system prevents microglia-synapse engulfment and ameliorates synapse loss in two independent amyloidosis mouse models of AD. Altogether, our findings highlight astrocyte-microglia crosstalk in determining synapse fate in amyloid models and nominate astrocytic MFGE8 as a potential target to ameliorate synapse loss during the earliest stages of AD.
Collapse
Affiliation(s)
- Dimitra Sokolova
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
- Neuroscience BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Shari Addington Ghansah
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Francesca Puletti
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Tatiana Georgiades
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Sebastiaan De Schepper
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Yongjing Zheng
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Gerard Crowley
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Ling Wu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Javier Rueda-Carrasco
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Angeliki Koutsiouroumpa
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Philip Muckett
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Oliver J. Freeman
- Neuroscience BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Baljit S. Khakh
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Soyon Hong
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| |
Collapse
|
34
|
Nagappan-Chettiar S, Burbridge TJ, Umemori H. Activity-Dependent Synapse Refinement: From Mechanisms to Molecules. Neuroscientist 2024; 30:673-689. [PMID: 37140155 PMCID: PMC11584027 DOI: 10.1177/10738584231170167] [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] [Indexed: 05/05/2023]
Abstract
The refinement of immature neuronal networks into efficient mature ones is critical to nervous system development and function. This process of synapse refinement is driven by the neuronal activity-dependent competition of converging synaptic inputs, resulting in the elimination of weak inputs and the stabilization of strong ones. Neuronal activity, whether in the form of spontaneous activity or experience-evoked activity, is known to drive synapse refinement in numerous brain regions. More recent studies are now revealing the manner and mechanisms by which neuronal activity is detected and converted into molecular signals that appropriately regulate the elimination of weaker synapses and stabilization of stronger ones. Here, we highlight how spontaneous activity and evoked activity instruct neuronal activity-dependent competition during synapse refinement. We then focus on how neuronal activity is transformed into the molecular cues that determine and execute synapse refinement. A comprehensive understanding of the mechanisms underlying synapse refinement can lead to novel therapeutic strategies in neuropsychiatric diseases characterized by aberrant synaptic function.
Collapse
Affiliation(s)
- Sivapratha Nagappan-Chettiar
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy J. Burbridge
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hisashi Umemori
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
35
|
Hu Y, Qi H, Yang J, Wang F, Peng X, Chen X, Zhu X. Wogonin mitigates microglia-mediated synaptic over-pruning and cognitive impairment following epilepsy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156222. [PMID: 39547095 DOI: 10.1016/j.phymed.2024.156222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/21/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Epilepsy, a neurological disorder characterized by recurrent abnormal neuronal discharges, leading to brain dysfunction and imposing significant psychological and economic burdens on patients. Microglia, the resident immune cells within the central nervous system (CNS), play a crucial role in maintaining CNS homeostasis. However, activated microglia can excessively prune synapses, exacerbating neuronal damage and cognitive dysfunction following epilepsy. Wogonin, a flavonoid from Scutellaria Baicalensis, has known neuroprotective effects via anti-inflammatory and antioxidative mechanisms, but its impact on microglial activation and synaptic pruning in neurons post-epilepsy remains unclear. METHODS Synaptic density was assessed using presynaptic marker Synaptophysin and postsynaptic marker Psd-95, and microglial phagocytosis was evaluated with fluorescent microspheres. Pilocarpine-induced mouse model of status epilepticus was used to evaluate synaptic density changes of mouse hippocampus following an intraperitoneal injection of wogonin (50 and 100 mg/kg). Memory and cognitive function in mice were subsequently evaluated using the Y-maze, object recognition, and Morris water maze tests. Single-cell sequencing was employed to investigate the underlying causes of microglial state alterations, followed by experimental validation. RESULTS Microglia were transitioned to an activated state post-epilepsy, exhibiting significantly enhanced phagocytic capacity. Correspondingly, levels of synaptophysin and Psd-95 were markedly reduced in neurons. Treatment with wogonin (100 mg/kg) significantly increased neuronal synaptic density and improved learning and memory deficits in epileptic mice. Further investigation revealed that wogonin inhibits the release of pro-inflammatory cytokines and synaptic phagocytosis of microglia by activating the AKT/FoxO1 pathway. CONCLUSIONS Wogonin could alleviate excessive synaptic pruning of epileptic neurons by microglia and improve cognitive dysfunction of epileptic mice via the AKT/FoxO1 pathway.
Collapse
Affiliation(s)
- Yang Hu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Medical School of Southeast University, Nanjing, China
| | - Honggang Qi
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Medical School of Southeast University, Nanjing, China
| | - Jiurong Yang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Medical School of Southeast University, Nanjing, China
| | - Feiyu Wang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Medical School of Southeast University, Nanjing, China
| | - Xintao Peng
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Xiang Chen
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Medical School of Southeast University, Nanjing, China
| | - Xinjian Zhu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Medical School of Southeast University, Nanjing, China.
| |
Collapse
|
36
|
Neziri S, Köseoğlu AE, Deniz Köseoğlu G, Özgültekin B, Özgentürk NÖ. Animal models in neuroscience with alternative approaches: Evolutionary, biomedical, and ethical perspectives. Animal Model Exp Med 2024; 7:868-880. [PMID: 39375824 DOI: 10.1002/ame2.12487] [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: 05/27/2024] [Accepted: 08/07/2024] [Indexed: 10/09/2024] Open
Abstract
Animal models have been a crucial tool in neuroscience research for decades, providing insights into the biomedical and evolutionary mechanisms of the nervous system, disease, and behavior. However, their use has raised concerns on several ethical, clinical, and scientific considerations. The welfare of animals and the 3R principles (replacement, reduction, refinement) are the focus of the ethical concerns, targeting the importance of reducing the stress and suffering of these models. Several laws and guidelines are applied and developed to protect animal rights during experimenting. Concurrently, in the clinic and biomedical fields, discussions on the relevance of animal model findings on human organisms have increased. Latest data suggest that in a considerable amount of time the animal model results are not translatable in humans, costing time and money. Alternative methods, such as in vitro (cell culture, microscopy, organoids, and micro physiological systems) techniques and in silico (computational) modeling, have emerged as potential replacements for animal models, providing more accurate data in a minimized cost. By adopting alternative methods and promoting ethical considerations in research practices, we can achieve the 3R goals while upholding our responsibility to both humans and other animals. Our goal is to present a thorough review of animal models used in neuroscience from the biomedical, evolutionary, and ethical perspectives. The novelty of this research lies in integrating diverse points of views to provide an understanding of the advantages and disadvantages of animal models in neuroscience and in discussing potential alternative methods.
Collapse
Affiliation(s)
- Sabina Neziri
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Yıldız Technical University, Istanbul, Turkey
| | | | | | - Buminhan Özgültekin
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acıbadem University, Istanbul, Turkey
| | - Nehir Özdemir Özgentürk
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Yıldız Technical University, Istanbul, Turkey
| |
Collapse
|
37
|
Sebastian R, Song Y, Pak C. Probing the molecular and cellular pathological mechanisms of schizophrenia using human induced pluripotent stem cell models. Schizophr Res 2024; 273:4-23. [PMID: 35835709 PMCID: PMC9832179 DOI: 10.1016/j.schres.2022.06.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 01/13/2023]
Abstract
With recent advancements in psychiatric genomics, as a field, "stem cell-based disease modelers" were given the exciting yet daunting task of translating the extensive list of disease-associated risks into biologically and clinically relevant information in order to deliver therapeutically meaningful leads and insights. Despite their limitations, human induced pluripotent stem cell (iPSCs) based models have greatly aided our understanding of the molecular and cellular mechanisms underlying the complex etiology of brain disorders including schizophrenia (SCZ). In this review, we summarize the major findings from studies in the past decade which utilized iPSC models to investigate cell type-specific phenotypes relevant to idiopathic SCZ and disease penetrant alleles. Across cell type differences, several biological themes emerged, serving as potential neurodevelopmental mechanisms of SCZ, including oxidative stress and mitochondrial dysfunction, depletion of progenitor pools and insufficient differentiation potential of these progenitors, and structural and functional deficits of neurons and other supporting cells. Here, we discuss both the recent progress as well as challenges and improvements needed for future studies utilizing iPSCs as a model for SCZ and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Rebecca Sebastian
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA; Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Yoonjae Song
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA
| | - ChangHui Pak
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
38
|
Makowski C, Nichols TE, Dale AM. Quality over quantity: powering neuroimaging samples in psychiatry. Neuropsychopharmacology 2024; 50:58-66. [PMID: 38902353 PMCID: PMC11525971 DOI: 10.1038/s41386-024-01893-4] [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: 03/22/2024] [Revised: 05/06/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024]
Abstract
Neuroimaging has been widely adopted in psychiatric research, with hopes that these non-invasive methods will provide important clues to the underpinnings and prediction of various mental health symptoms and outcomes. However, the translational impact of neuroimaging has not yet reached its promise, despite the plethora of computational methods, tools, and datasets at our disposal. Some have lamented that too many psychiatric neuroimaging studies have been underpowered with respect to sample size. In this review, we encourage this discourse to shift from a focus on sheer increases in sample size to more thoughtful choices surrounding experimental study designs. We propose considerations at multiple decision points throughout the study design, data modeling and analysis process that may help researchers working in psychiatric neuroimaging boost power for their research questions of interest without necessarily increasing sample size. We also provide suggestions for leveraging multiple datasets to inform each other and strengthen our confidence in the generalization of findings to both population-level and clinical samples. Through a greater emphasis on improving the quality of brain-based and clinical measures rather than merely quantity, meaningful and potentially translational clinical associations with neuroimaging measures can be achieved with more modest sample sizes in psychiatry.
Collapse
Affiliation(s)
- Carolina Makowski
- Department of Radiology, University of California San Diego, San Diego, CA, USA.
| | - Thomas E Nichols
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Anders M Dale
- Departments of Radiology and Neurosciences, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
39
|
Choudhary A, Peles D, Nayak R, Mizrahi L, Stern S. Current progress in understanding schizophrenia using genomics and pluripotent stem cells: A meta-analytical overview. Schizophr Res 2024; 273:24-38. [PMID: 36443183 DOI: 10.1016/j.schres.2022.11.001] [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: 06/23/2022] [Revised: 10/16/2022] [Accepted: 11/01/2022] [Indexed: 11/27/2022]
Abstract
Schizophrenia (SCZ) is a complex, heritable and polygenic neuropsychiatric disease, which disables the patients as well as decreases their life expectancy and quality of life. Common and rare variants studies on SCZ subjects have provided >100 genomic loci that hold importance in the context of SCZ pathophysiology. Transcriptomic studies from clinical samples have informed about the differentially expressed genes (DEGs) and non-coding RNAs in SCZ patients. Despite these advancements, no causative genes for SCZ were found and hence SCZ is difficult to recapitulate in animal models. In the last decade, induced Pluripotent Stem Cells (iPSCs)-based models have helped in understanding the neural phenotypes of SCZ by studying patient iPSC-derived 2D neuronal cultures and 3D brain organoids. Here, we have aimed to provide a simplistic overview of the current progress and advancements after synthesizing the enormous literature on SCZ genetics and SCZ iPSC-based models. Although further understanding of SCZ genetics and pathophysiological mechanisms using these technological advancements is required, the recent approaches have allowed to delineate important cellular mechanisms and biological pathways affected in SCZ.
Collapse
Affiliation(s)
- Ashwani Choudhary
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - David Peles
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Ritu Nayak
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Liron Mizrahi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
40
|
Muhtaseb AW, Duan J. Modeling common and rare genetic risk factors of neuropsychiatric disorders in human induced pluripotent stem cells. Schizophr Res 2024; 273:39-61. [PMID: 35459617 PMCID: PMC9735430 DOI: 10.1016/j.schres.2022.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 12/13/2022]
Abstract
Recent genome-wide association studies (GWAS) and whole-exome sequencing of neuropsychiatric disorders, especially schizophrenia, have identified a plethora of common and rare disease risk variants/genes. Translating the mounting human genetic discoveries into novel disease biology and more tailored clinical treatments is tied to our ability to causally connect genetic risk variants to molecular and cellular phenotypes. When combined with the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated (Cas) nuclease-mediated genome editing system, human induced pluripotent stem cell (hiPSC)-derived neural cultures (both 2D and 3D organoids) provide a promising tractable cellular model for bridging the gap between genetic findings and disease biology. In this review, we first conceptualize the advances in understanding the disease polygenicity and convergence from the past decade of iPSC modeling of different types of genetic risk factors of neuropsychiatric disorders. We then discuss the major cell types and cellular phenotypes that are most relevant to neuropsychiatric disorders in iPSC modeling. Finally, we critically review the limitations of iPSC modeling of neuropsychiatric disorders and outline the need for implementing and developing novel methods to scale up the number of iPSC lines and disease risk variants in a systematic manner. Sufficiently scaled-up iPSC modeling and a better functional interpretation of genetic risk variants, in combination with cutting-edge CRISPR/Cas9 gene editing and single-cell multi-omics methods, will enable the field to identify the specific and convergent molecular and cellular phenotypes in precision for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Abdurrahman W Muhtaseb
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, United States of America; Department of Human Genetics, The University of Chicago, Chicago, IL 60637, United States of America
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, United States of America; Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL 60637, United States of America.
| |
Collapse
|
41
|
Santarriaga S, Gerlovin K, Layadi Y, Karmacharya R. Human stem cell-based models to study synaptic dysfunction and cognition in schizophrenia: A narrative review. Schizophr Res 2024; 273:78-97. [PMID: 36925354 PMCID: PMC10500041 DOI: 10.1016/j.schres.2023.02.029] [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: 10/21/2022] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
Cognitive impairment is the strongest predictor of functional outcomes in schizophrenia and is hypothesized to result from synaptic dysfunction. However, targeting synaptic plasticity and cognitive deficits in patients remains a significant clinical challenge. A comprehensive understanding of synaptic plasticity and the molecular basis of learning and memory in a disease context can provide specific targets for the development of novel therapeutics targeting cognitive impairments in schizophrenia. Here, we describe the role of synaptic plasticity in cognition, summarize evidence for synaptic dysfunction in schizophrenia and demonstrate the use of patient derived induced-pluripotent stem cells for studying synaptic plasticity in vitro. Lastly, we discuss current advances and future technologies for bridging basic science research of synaptic dysfunction with clinical and translational research that can be used to predict treatment response and develop novel therapeutics.
Collapse
Affiliation(s)
- Stephanie Santarriaga
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Kaia Gerlovin
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yasmine Layadi
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chimie ParisTech, Université Paris Sciences et Lettres, Paris, France
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
42
|
Liu D, Yang S, Yu S. Interactions Between Ferroptosis and Oxidative Stress in Ischemic Stroke. Antioxidants (Basel) 2024; 13:1329. [PMID: 39594471 PMCID: PMC11591163 DOI: 10.3390/antiox13111329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 11/28/2024] Open
Abstract
Ischemic stroke is a devastating condition that occurs due to the interruption of blood flow to the brain, resulting in a range of cellular and molecular changes. In recent years, there has been growing interest in the role of ferroptosis, a newly identified form of regulated cell death, in ischemic stroke. Ferroptosis is driven by the accumulation of lipid peroxides and is characterized by the loss of membrane integrity. Additionally, oxidative stress, which refers to an imbalance between prooxidants and antioxidants, is a hallmark of ischemic stroke and significantly contributes to the pathogenesis of the disease. In this review, we explore the interactions between ferroptosis and oxidative stress in ischemic stroke. We examine the underlying mechanisms through which oxidative stress induces ferroptosis and how ferroptosis, in turn, exacerbates oxidative stress. Furthermore, we discuss potential therapeutic strategies that target both ferroptosis and oxidative stress in the treatment of ischemic stroke. Overall, this review highlights the complex interplay between ferroptosis and oxidative stress in ischemic stroke and underscores the need for further research to identify novel therapeutic targets for this condition.
Collapse
Affiliation(s)
| | - Sha Yang
- College of Acupuncture and Massage, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China;
| | - Shuguang Yu
- College of Acupuncture and Massage, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China;
| |
Collapse
|
43
|
Stern S, Zhang L, Wang M, Wright R, Rosh I, Hussein Y, Stern T, Choudhary A, Tripathi U, Reed P, Sadis H, Nayak R, Shemen A, Agarwal K, Cordeiro D, Peles D, Hang Y, Mendes APD, Baul TD, Roth JG, Coorapati S, Boks MP, McCombie WR, Hulshoff Pol H, Brennand KJ, Réthelyi JM, Kahn RS, Marchetto MC, Gage FH. Monozygotic twins discordant for schizophrenia differ in maturation and synaptic transmission. Mol Psychiatry 2024; 29:3208-3222. [PMID: 38704507 PMCID: PMC11449799 DOI: 10.1038/s41380-024-02561-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 04/01/2024] [Accepted: 04/12/2024] [Indexed: 05/06/2024]
Abstract
Schizophrenia affects approximately 1% of the world population. Genetics, epigenetics, and environmental factors are known to play a role in this psychiatric disorder. While there is a high concordance in monozygotic twins, about half of twin pairs are discordant for schizophrenia. To address the question of how and when concordance in monozygotic twins occur, we have obtained fibroblasts from two pairs of schizophrenia discordant twins (one sibling with schizophrenia while the second one is unaffected by schizophrenia) and three pairs of healthy twins (both of the siblings are healthy). We have prepared iPSC models for these 3 groups of patients with schizophrenia, unaffected co-twins, and the healthy twins. When the study started the co-twins were considered healthy and unaffected but both the co-twins were later diagnosed with a depressive disorder. The reprogrammed iPSCs were differentiated into hippocampal neurons to measure the neurophysiological abnormalities in the patients. We found that the neurons derived from the schizophrenia patients were less arborized, were hypoexcitable with immature spike features, and exhibited a significant reduction in synaptic activity with dysregulation in synapse-related genes. Interestingly, the neurons derived from the co-twin siblings who did not have schizophrenia formed another distinct group that was different from the neurons in the group of the affected twin siblings but also different from the neurons in the group of the control twins. Importantly, their synaptic activity was not affected. Our measurements that were obtained from schizophrenia patients and their monozygotic twin and compared also to control healthy twins point to hippocampal synaptic deficits as a central mechanism in schizophrenia.
Collapse
Affiliation(s)
- Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel.
| | - Lei Zhang
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Meiyan Wang
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Rebecca Wright
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Idan Rosh
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Yara Hussein
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Tchelet Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Ashwani Choudhary
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Utkarsh Tripathi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Patrick Reed
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Hagit Sadis
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Ritu Nayak
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Aviram Shemen
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Karishma Agarwal
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Diogo Cordeiro
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - David Peles
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Yuqing Hang
- Razavi Newman Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ana P D Mendes
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Tithi D Baul
- Department of Psychiatry at the Boston Medical Center, Boston, MA, USA
| | - Julien G Roth
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Shashank Coorapati
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Marco P Boks
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| | | | - Hilleke Hulshoff Pol
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
- Department of Experimental Psychology, Utrecht University, Heidelberglaan 1, 3584CS, Utrecht, The Netherlands
| | - Kristen J Brennand
- Nash Family Department of Neuroscience, Friedman Brain Institute, Pamela Sklar Division of Psychiatric Genomics, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Department of Genetics, Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - János M Réthelyi
- Molecular Psychiatry Research Group and Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - René S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, New York, NY, USA
| | - Maria C Marchetto
- Department of Anthropology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
44
|
Cavaleri D, De Pietra A, Gazzola M, Crocamo C, Bartoli F, Carrà G. Complete blood count-based inflammation indexes and symptom severity in people with schizophrenia spectrum disorders: An analysis based on structural equation modelling. Psychoneuroendocrinology 2024; 168:107134. [PMID: 39059225 DOI: 10.1016/j.psyneuen.2024.107134] [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: 04/12/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024]
Abstract
INTRODUCTION Schizophrenia spectrum disorders (SSDs) are associated with immune-inflammatory activation. Recently, complete blood count (CBC)-based inflammation indexes such as the neutrophil-to-lymphocyte ratio (NLR), the monocyte-to-lymphocyte ratio (MLR), and the platelet-to-lymphocyte ratio (PLR) have emerged as reproducible and cost-effective inflammation markers in mental disorders. In this study, we aimed at investigating the relationship of NLR, MLR, and PLR with symptom severity in people with SSDs, testing interactions with relevant clinical variables. METHODS We included inpatients with SSDs aged 18-65 consecutively hospitalized from May 2020 to March 2024. Socio-demographic and clinical data were recorded. CBC-based ratios were estimated from routinely collected blood samples. Structural equation modelling (SEM) was performed to test relationships involving symptom severity constructs and CBC-based ratios, accounting for substance use disorder, antipsychotic treatment, and obesity. RESULTS Two hundred sixty-six participants met inclusion criteria. The SEM analysis uncovered a significant relationship of MLR with positive (coeff.: 0.19, p=0.048) and negative (coeff.: 0.27, p=0.004) symptoms, also showing a significant link of substance use disorder and antipsychotic treatment with symptom severity as well as of antipsychotic treatment with obesity. CONCLUSIONS Notwithstanding the cross-sectional design and the somewhat limited sample representativeness, this study showed a significant relationship between the MLR - but not the NLR or the PLR - and the severity of both positive and negative symptoms, testing at the same time the interactions with other clinical variables. Considering the insufficiency and inconsistency of data in this field, further research is needed to validate our findings and elucidate the underlying mechanisms driving the observed relationships between the MLR and SSD symptoms.
Collapse
Affiliation(s)
- Daniele Cavaleri
- School of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Aldo De Pietra
- School of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Marco Gazzola
- School of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Cristina Crocamo
- School of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Francesco Bartoli
- School of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy.
| | - Giuseppe Carrà
- School of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| |
Collapse
|
45
|
Sharkey RJ, Bacon C, Peterson Z, Rootes-Murdy K, Salvador R, Pomarol-Clotet E, Karuk A, Homan P, Ji E, Omlor W, Homan S, Georgiadis F, Kaiser S, Kirschner M, Ehrlich S, Dannlowski U, Grotegerd D, Goltermann J, Meinert S, Kircher T, Stein F, Brosch K, Krug A, Nenadic I, Sim K, Spalletta G, Banaj N, Sponheim SR, Demro C, Ramsay IS, King M, Quidé Y, Green MJ, Nguyen D, Preda A, Calhoun V, Turner J, van Erp T, Nickl-Jockschat T. Differences in the neural correlates of schizophrenia with positive and negative formal thought disorder in patients with schizophrenia in the ENIGMA dataset. Mol Psychiatry 2024; 29:3086-3096. [PMID: 38671214 PMCID: PMC11449795 DOI: 10.1038/s41380-024-02563-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024]
Abstract
Formal thought disorder (FTD) is a clinical key factor in schizophrenia, but the neurobiological underpinnings remain unclear. In particular, the relationship between FTD symptom dimensions and patterns of regional brain volume loss in schizophrenia remains to be established in large cohorts. Even less is known about the cellular basis of FTD. Our study addresses these major obstacles by enrolling a large multi-site cohort acquired by the ENIGMA Schizophrenia Working Group (752 schizophrenia patients and 1256 controls), to unravel the neuroanatomy of FTD in schizophrenia and using virtual histology tools on implicated brain regions to investigate the cellular basis. Based on the findings of previous clinical and neuroimaging studies, we decided to separately explore positive, negative and total formal thought disorder. We used virtual histology tools to relate brain structural changes associated with FTD to cellular distributions in cortical regions. We identified distinct neural networks positive and negative FTD. Both networks encompassed fronto-occipito-amygdalar brain regions, but positive and negative FTD demonstrated a dissociation: negative FTD showed a relative sparing of orbitofrontal cortical thickness, while positive FTD also affected lateral temporal cortices. Virtual histology identified distinct transcriptomic fingerprints associated for both symptom dimensions. Negative FTD was linked to neuronal and astrocyte fingerprints, while positive FTD also showed associations with microglial cell types. These results provide an important step towards linking FTD to brain structural changes and their cellular underpinnings, providing an avenue for a better mechanistic understanding of this syndrome.
Collapse
Affiliation(s)
- Rachel J Sharkey
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Chelsea Bacon
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Zeru Peterson
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | | | - Raymond Salvador
- FIDMAG Germanes Hospitalàries Research Foundation, CIBERSAM ISCIII, Barcelona, Spain
| | - Edith Pomarol-Clotet
- FIDMAG Germanes Hospitalàries Research Foundation, CIBERSAM ISCIII, Barcelona, Spain
| | - Andriana Karuk
- FIDMAG Germanes Hospitalàries Research Foundation, CIBERSAM ISCIII, Barcelona, Spain
| | - Philipp Homan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich (PUK), Zurich, 8008, Switzerland
| | - Ellen Ji
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich (PUK), Zurich, 8008, Switzerland
| | - Wolfgang Omlor
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich (PUK), Zurich, 8008, Switzerland
| | - Stephanie Homan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich (PUK), Zurich, 8008, Switzerland
| | - Foivos Georgiadis
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich (PUK), Zurich, 8008, Switzerland
| | - Stefan Kaiser
- Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Matthias Kirschner
- Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Stefan Ehrlich
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Janik Goltermann
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Axel Krug
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Igor Nenadic
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Kang Sim
- West Region, Institute of Mental Health, Singapore, Singapore
| | | | - Nerisa Banaj
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Scott R Sponheim
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Caroline Demro
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Ian S Ramsay
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | | | - Yann Quidé
- School of Psychiatry, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
| | - Melissa Jane Green
- School of Psychiatry, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
| | - Dana Nguyen
- Department of Pediatric Neurology, University of California Irvine, Irvine, CA, USA
| | - Adrian Preda
- Department of Pediatric Neurology, University of California Irvine, Irvine, CA, USA
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, USA
| | - Vince Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GE, USA
| | - Jessica Turner
- Department of Psychiatry and Behavioral Medicine, Ohio State University, Columbus, OH, USA
| | - Theo van Erp
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA, USA
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, USA
| | - Thomas Nickl-Jockschat
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA.
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Department of Psychiatry and Psychotherapy, Otto-von-Guericke University, Magdeburg, Germany.
- German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Magdeburg, Germany.
- Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Magdeburg, Germany.
| |
Collapse
|
46
|
Koss KM, Son T, Li C, Hao Y, Cao J, Churchward MA, Zhang ZJ, Wertheim JA, Derda R, Todd KG. Toward discovering a novel family of peptides targeting neuroinflammatory states of brain microglia and astrocytes. J Neurochem 2024; 168:3386-3414. [PMID: 37171455 PMCID: PMC10640667 DOI: 10.1111/jnc.15840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/13/2023]
Abstract
Microglia are immune-derived cells critical to the development and healthy function of the brain and spinal cord, yet are implicated in the active pathology of many neuropsychiatric disorders. A range of functional phenotypes associated with the healthy brain or disease states has been suggested from in vivo work and were modeled in vitro as surveying, reactive, and primed sub-types of primary rat microglia and mixed microglia/astrocytes. It was hypothesized that the biomolecular profile of these cells undergoes a phenotypical change as well, and these functional phenotypes were explored for potential novel peptide binders using a custom 7 amino acid-presenting M13 phage library (SX7) to identify unique peptides that bind differentially to these respective cell types. Surveying glia were untreated, reactive were induced with a lipopolysaccharide treatment, recovery was modeled with a potent anti-inflammatory treatment dexamethasone, and priming was determined by subsequently challenging the cells with interferon gamma. Microglial function was profiled by determining the secretion of cytokines and nitric oxide, and expression of inducible nitric oxide synthase. After incubation with the SX7 phage library, populations of SX7-positive microglia and/or astrocytes were collected using fluorescence-activated cell sorting, SX7 phage was amplified in Escherichia coli culture, and phage DNA was sequenced via next-generation sequencing. Binding validation was done with synthesized peptides via in-cell westerns. Fifty-eight unique peptides were discovered, and their potential functions were assessed using a basic local alignment search tool. Peptides potentially originated from proteins ranging in function from a variety of supportive glial roles, including synapse support and pruning, to inflammatory incitement including cytokine and interleukin activation, and potential regulation in neurodegenerative and neuropsychiatric disorders.
Collapse
Affiliation(s)
- K M Koss
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Alberta, Edmonton, Canada
- Department of Surgery, University of Arizona College of Medicine, Arizona, Tucson, USA
| | - T Son
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
| | - C Li
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
| | - Y Hao
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
| | - J Cao
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
- 48Hour Discovery Inc, 11421 Saskatchewan Dr NW, Edmonton, AB T6G 2M9, Canada
| | - M A Churchward
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Alberta, Edmonton, Canada
- Department of Biology and Environmental Sciences, Concordia University of Edmonton, Alberta, Edmonton, Canada
| | - Z J Zhang
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
| | - J A Wertheim
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
- Department of Surgery, University of Arizona College of Medicine, Arizona, Tucson, USA
| | - R Derda
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
- 48Hour Discovery Inc, 11421 Saskatchewan Dr NW, Edmonton, AB T6G 2M9, Canada
| | - K G Todd
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Alberta, Edmonton, Canada
- Department of Biomedical Engineering, University of Alberta, Alberta, Edmonton, Canada
| |
Collapse
|
47
|
Jiang Y, Sachdeva K, Goulbourne CN, Berg MJ, Peddy J, Stavrides PH, Pensalfini A, Pawlik M, Whyte L, Balapal BS, Shivakumar S, Bleiwas C, Smiley JF, Mathews PM, Nixon RA. Increased neuronal expression of the early endosomal adaptor APPL1 leads to endosomal and synaptic dysfunction with cholinergic neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613736. [PMID: 39345644 PMCID: PMC11430014 DOI: 10.1101/2024.09.19.613736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Dysfunction of the endolysosomal system within neurons is a prominent feature of Alzheimer's disease (AD) pathology. Multiple AD-risk factors are known to cause hyper-activity of the early-endosome small GTPase rab5, resulting in neuronal endosomal pathway disruption. APPL1, an important rab5 effector protein, is an interface between endosomal and neuronal function through a rab5-activating interaction with the BACE1-generated C-terminal fragment (βCTF or C99) of the amyloid precursor protein (APP), a pathogenic APP fragment generated within endolysosomal compartments. To better understand the role of APPL1 in the AD endosomal phenotype, we generated a transgenic mouse model over-expressing human APPL1 within neurons (Thy1-APPL1 mice). Consistent with the important endosomal regulatory role of APPL1, Thy1-APPL1 mice have enlarged neuronal early endosomes and increased synaptic endocytosis due to increased rab5 activation. We additionally demonstrate pathological consequences of APPL1 overexpression, including functional changes in hippocampal long-term potentiation (LTP) and long-term depression (LTD), as well as degeneration of the large projection cholinergic neurons of the basal forebrain and impairment of hippocampal-dependent memory. Our findings show that increased neuronal APPL1 levels lead to a cascade of pathological effects within neurons, including early endosomal alterations, synaptic dysfunction, and neurodegeneration. Multiple risk factors and molecular regulators, including APPL1 activity, are known to contribute to the endosomal dysregulation seen in the early stages of AD, and these findings further highlight the shared pathobiology and consequences to a neuron of early endosomal pathway disruption. Significance Statement Dysfunction in the endolysosomal system within neurons is a key feature of Alzheimer's disease (AD). Multiple AD risk factors lead to hyperactivity of the early-endosome GTPase rab5, disrupting neuronal pathways including the cholinergic circuits involved early in memory decline. APPL1, a crucial rab5 effector, connects endosomal and neuronal functions through its interaction with a specific amyloid precursor protein (APP) fragment generated within endosomes. To understand APPL1's role, a transgenic mouse model over-expressing human APPL1 in neurons (Thy1-APPL1 mice) was developed. These mice show enlarged early endosomes and increased synaptic endocytosis due to rab5 activation, resulting in impaired hippocampal long-term potentiation and depression, the degeneration of basal forebrain cholinergic neurons, and memory deficits, highlighting a pathological cascade mediated through APPL1 at the early endosome.
Collapse
|
48
|
de Deus JL, Faborode OS, Nandi S. Synaptic Pruning by Microglia: Lessons from Genetic Studies in Mice. Dev Neurosci 2024:1-21. [PMID: 39265565 DOI: 10.1159/000541379] [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: 06/14/2024] [Accepted: 09/06/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Neural circuits are subjected to refinement throughout life. The dynamic addition and elimination (pruning) of synapses are necessary for maturation of neural circuits and synaptic plasticity. Due to their phagocytic nature, microglia have been considered as the primary mediators of synaptic pruning. Synaptic pruning can strengthen an active synapse by removing excess weaker synapses during development. Inappropriate synaptic pruning can often influence a disease outcome or an injury response. SUMMARY This review offers a focused discussion on microglial roles in synaptic pruning, based on the evidence gathered from genetic manipulations in mice. Genetically labeled microglia and synapses often allow assessment of their interactions in real time. Further manipulations involving synaptically localized molecules, neuronally or glial-derived diffusible factors, and their respective cognate receptors in microglia provide critical evidence in support of a direct role of microglia in synaptic pruning. KEY MESSAGE We discuss microglial contact-dependent "eat-me," "don't-eat-me," and "find-me" signals, as well as recently identified noncontact pruning, under the contexts of neural circuit, brain region, developmental window, and an injury or a disease state.
Collapse
Affiliation(s)
- Junia Lara de Deus
- Department of Anatomy, Howard University College of Medicine, Washington, DC, USA
| | | | - Sayan Nandi
- Department of Anatomy, Howard University College of Medicine, Washington, DC, USA
| |
Collapse
|
49
|
Quagliato LA, Nardi AE. Childhood Trauma and the Immune System: A Complex Interaction That Can Lead to Biopsychosocial Pathogenesis. PSYCHOTHERAPY AND PSYCHOSOMATICS 2024; 93:304-307. [PMID: 39265551 DOI: 10.1159/000541094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 08/21/2024] [Indexed: 09/14/2024]
Affiliation(s)
- Laiana A Quagliato
- Psychiatry Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Antonio E Nardi
- Psychiatry Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
50
|
Ketharanathan T, Pereira A, Sundram S. Gene expression changes in Brodmann's Area 46 differentiate epidermal growth factor and immune system interactions in schizophrenia and mood disorders. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:76. [PMID: 39242583 PMCID: PMC11379811 DOI: 10.1038/s41537-024-00488-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 07/16/2024] [Indexed: 09/09/2024]
Abstract
How early in life stress-immune related environmental factors increase risk predisposition to schizophrenia remains unknown. We examined if pro-inflammatory changes perturb the brain epidermal growth factor (EGF) system, a system critical for neurodevelopment and mature CNS functions including synaptic plasticity. We quantified genes from key EGF and immune system pathways for mRNA levels and eight immune proteins in post-mortem dorsolateral prefrontal (DLPFC; Brodmann's Area (BA) 46) and orbitofrontal (OFC; BA11) cortices from people with schizophrenia, mood disorders and neurotypical controls. In BA46, 64 genes were differentially expressed, predominantly in schizophrenia, where attenuated expression of the MAPK-ERK, NRG1-PI3K-AKT and mTOR cascades indicated reduced EGF system signalling, and similarly diminished immune molecular expression, notably in TLR, TNF and complement pathways, along with low NF-κB1 and elevated IL12RB2 protein levels were noted. There was nominal evidence for altered convergence between ErbB-PI3K-AKT-mTOR and TLR pathways in BA46 in schizophrenia. Comparatively minimal changes were noted in BA11. Overall, distinct pathway gene expression changes may reflect variant pathological processes involving immune and EGF system signalling between schizophrenia and mood disorder, particularly in DLPFC. Further, the abnormal convergence between innate immune signalling and candidate EGF signalling pathways may indicate a pathologically important interaction in the developing brain in response to environmental stressors.
Collapse
Affiliation(s)
- Tharini Ketharanathan
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia.
- Department of Psychiatry, University of Melbourne, Parkville, VIC 3052, Australia.
- Northern Health, Epping, VIC 3076, Australia.
| | - Avril Pereira
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia
- Department of Psychiatry, University of Melbourne, Parkville, VIC 3052, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
- Mental Health Program, Monash Health, Clayton, VIC 3168, Australia
| |
Collapse
|