1
|
Jiang H, Bai W, Yang Y, Zhou G, Li J, Li X, Wan X, Shao J. Biliverdin alleviates cerebral ischemia-reperfusion injury by regulating the P4hb/MAPK/mTOR pathway to inhibit autophagy. Cell Signal 2025; 132:111815. [PMID: 40258578 DOI: 10.1016/j.cellsig.2025.111815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/26/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND Biliverdin (BV) exhibits anti-inflammatory and antioxidative effects. Autophagy activation is crucial in the pathogenesis of cerebral ischemia-reperfusion injury (CIRI). This study aimed to investigate whether BV could ameliorate CIRI by regulating autophagy. METHODS A middle cerebral artery occlusion-reperfusion (MCAO/R) model in Sprague-Dawley (SD) rats and an oxygen-glucose deprivation/reoxygenation (OGD/R) model in PC12 cells were employed to explore the neuroprotective effects of BV and its underlying mechanisms. In these rats, once BV was administered post-MCAO/R, its treatment efficacy and underlying mechanisms were evaluated through behavioral, morphological, and molecular analyses. Alternatively, for PC12 cells, following successful OGD/R modeling, BV, autophagy activator rapamycin, prolyl 4-hydroxylase beta (P4hb) knockdown or overexpression, and the specific inhibitors of three classic autophagy pathways were applied. Cell viability (using CCK8 assay), Calcein/PI staining, autophagosome staining (using MDC assay), reverse transcription quantitative polymerase chain reaction, and western blot were subsequently carried out to investigate the mechanisms by which BV ameliorates CIRI. RESULTS BV alleviated CIRI by inhibiting autophagy. Further investigation suggested that BV downregulated P4hb expression. In vitro experiments showed that P4hb knockdown reduced autophagy in post-CIRI cells, while its overexpression reversed the effects of BV. Rescue experiments indicated that MAPK pathway inhibitors counteracted the effects of P4hb overexpression on autophagy post-CIRI. CONCLUSION BV improves CIRI by regulating the P4hb/MAPK/mTOR signaling pathway to inhibit autophagy, offering a novel therapeutic strategy for ischemic stroke.
Collapse
Affiliation(s)
- Huan Jiang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, 650032 Kunming, Yunnan Province, China
| | - Wenya Bai
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, 650032 Kunming, Yunnan Province, China
| | - Yuan Yang
- Department of Anesthesiology, Second Affiliated Hospital of Kunming Medical University, 650000 Kunming, Yunnan Province, China
| | - Guilin Zhou
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, 650032 Kunming, Yunnan Province, China
| | - Junjie Li
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, 650032 Kunming, Yunnan Province, China
| | - Xuelian Li
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, 650032 Kunming, Yunnan Province, China
| | - Xiaohong Wan
- Department of Anesthesiology, Second Affiliated Hospital of Kunming Medical University, 650000 Kunming, Yunnan Province, China.
| | - Jianlin Shao
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, 650032 Kunming, Yunnan Province, China.
| |
Collapse
|
2
|
Li J, Wu D, Mu Y, Wang Y, Zhai L, Jia L. The combined damage of bisphenol A and high fat diet to learning and memory in young male mice: the regulatory effect of BDNF/TrkB/PI3K/AKT pathway on autophagy. ENVIRONMENTAL RESEARCH 2025; 276:121538. [PMID: 40187395 DOI: 10.1016/j.envres.2025.121538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/12/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
With the popularity of takeaway and processed food, combined exposure to Bisphenol A (BPA) and high fat diet (HFD) is becoming increasingly common. BPA or HFD intake in children could impair learning and memory ability, but the combined effect and mechanisms remain unclear. In this study, we fed young male mice with 0.1 μg/mL BPA (L-BPA), 0.2 μg/mL BPA (H-BPA), 60 %HFD (HFD), 0.1 BPA + HFD (L-BPA + HFD) and 0.2 BPA + HFD (H-BPA + HFD) for 8 weeks. The results showed that recognition memory and free exploration of mice were impaired in the BPA or HFD group, and the damage of exploration was more severe in the combined group. All treated groups showed morphological changes in hippocampal neurons. The levels of synaptic structural protein PSD-95 and SYN were reduced in BPA and HFD alone or in combination groups. BPA or HFD led to changes in autophagy levels in the hippocampus, manifested by decreased protein levels of mTOR and P62, increased level of LC3B, and more significant changes in the combined group. The BDNF/TrkB/PI3K/AKT pathway was inhibited in BPA or HFD groups, especially in the combined group. Our results suggested that combined BPA with HFD exposure could impair learning and memory ability, and the combined effect might be related to the BDNF/TrkB/PI3K/AKT pathway, which regulated mTOR mediated autophagy and finally caused hippocampal synaptic damage.
Collapse
Affiliation(s)
- Jinshi Li
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Dan Wu
- Department of Child and Adolescent Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
| | - Yuyang Mu
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Yunzhu Wang
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Lingling Zhai
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Lihong Jia
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
3
|
Eissa N, Jayaprakash P, Aljneibi S, Alsaadi A, Alzaabi S, Łazewska D, Karcz T, Kieć-Kononowicz K, Sadek B. Mitigating effects of H3 receptor antagonism on cerebellar autophagic pathways and behavioral phenotypes in BTBR T+ tf/J mouse model of autism spectrum disorder. Eur J Pharmacol 2025; 997:177481. [PMID: 40058755 DOI: 10.1016/j.ejphar.2025.177481] [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: 12/13/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Accumulation of evidence suggested the involvement of autophagic pathways and their associated AktmTOR (mammalian target of rapamycin) signalling cascade in the pathogenesis of autism spectrum disorder (ASD). Histamine 3 receptors antagonism may be neuroprotective in ASD, as this antagonism modulates autophagy which is reported to be impaired in ASD. Therefore, the effects the novel H3 receptor antagonist E169 (2.5, 5, and 10 mg/kg, i.p.) on short-term memory (STM), long-term memory (LTM), and anxiety level in male Black and Tan BRachyury (BTBR) mice were evaluated using Novel object recognition test (NORT) and open field locomotor (OFT) tests respectively. In NORT, E169 (2.5 mg/kg, i.p.) significantly improved the memory of tested BTBR mice, and the effects of E169 were similar to those of the reference mTOR inhibitor rapamycin, and were reversed following co-administration with the centrally penetrant H3 receptor agonist (R)-α-methylhistamine (RAMH). Furthermore, E169 enhanced the BTBR memory by inhibiting H3 receptors and regulating the extent of disruption in the expression of cerebellar Akt, mTOR, and LC-3 proteins of treated mice. Moreover, E169 (2.5 mg/kg, i.p.) restored the disturbed anxiety levels and hyperactivity observed in OFT. In summary, the findings indicate that H3 receptor antagonists like E169 could play a role in simultaneously regulating disrupted brain neurotransmitters and the dysregulated cerebellar Akt-mTOR signaling pathway associated with autophagy in neurological diseases. Therefore, activation of cerebellar autophagy represented by H3 receptor antagonist E169 may serve as an effective pharmacological therapeutic target for the ASD-like behavioral phenotypes and may add new therapeutic management strategy for the multifactorial disorder ASD.
Collapse
Affiliation(s)
- Nermin Eissa
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, P.O. Box 59911, Abu Dhabi, United Arab Emirates
| | - Petrilla Jayaprakash
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates
| | - Shouq Aljneibi
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates
| | - Abdallah Alsaadi
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates
| | - Shaikha Alzaabi
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates
| | - Dorota Łazewska
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University-Medical College, Kraków, Poland
| | - Tadeusz Karcz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University-Medical College, Kraków, Poland
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University-Medical College, Kraków, Poland
| | - Bassem Sadek
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates; Zayed Center for Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates.
| |
Collapse
|
4
|
Li R, Anzai M, Shibata A, Ito-Ishida A. Synaptic disturbance in neurodevelopmental disorders: Perspectives from fragile X and Rett syndromes. Brain Dev 2025; 47:104358. [PMID: 40228442 DOI: 10.1016/j.braindev.2025.104358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025]
Abstract
Neurodevelopmental disorders (NDDs) are often referred to as "synaptopathies" because many of their behavioral symptoms arise from impaired synaptic development and function. However, the mechanisms that connect synaptic dysfunction to neurological symptoms remain unclear, mainly due to the wide variety of genetic and environmental factors involved in these disorders. Fragile X syndrome and Rett syndrome, two extensively studied monogenic NDDs, provide a unique opportunity to explore these mechanisms at molecular, cellular, and synaptic levels. This review summarizes the current understanding of how synaptic alterations contribute to the neurological symptoms observed in fragile X and Rett syndromes. A comparison of findings from mouse models indicates that an imbalance in local and distal connectivity may serve as a common feature of both disorders.
Collapse
Affiliation(s)
- Ruixiang Li
- Laboratory for Brain Development and Disorders, RIKEN Center for Brain Science, Saitama, Japan; Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Mai Anzai
- Laboratory for Brain Development and Disorders, RIKEN Center for Brain Science, Saitama, Japan
| | - Akiko Shibata
- Laboratory for Brain Development and Disorders, RIKEN Center for Brain Science, Saitama, Japan
| | - Aya Ito-Ishida
- Laboratory for Brain Development and Disorders, RIKEN Center for Brain Science, Saitama, Japan.
| |
Collapse
|
5
|
Zheng S, Chen C. Auditory processing deficits in autism spectrum disorder: mechanisms, animal models, and therapeutic directions. J Neural Transm (Vienna) 2025; 132:781-791. [PMID: 40353881 DOI: 10.1007/s00702-025-02919-x] [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: 11/19/2024] [Accepted: 03/24/2025] [Indexed: 05/14/2025]
Abstract
Auditory processing abnormalities are a prominent feature of Autism Spectrum Disorder (ASD), significantly affecting sensory integration, communication, and social interaction. This review delves into the neurobiological mechanisms underlying these deficits, including structural and functional disruptions in the auditory cortex, imbalances in excitatory and inhibitory signaling, and synaptic dysfunction. Genetic contributions from mutations in CNTNAP2, SHANK3, FMR1, and FOXP2 are explored, highlighting their roles in auditory abnormalities. Animal models, such as BTBRT+tf/J mice (BTBR) and valproic acid (VPA)-exposed rodents, provide critical insights into the sensory abnormalities observed in ASD. In addition, the review discusses current pharmacological strategies and emerging interventions targeting neurotransmitter systems and synaptic plasticity. Notably, future directions are emphasized, highlighting the need for integrated pharmacological and auditory-specific therapies to enhance sensory processing and communication outcomes in ASD. Overall, this review aims to bridge the gap between basic neurobiological research and clinical application, guiding future studies and therapeutic developments in ASD-related auditory processing deficits.
Collapse
Affiliation(s)
- Shuyu Zheng
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310051, Zhejiang, China
- Department of Traditional Chinese Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Chen Chen
- Department of Traditional Chinese Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
| |
Collapse
|
6
|
Wosnitzka E, Gambarotto L, Nikoletopoulou V. Macroautophagy at the service of synapses. Curr Opin Neurobiol 2025; 93:103054. [PMID: 40414166 DOI: 10.1016/j.conb.2025.103054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/02/2025] [Accepted: 05/02/2025] [Indexed: 05/27/2025]
Abstract
Post-mitotic and highly polarized neurons are dependent on the fitness of their synapses, which are often found a long distance away from the soma. How the synaptic proteome is maintained, dynamically reshaped, and continuously turned over is a topic of intense investigation. Autophagy, a highly conserved, lysosome-mediated degradation pathway has emerged as a vital component of long-term neuronal maintenance, and now more specifically of synaptic homeostasis. Here, we review the most recent findings on how autophagy undergoes both dynamic and local regulation at the synapse, and how it contributes to pre- and post-synaptic proteostasis and function. We also discuss the insights and open questions that this new evidence brings.
Collapse
Affiliation(s)
- Erin Wosnitzka
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005, Lausanne, Switzerland
| | - Lisa Gambarotto
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005, Lausanne, Switzerland
| | - Vassiliki Nikoletopoulou
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005, Lausanne, Switzerland.
| |
Collapse
|
7
|
Saleh NK, Farrag SM, El-Yamany MF, Kamel AS. Exploring Dapagliflozin's Influence on Autophagic Flux in Mania-like Behaviour: Insights from the LKB1/AMPK/LC3 Pathway in a Mouse Model. J Neuroimmune Pharmacol 2025; 20:57. [PMID: 40402300 PMCID: PMC12098488 DOI: 10.1007/s11481-025-10218-1] [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: 01/06/2025] [Accepted: 04/30/2025] [Indexed: 05/23/2025]
Abstract
Mania-like episodes are neuropsychiatric disturbances associated with bipolar disorder (BD). Autophagic flux disturbance evolved as one of the molecular mechanisms implicated in mania. Recently, Dapagliflozin (DAPA) has corrected autophagic signaling in several neurological disorders. Yet, no endeavours examined the autophagic impact of DAPA in mania-like behaviours. This study aimed to investigate the effect of DAPA on disrupted autophagic pathways in a mouse model of mania-like behaviour. Mania-like behaviour was induced through paradoxical sleep deprivation (PSD) using the multiple-platform method for a duration of 36 h. Mice were divided into three groups, with DAPA (1 mg/kg/day, orally) administered for one week. Behavioural assessments were conducted on the 7th day. DAPA mitigated anxiety-like behaviour in the open field test and improved motor coordination and muscle tone in the rotarod test. Mechanistically, DAPA activated hippocampal autophagy-related markers; liver kinase B1/AMP-activated protein kinase (LKB1/AMPK) pathway, autophagy related gene 7 (ATG7), and microtubule-associated protein light chain 3II (LC3II). This was associated with reduced levels of the autophagosome receptor p62 protein, which subsequently enhanced GABAA receptor-associated protein (GABARAP), facilitating the surface presentation of GABAA receptors. Additionally, DAPA upregulated the GABAB receptor R2 subunit through trophic factors such as brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF). Furthermore, DAPA mitigated elevated serum stress hormones and restored the balance between proinflammatory and anti-inflammatory cytokines in both cortical and hippocampal tissues. These findings highlight the role of autophagic flux modulation by DAPA and its therapeutic potential in mitigating mania-like behaviours.
Collapse
Affiliation(s)
- Nada K Saleh
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Sama M Farrag
- Pharmacology and Toxicology Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), Giza, Egypt
| | - Mohamed F El-Yamany
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Ahmed S Kamel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
8
|
Kaneda E, Koguchi-Yoshioka H, Nimura K, Hattori S, Ishino S, Fujimoto M, Wataya-Kaneda M. Genotype-phenotype correlation for skin and neuropsychiatric features in tuberous sclerosis complex. Br J Dermatol 2025; 192:1122-1124. [PMID: 39870113 DOI: 10.1093/bjd/ljaf038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/29/2025]
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant disorder caused by pathogenic variants in TSC1 and TSC2. The condition manifests as diverse symptoms across multiple organ systems and is frequently associated with neuropsychiatric symptoms. However, the relationship between the severity of these manifestations remains poorly understood. This study aimed to elucidate correlations between the severity of various skin and neuropsychiatric symptoms and the underlying TSC1/TSC2 pathology.
Collapse
Affiliation(s)
- Emi Kaneda
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hanako Koguchi-Yoshioka
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Neurocutaneous Medicine, Division of Health Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Keisuke Nimura
- Division of Gene Therapy Science, Department of Genome Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Gunma University Initiative for Advanced Research (GIAR), Gunma University, Maebashi, Gunma, Japan
| | - Satoshi Hattori
- Department of Biomedical Statistics, Graduate School of Medicine, Osaka University, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI), Osaka University, Osaka, Japan
| | - Saki Ishino
- CoMIT Omics Center, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mari Wataya-Kaneda
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Neurocutaneous Medicine, Division of Health Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
9
|
Shen L, Chen L, Tang Y, Yan Y, Xiong T, Liu Y, Li H, Gu H. PRRG4 Brain-Specific Conditional Knockout Mice Display Autism Spectrum Disorder-Like Behaviors. Biol Proced Online 2025; 27:16. [PMID: 40380139 DOI: 10.1186/s12575-025-00280-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized primarily by social deficits and repetitive behaviors. The mechanisms of ASD are complex and are not yet fully understood, although many ASD risk genes and mouse models have been reported. It has been suggested that deletion of PRRG4 (proline-rich and Gla domain 4) deletion may contribute to autism symptoms in patients with WAGR (Wilms' tumor, aniridia, gonadoblastoma, mental retardation) syndrome. The mouse model with PRRG4 gene deletion has not been reported so far. This study investigated whether brain-specific conditional knockout of PRRG4 induces ASD-like symptoms in mice by crossing the PRRG4fl/fl mice with Emx1-Cre mice, which express Cre in the cerebral cortex and hippocampus. RESULTS The PRRG4 brain-specific knockout (PRRG4fl/fl-Cre+, PRRG4-CKO) mice exhibited social deficits, repetitive behaviors, and anxiety-like symptoms compared to PRRG4fl/fl control mice according to the results of various behavioral tests. PRRG4 knockout led to the increase in total dendritic length, branching, and dendritic spine density in the pyramidal neurons of the cerebral cortex and hippocampus, as well as enhanced levels of synaptic proteins including SYP and PSD95. Immunoprecipitation experiment with PRRG4 antibodies showed dramatic decreased interaction of PRRG4 and MAGI2 proteins in brain tissues from PRRG4-CKO mice compared to PRRG4fl/fl control mice. GST-RBD pull-down assay showed a significant decrease in RhoA-GTP levels in the cerebral cortex and hippocampus of PRRG4-CKO mice. CONCLUSIONS Brain-specific conditional knockout of the PRRG4 in mice leads to ASD-like symptoms. PRRG4 protein may regulate dendritic and synaptic development in mice by activating RhoA through interaction with MAGI2. These findings provide evidence for a comprehensive understanding of PRRG4 function in vivo and support the association between PRRG4 loss and ASD phenotypes observed in WAGR syndrome.
Collapse
Affiliation(s)
- Luxi Shen
- Department of Internal Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Lan Chen
- Department of Laboratory Medicine, Ganzhou Municipal Hospital, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou, 341000, China
| | - Yuping Tang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan University Town, Northern Zhongxin Road, Wenzhou, Zhejiang, 325035, China
| | - Yeyao Yan
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan University Town, Northern Zhongxin Road, Wenzhou, Zhejiang, 325035, China
| | - Ting Xiong
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan University Town, Northern Zhongxin Road, Wenzhou, Zhejiang, 325035, China
| | - Yong Liu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan University Town, Northern Zhongxin Road, Wenzhou, Zhejiang, 325035, China
| | - Hongzhi Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan University Town, Northern Zhongxin Road, Wenzhou, Zhejiang, 325035, China.
| | - Haihua Gu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan University Town, Northern Zhongxin Road, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
10
|
Zhang Q, Zheng J, Sun H, Zheng J, Ma Y, Ji Q, Chen D, Tang Z, Zhang J, He Y, Song T. The Notch Signaling Pathway: A Potential Target for Mental Disorders. Mol Neurobiol 2025:10.1007/s12035-025-05034-w. [PMID: 40372672 DOI: 10.1007/s12035-025-05034-w] [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: 08/04/2024] [Accepted: 05/02/2025] [Indexed: 05/16/2025]
Abstract
The highly conserved Notch signaling pathway plays a critical role in cell fate determination during metazoan development through cell-to-cell communication. The classical pathway consists of Notch receptors, ligands, intracellular effectors, DNA-binding proteins, and other regulatory molecules. Recent research has highlighted its involvement in the pathogenesis of several diseases. In autism, bipolar disorder, and schizophrenia, the Notch signaling pathway is implicated in key processes such as neuronal development and synaptic plasticity. Furthermore, it has been shown to play significant roles in other mental health conditions, including anxiety, depression, post-traumatic stress disorder, and neurocognitive disorders. However, the precise mechanisms underlying the contribution of Notch to these conditions remain poorly understood. This review examines the current understanding of the Notch signaling pathway in mental disorders, highlighting its role in their pathophysiology and summarizing therapeutic strategies aimed at modulating this pathway to improve mental health outcomes.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Jingxuan Zheng
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hongqin Sun
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Jishan Zheng
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Yunyan Ma
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Qinglu Ji
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Dengwang Chen
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Zhengzhen Tang
- Department of Pediatrics, Third Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jidong Zhang
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Yuqi He
- School of Pharmacy, Zunyi Medical University, Zunyi, China
- Engineering Research Center of Key Technologies for Industrial Development of Dendrobium in Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Tao Song
- Department of Immunology, Zunyi Medical University, Zunyi, China.
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
11
|
Si Y, Zhang H, Du L, Deng Z. Abnormalities of brain dynamics based on large-scale cortical network modeling in autism spectrum disorder. Neural Netw 2025; 189:107561. [PMID: 40388872 DOI: 10.1016/j.neunet.2025.107561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/12/2025] [Accepted: 04/27/2025] [Indexed: 05/21/2025]
Abstract
Synaptic increase is a common phenomenon in the brain of autism spectrum disorder (ASD). However, the impact of increased synapses on the neurophysiological activity of ASD remains unclear. To address this, we propose a large-scale cortical network model based on empirical structural connectivity data using the Wendling model, which successfully simulates both pathological and physiological electroencephalography (EEG) signals. Building on this, the EEG functional network is constructed using the phase lag index, effectively characterizing the functional connectivity. Our modeling results indicate that EEG activity and functional network properties undergo significant changes by globally increasing synaptic coupling strength. Specifically, it leads to abnormal neural oscillations clinically reported in ASD, including the decreased dominant frequency, the decreased relative power in the α band and the increased relative power in the δ+θ band, particularly in the frontal lobe. At the same time, the clustering coefficient and global efficiency of the functional network decrease, while the characteristic path length increases, suggesting that the functional network of ASD is inefficient and poorly integrated. Additionally, we find insufficient functional connectivity across multiple brain regions in ASD, along with decreased wavelet coherence in the α band within the frontal lobe and between the frontal and temporal lobes. Considering that most of the synaptic increases in ASD are limited, brain regions are further randomly selected to increase the local synaptic coupling strength. The results show that disturbances in local brain regions can also facilitate the development of ASD. This study reveals the intrinsic link between synapse increase and abnormal brain activity in ASD, and inspires treatments related to synapse pruning.
Collapse
Affiliation(s)
- Youyou Si
- School of Mathematics and Statistics, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; MIIT Key Laboratory of Dynamics and Control of Complex Systems, Xi'an, Shaanxi, 710072, China
| | - Honghui Zhang
- School of Mathematics and Statistics, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; MIIT Key Laboratory of Dynamics and Control of Complex Systems, Xi'an, Shaanxi, 710072, China.
| | - Lin Du
- School of Mathematics and Statistics, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; MIIT Key Laboratory of Dynamics and Control of Complex Systems, Xi'an, Shaanxi, 710072, China
| | - Zichen Deng
- School of Aeronautics, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; MIIT Key Laboratory of Dynamics and Control of Complex Systems, Xi'an, Shaanxi, 710072, China
| |
Collapse
|
12
|
Yang L, Guo C, Zheng Z, Dong Y, Xie Q, Lv Z, Li M, Lu Y, Guo X, Deng R, Liu Y, Feng Y, Mu R, Zhang X, Ma H, Chen Z, Zhang Z, Dong Z, Yang W, Zhang X, Cui Y. Stress dynamically modulates neuronal autophagy to gate depression onset. Nature 2025; 641:427-437. [PMID: 40205038 PMCID: PMC12058529 DOI: 10.1038/s41586-025-08807-4] [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: 07/24/2023] [Accepted: 02/19/2025] [Indexed: 04/11/2025]
Abstract
Chronic stress remodels brain homeostasis, in which persistent change leads to depressive disorders1. As a key modulator of brain homeostasis2, it remains elusive whether and how brain autophagy is engaged in stress dynamics. Here we discover that acute stress activates, whereas chronic stress suppresses, autophagy mainly in the lateral habenula (LHb). Systemic administration of distinct antidepressant drugs similarly restores autophagy function in the LHb, suggesting LHb autophagy as a common antidepressant target. Genetic ablation of LHb neuronal autophagy promotes stress susceptibility, whereas enhancing LHb autophagy exerts rapid antidepressant-like effects. LHb autophagy controls neuronal excitability, synaptic transmission and plasticity by means of on-demand degradation of glutamate receptors. Collectively, this study shows a causal role of LHb autophagy in maintaining emotional homeostasis against stress. Disrupted LHb autophagy is implicated in the maladaptation to chronic stress, and its reversal by autophagy enhancers provides a new antidepressant strategy.
Collapse
Affiliation(s)
- Liang Yang
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Chen Guo
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Zhiwei Zheng
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yiyan Dong
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qifeng Xie
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Zijian Lv
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Min Li
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yangyang Lu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Xiaonan Guo
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Rongshan Deng
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiqin Liu
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yirong Feng
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruiqi Mu
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Xuliang Zhang
- Laboratory Animal Center, Zhejiang University, Hangzhou, China
| | - Huan Ma
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Zhong Chen
- Zhejiang Key Laboratory of Neuropsychopharmacology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhijun Zhang
- Department of Neurology, Affiliated Zhongda Hospital, School of Medicine, Institute of Neuropsychiatry, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
- Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhaoqi Dong
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wei Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangnan Zhang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
| | - Yihui Cui
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
13
|
Vellucci L, Barone A, Buonaguro EF, Ciccarelli M, De Simone G, Iannotta F, Matrone M, Mazza B, Vitelli R, de Bartolomeis A, Iasevoli F. Severity of autism-related symptoms in treatment-resistant schizophrenia: associations with cognitive performance, psychosocial functioning, and neurological soft signs - Clinical evidence and ROC analysis. J Psychiatr Res 2025; 185:119-129. [PMID: 40179689 DOI: 10.1016/j.jpsychires.2025.03.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/19/2025] [Accepted: 03/22/2025] [Indexed: 04/05/2025]
Abstract
Treatment-resistant schizophrenia (TRS) occurs when symptoms persist despite adequate antipsychotic treatment in terms of both timing and dosage. This severe condition is often overlooked, despite the existence of guidelines, with an average delay of 4-9 years before the introduction of clozapine, the gold standard treatment. We hypothesized that schizophrenia patients with severe autistic symptoms are more prone to develop TRS. To test this, we administered the Positive and Negative Syndrome Scale for Schizophrenia Autism Severity Scale (PAUSS) to 117 patients diagnosed with schizophrenia. Our results revealed that both TRS and clozapine non-responder (CLZ-nR) groups had higher rates of autistic symptoms than non-TRS patients. A machine learning model was developed to examine the relationship between PAUSS scores and TRS, obtaining an accuracy of 0.65 and an AUC of 0.67. Specifically, PAUSS items N6 ("lack of spontaneity and flow of conversation") and N7 ("stereotypical thinking") emerged as the most significant factors in the model. In addition, PAUSS was correlated with cognitive and social functions, as well as soft neurological signs, in TRS patients. Autism-related symptoms were found to predict significant variance in motor coordination, verbal fluency, functional ability and soft neurological signs. These results suggest that autism-related symptoms in schizophrenia may define a distinct subgroup with unique neurobiological characteristics.
Collapse
Affiliation(s)
- Licia Vellucci
- Unit for Treatment-Resistant Psychoses, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry - University of Naples "Federico II", Naples, Italy; Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Annarita Barone
- Unit for Treatment-Resistant Psychoses, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry - University of Naples "Federico II", Naples, Italy
| | - Elisabetta Filomena Buonaguro
- Unit for Treatment-Resistant Psychoses, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry - University of Naples "Federico II", Naples, Italy; Unità Operativa di Salute Mentale Terzigno, ASL NAPOLI 3 SUD, Naples, Italy
| | - Mariateresa Ciccarelli
- Unit for Treatment-Resistant Psychoses, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry - University of Naples "Federico II", Naples, Italy
| | - Giuseppe De Simone
- Unit for Treatment-Resistant Psychoses, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry - University of Naples "Federico II", Naples, Italy
| | - Federica Iannotta
- Unit for Treatment-Resistant Psychoses, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry - University of Naples "Federico II", Naples, Italy
| | - Marta Matrone
- NESMOS (Neurosciences, Mental Health, and Sensory Organs) Department, Sapienza University of Rome, Faculty of Medicine and Psychology, Via di Grottarossa 1035-1039, 00189, Rome, Italy; Department of Mental Health Protection and Promotion, Unit of Addiction Pathology, Via Salaria per Roma, 36, 02100, Rieti, Italy
| | - Benedetta Mazza
- Unit for Treatment-Resistant Psychoses, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry - University of Naples "Federico II", Naples, Italy
| | - Roberto Vitelli
- Unit for Treatment-Resistant Psychoses, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry - University of Naples "Federico II", Naples, Italy
| | - Andrea de Bartolomeis
- Unit for Treatment-Resistant Psychoses, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry - University of Naples "Federico II", Naples, Italy; UNESCO Staff Chair on Health Education and Sustainable Development, University "Federico II", Naples, Italy.
| | - Felice Iasevoli
- Unit for Treatment-Resistant Psychoses, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry - University of Naples "Federico II", Naples, Italy
| |
Collapse
|
14
|
Li YZ, Gao L, Sun XL, Duan L, Jiang M, Wu QF. Neural cell competition sculpting brain from cradle to grave. Natl Sci Rev 2025; 12:nwaf057. [PMID: 40309342 PMCID: PMC12042753 DOI: 10.1093/nsr/nwaf057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/18/2025] [Accepted: 02/13/2025] [Indexed: 05/02/2025] Open
Abstract
Darwinian selection, operating within the cellular ecosystem of multicellular organisms, drives a pervasive surveillance mechanism of cell-cell competition that shapes tissue architecture and function. While cell competition eliminates suboptimal cells to ensure tissue integrity across various tissues, neuronal competition specifically sculpts neural networks to establish precise circuits for sensory, motor and cognitive functions. However, our understanding of cell competition across diverse neural cell types in both developmental and pathological contexts remains limited. Here, we review recent advances on the phenomenon, and mechanisms and potential functions of neural cell competition (NCC), ranging from neural progenitors, neurons, astrocytes and oligodendrocytes to microglia. Physiological NCC governs cellular survival, proliferation, arborization, organization, function and territorial colonization, whereas dysregulated NCC may cause neurodevelopmental disorders, accelerate aging, exacerbate neurodegenerative diseases and drive brain tumor progression. Future work that leverages cell competition mechanisms may help to improve cognition and curb diseases.
Collapse
Affiliation(s)
- Yu Zheng Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lisen Gao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xue-Lian Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Lihui Duan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Man Jiang
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qing-Feng Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Children's Hospital, Beijing 100045, China
| |
Collapse
|
15
|
Tang H, Ma T, Wang Y, Zhang C, Chu Y, Guo Y, Xi J, Jiao D, Li B, Xie C, Wang Y. Paeoniflorin modulates AGEs/RAGE/P38MAPK/ERK/mTOR autophagy pathway to improve cognitive dysfunction in MRL/lpr mice. Int J Biol Macromol 2025; 307:141765. [PMID: 40049494 DOI: 10.1016/j.ijbiomac.2025.141765] [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: 08/26/2024] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
OBJECTIVE The objective of this study was to investigate the therapeutic effects of paeoniflorin (PA) on cognitive impairment and to elucidate its potential mechanisms in MRL/lpr mice, a model of systemic lupus erythematosus-associated cognitive dysfunction. METHOD Cognitive performance and behavioral responses were assessed using a comprehensive battery of tests, including the Morris water maze, the Novel object recognition test, and the Y maze. Neuropathological changes in the hippocampal regions were visualized through Nissl, HE and Immunohistochemistry staining. Protein expression levels of receptor for advanced glycation end-products (RAGE) and LC3B were quantified by immunofluorescence, while the ultrastructure of autophagic organelles was examined using transmission electron microscopy (TEM). Inflammatory cytokines, namely tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6) were quantified in both serum and hippocampal homogenates by enzyme-linked immunosorbent assay (ELISA). The hippocampal expression of advanced glycation end-products (AGEs), RAGE, p62, Beclin-1, and key proteins involved in the mitogen-activated protein kinase (MAPK) pathways, including p38MAPK, ERK, and mTOR were analyzed by Western blotting. RESULT Paeoniflorin ameliorates cognitive dysfunction, neuronal damage, pro-inflammatory cytokine production in MRL/lpr mice. Paeoniflorin suppresses RAGE and autophagy levels and P38 MAPK/ERK/mTOR signaling pathway activation in the hippocampus of MRL/lpr mice. CONCLUSION Paeoniflorin may exert its neuroprotective effects by modulating the AGEs/RAGE/P38MAPK/ERK/mTOR autophagy signaling pathway.
Collapse
Affiliation(s)
- Honghui Tang
- School of Clinical Medicine, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Tianzhen Ma
- Department of Embryology, Bengbu Medical University, Bengbu, Anhui 233030, China; Anhui Key Laboratory of Tissue Transplantation, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China
| | - Yanxin Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Chuanmeng Zhang
- School of Mental Health, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Yuanding Chu
- School of Clinical Medicine, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Yuqing Guo
- School of Clinical Medicine, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Jin Xi
- Bengbu Medical University Research Center, Bengbu, Anhui 233030, China; Anhui Key Laboratory of Tissue Transplantation, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China
| | - Dongliang Jiao
- School of Mental Health, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Baiqing Li
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China
| | - Changhao Xie
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China; Anhui Province Key Laboratory of Immunology in Chronic Diseases, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, 287 Changhuai Road, Bengbu, Anhui 233004, China.
| | - Yuanyuan Wang
- Department of Embryology, Bengbu Medical University, Bengbu, Anhui 233030, China; Anhui Key Laboratory of Tissue Transplantation, 2600 Donghai Avenue, Longzihu District, Bengbu, Anhui 233030, China.
| |
Collapse
|
16
|
Zuo Z, Zhang H, Li Z, Qi F, Hu H, Yang J, Yao Z. Activation of Hippocampal Neuronal NADPH Oxidase NOX2 Promotes Depressive-Like Behaviour and Cognition Deficits in Chronic Restraint Stress Mouse Model. PHARMACOPSYCHIATRY 2025; 58:117-126. [PMID: 39547705 DOI: 10.1055/a-2429-4023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
BACKGROUND Nicotinamide adenosine dinucleotide phosphate oxidases (NOX) play important roles in mediating stress-induced depression. Three NOX isotypes are expressed mainly in the brain: NOX2, NOX3 and NOX4. In this study, the expression and cellular sources of these NOX isoforms was investigated in the context of stress-induced depression. METHODS Chronic restraint stress (CRS)-induced depressive-like behaviour and cognitive deficits were evaluated by tail suspension tests, forced swimming tests and the Morris water maze test. Hippocampal NOX expression was determined by immunofluorescence staining and western blotting. The hippocampal levels of the brain-derived neurotrophic factor (BDNF) mRNA were determined via quantitative real-time -polymerase chain reaction. Glucocorticoid levels in the hippocampus were measured using ELISA kits. RESULTS In the mouse CRS model, a significant increase in NOX2 expression was observed in the hippocampus, whereas no significant changes in NOX3 and NOX4 expression were detected. Next, NOX2 expression was primarily localised to neurons (NeuN+) but not microglia (Iba-1+) or astrocytes (GFAP+). Treatment with gp91ds-tat, a specific NOX2 inhibitor, effectively mitigated the behavioural deficits induced by CRS. The decreased expression of the BDNF mRNA in the hippocampus of CRS mice was restored upon gp91ds-tat treatment. A positive correlation was identified between neuronal NOX2 expression and serum glucocorticoid levels. CONCLUSIONS Our study indicated that neuronal NOX2 may be a critical mediator of depression-like behaviours and spatial cognitive deficits in mice subjected to CRS. Blockade of NOX2 signalling may be a promising therapeutic strategy for depression.
Collapse
Affiliation(s)
- Zejie Zuo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongyang Zhang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhihui Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Fangfang Qi
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Haojie Hu
- Department of Psychology, College of Arts and Sciences, New York University, NY, USA
| | - Junhua Yang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhibin Yao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
17
|
Cantua R, Mulligan K. Developmental neurotoxicity of bisphenol F and bisphenol S in animal model systems: A literature review. Neurotoxicology 2025; 108:263-280. [PMID: 40280242 DOI: 10.1016/j.neuro.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Neurodevelopmental disorders have complex etiologies, stemming both from genetic and environmental risk factors, including gestational exposure to bisphenol A (BPA). BPA is an endocrine-disrupting chemical widely used in the synthesis of plastics and epoxy-resins. In 2012, the Food and Drug Administration issued a ban on the use of BPA in certain baby and childhood products, which contributed to the proliferation of BPA-free products. To make products without BPA, plastic and epoxy manufacturers often use chemical analogs, including bisphenol F (BPF) and bisphenol S (BPS). However, the structural and biochemical similarities BPF and BPS share with BPA suggest they may have similar molecular and cellular impacts on the developing nervous system, despite consumers generally regarding BPA-free products as safer alternatives. In this review, we synthesized all available peer-reviewed primary literature to date reporting on the neurodevelopmental impacts of BPF and/or BPS in animal models. In total, 61 papers were identified as relevant to the topic, including evaluation of BPF- and BPS-associated neurodevelopmental phenotypes such as changes in neurodevelopmental gene expression, the proliferation and differentiation of neural stem cells, synaptogenesis, central nervous system morphology, neuronal cell death, and behavior. Though less extensively studied than BPA, the collective works described here indicate that BPF and BPS can act as developmental neurotoxicants in animal models, urging further mechanistic and epidemiological analyses of these bisphenol analogs, as well as a reconsideration by regulatory agencies of policies surrounding their usage.
Collapse
Affiliation(s)
- Ricardo Cantua
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA 95819, USA.
| | - Kimberly Mulligan
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA 95819, USA.
| |
Collapse
|
18
|
Wei R, Wang Y, Fang H, Guan L, Gao J, Xu X, Ke X, Jin H. Distinct features of EEG microstates in autism spectrum disorder revealed by meta-analysis: the contribution of individual age to heterogeneity across studies. Front Psychiatry 2025; 16:1531694. [PMID: 40330653 PMCID: PMC12052564 DOI: 10.3389/fpsyt.2025.1531694] [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: 11/21/2024] [Accepted: 03/28/2025] [Indexed: 05/08/2025] Open
Abstract
Background and purpose Electroencephalographic (EEG) microstates, as quasi-stable scalp EEG spatial patterns, are characterized by their high temporal resolution, making them a potentially powerful approach for studying the function of large-scale brain networks. A substantial body of research has demonstrated that abnormalities in the function or structure of large-scale brain networks are closely related to many characteristics of autism spectrum disorder (ASD). Investigating the EEG microstate features of individuals with autism can help reveal the nature of autism. To date, numerous studies have observed unique resting-state microstate patterns in individuals with autism. However, the results of these studies have not been consistent. Therefore, the present study aims to assess the differences in microstate parameters between ASD and non-autistic groups through meta-analysis and to explore the sources of research heterogeneity. Method This meta-analysis was preregistered with PROSPERO (CRD42024599897) and followed PRISMA guidelines. Studies in English comparing EEG microstate patterns between ASD and Non-autistic groups were retrieved by database search to October 20, 2024. The meta-analysis was then conducted using RevMan5.2. Pooled results are expressed as standardized mean difference (SMD). Heterogeneity (I²) and publication bias were assessed using Stata15.0. Result Seven studies enrolling 194 ASD individuals were included, four deemed high quality and three moderate quality according to bias risk assessment. Microstate B duration and coverage were significantly greater in the pooled ASD group (duration SMD=0.83, 95%CI: 0.17-1.5; coverage SMD=0.54, 95%CI: 0.18-0.90), but heterogeneity could not be excluded. Microstate C occurrence frequency was also in the ASD group (SMD= -0.61, 95%CI: -1.08 to -0.15), and heterogeneity was significant. Sensitivity analysis revealed that only the group difference in microstate B coverage was robust. Subgroup analysis suggested that age was the main source of heterogeneity in microstate B and C coverage. Results were not affected by publication bias according to Egger's test. Conclusion Future studies on the EEG microstate characteristics of ASD must control for age as an important cofounding variable. Systematic Review Registration PROSPERO, identifier CRD42024599897.
Collapse
Affiliation(s)
- Ran Wei
- Children's Mental Health Research Center of the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Child Healthcare Department of the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Child Healthcare Department of the Suzhou Maternal and Child Health Hospital, Suzhou, China
| | - Yonglu Wang
- Children's Mental Health Research Center of the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Hui Fang
- Children's Mental Health Research Center of the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Luyang Guan
- Children's Mental Health Research Center of the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jianxing Gao
- Children's Mental Health Research Center of the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyue Xu
- Children's Mental Health Research Center of the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoyan Ke
- Children's Mental Health Research Center of the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Hua Jin
- Child Healthcare Department of the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Child Healthcare Department of the Suzhou Maternal and Child Health Hospital, Suzhou, China
| |
Collapse
|
19
|
Thakur RS, O'Connor-Giles KM. PDZD8 promotes autophagy at ER-lysosome membrane contact sites to regulate activity-dependent synaptic growth. Cell Rep 2025; 44:115483. [PMID: 40156832 DOI: 10.1016/j.celrep.2025.115483] [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: 08/05/2024] [Revised: 02/07/2025] [Accepted: 03/07/2025] [Indexed: 04/01/2025] Open
Abstract
Building synaptic connections requires coordinating a host of cellular activities from cell signaling to protein turnover, placing a high demand on intracellular communication. Membrane contact sites (MCSs) formed between organelles have emerged as key signaling hubs for coordinating diverse cellular activities, yet their roles in the developing nervous system remain obscure. We investigate the in vivo function of the endoplasmic reticulum (ER) MCS tethering and lipid-transfer protein PDZD8, which was recently linked to intellectual disability, in the nervous system. We find that PDZD8 is required for activity-dependent synaptic bouton formation in multiple paradigms. PDZD8 is sufficient to drive excess synaptic bouton formation through an autophagy-dependent mechanism and required for synapse development when autophagy is limited. PDZD8 accelerates autophagic flux by promoting lysosome maturation at ER-late endosome/lysosome MCSs. We propose that PDZD8 functions in the nervous system to increase autophagy during periods of high demand, including activity-dependent synaptic growth.
Collapse
Affiliation(s)
- Rajan S Thakur
- Department of Neuroscience, Brown University, Providence, RI, USA.
| | - Kate M O'Connor-Giles
- Department of Neuroscience, Brown University, Providence, RI, USA; Carney Institute for Brain Science, Providence, RI, USA.
| |
Collapse
|
20
|
Zhang H, Meléndez A. Conserved components of the macroautophagy machinery in Caenorhabditis elegans. Genetics 2025; 229:iyaf007. [PMID: 40180610 PMCID: PMC12005284 DOI: 10.1093/genetics/iyaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 12/13/2024] [Indexed: 04/05/2025] Open
Abstract
Macroautophagy involves the sequestration of cytoplasmic contents in a double-membrane autophagosome and its subsequent delivery to lysosomes for degradation and recycling. In Caenorhabditis elegans, autophagy participates in diverse processes such as stress resistance, cell fate specification, tissue remodeling, aging, and adaptive immunity. Genetic screens in C. elegans have identified a set of metazoan-specific autophagy genes that form the basis for our molecular understanding of steps unique to the autophagy pathway in multicellular organisms. Suppressor screens have uncovered multiple mechanisms that modulate autophagy activity under physiological conditions. C. elegans also provides a model to investigate how autophagy activity is coordinately controlled at an organismal level. In this chapter, we will discuss the molecular machinery, regulation, and physiological functions of autophagy, and also methods utilized for monitoring autophagy during C. elegans development.
Collapse
Affiliation(s)
- Hong Zhang
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Alicia Meléndez
- Department of Biology, Queens College, City University of New York, Flushing, NY 11367, USA
- Molecular, Cellular and Developmental Biology and Biochemistry Ph.D. Programs, The Graduate Center of the City University of New York, New York, NY 10016, USA
| |
Collapse
|
21
|
Abromeit A, Hooijmans CR, LeMaoult C, Drion CM, Kas M. Animal studies reveal downregulation of the Beclin-1 autophagy pathway as shared mechanism in Autism Spectrum Disorder: a systematic review and meta-analysis. Mol Psychiatry 2025:10.1038/s41380-025-03028-7. [PMID: 40247126 DOI: 10.1038/s41380-025-03028-7] [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: 01/11/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND Autism Spectrum Disorder (ASD) is a heterogeneous neurodevelopmental condition with complex etiology, involving genetic and environmental influences on brain development and behavior. Dysregulation of mammalian target of rapamycin (mTOR) signaling alters neuronal growth and synaptic plasticity, and has emerged as a potential underlying pathway in ASD. GOAL AND METHODS To investigate mTOR dysregulation as a common mechanism in ASD, we performed a systematic review, and a meta-analysis of 192 studies examining mTOR signaling in diverse genetic and environmental animal models. RESULTS Our random-effects model identified significant alterations in mTOR pathway-related proteins. For several proteins (p-AKT, PTEN, p-mTOR, p-EIF4e, LC3-II, p-S6K and p-S6), subgroup analyses revealed clear species-, sex-, age-, or brain region-specific effects. Interestingly, Beclin-1 was consistently downregulated across all subgroups. CONCLUSION Our findings support mTOR-pathway dysregulation in ASD. The observed consistent downregulation of Beclin-1 highlights autophagy as a common mechanism, and provides new leads for novel ASD biomarker and treatment development.
Collapse
Affiliation(s)
- A Abromeit
- Faculty of Science and Engineering, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - C R Hooijmans
- Department of Anaesthesiology, Pain and Palliative Care, Radboud University Medical Center, Nijmegen, The Netherlands
| | - C LeMaoult
- Faculty of Science and Engineering, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - C M Drion
- Faculty of Science and Engineering, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands.
| | - Mjh Kas
- Faculty of Science and Engineering, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
22
|
Yang J, Li X, Tan J, Zhou P, Hu L, Chen J, Li T, Liu Y, Chen L. Prenatal Exposure To Valproic Acid Induces Increased Autism-Like Behaviors and Impairment of Learning and Memory Functions in Rat Offspring by Upregulating ADAM10 Expression. Neurochem Res 2025; 50:146. [PMID: 40240730 PMCID: PMC12003443 DOI: 10.1007/s11064-025-04398-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Autism spectrum disorder (ASD) involves a complex neurodevelopmental pathogenesis. A disintegrin and metalloproteinase 10 (ADAM10) plays a crucial role in embryonic brain development and neural network stability. This study aimed to investigate the influence of ADAM10 on excitation/inhibition (E/I) balance, autism-like behaviors, and learning and memory dysfunction in rats prenatally exposed to valproic acid (VPA) and determine potential intervention strategies. The VPA-exposed group exhibited increased levels of ADAM10 and secreted amyloid precursor protein-α (sAPPα). Moreover, overexpression of glutamate decarboxylase 1 and N-methyl-D-aspartate receptors was observed. High-performance liquid chromatography-mass spectrometry revealed elevated levels of glutamate, glutamine, and γ-aminobutyric acid, as well as an E/I imbalance in the VPA group. Additionally, narrower synaptic clefts as well as increased postsynaptic density and synaptic vesicles were observed. Remarkably, intraperitoneal administration of ADAM10 inhibitor during the critical period of synaptic development significantly improved ASD-like behavior and learning and memory function in VPA-exposed rats. This intervention effectively reduced abnormally high sAPPα levels in the prefrontal cortex and corrected abnormal E/I balance. Thus, inhibiting ADAM10 overexpression may improve the E/I imbalance, alleviate core symptoms of ASD, and improve learning and memory dysfunction. The use of ADAM10 inhibitor represents a potential therapeutic strategy for treating ASD patients with intellectual disabilities.
Collapse
Affiliation(s)
- Jingyuan Yang
- Growth, Development and Mental Health Center of Children and Adolescents, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- Yibin Hospital Affiliated to Children's Hospital of Chongqing Medical University, Yibin, 644000, China
| | - Xiaoli Li
- Growth, Development and Mental Health Center of Children and Adolescents, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Jing Tan
- Growth, Development and Mental Health Center of Children and Adolescents, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Ping Zhou
- Department of Anatomy and Developmental Biology, Kyoto University Graduate School of Medicine, Yoshidakonoe, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Lingjun Hu
- Growth, Development and Mental Health Center of Children and Adolescents, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Jie Chen
- Growth, Development and Mental Health Center of Children and Adolescents, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Tingyu Li
- Growth, Development and Mental Health Center of Children and Adolescents, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yonggang Liu
- Laboratory of stem cell and Tissue Engineering, Chongqing Medical University, Chongqing, 400010, China
| | - Li Chen
- Growth, Development and Mental Health Center of Children and Adolescents, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
23
|
Teter OM, McQuade A, Hagan V, Liang W, Dräger NM, Sattler SM, Holmes BB, Castillo VC, Papakis V, Leng K, Boggess S, Nowakowski TJ, Wells J, Kampmann M. CRISPRi-based screen of autism spectrum disorder risk genes in microglia uncovers roles of ADNP in microglia endocytosis and synaptic pruning. Mol Psychiatry 2025:10.1038/s41380-025-02997-z. [PMID: 40188316 DOI: 10.1038/s41380-025-02997-z] [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: 06/01/2024] [Revised: 03/06/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
Autism Spectrum Disorders (ASD) are a set of neurodevelopmental disorders with complex biology. The identification of ASD risk genes from exome-wide association studies and de novo variation analyses has enabled mechanistic investigations into how ASD-risk genes alter development. Most functional genomics studies have focused on the role of these genes in neurons and neural progenitor cells. However, roles for ASD risk genes in other cell types are largely uncharacterized. There is evidence from postmortem tissue that microglia, the resident immune cells of the brain, appear activated in ASD. Here, we used CRISPRi-based functional genomics to systematically assess the impact of ASD risk gene knockdown on microglia activation and phagocytosis. We developed an iPSC-derived microglia-neuron coculture system and high-throughput flow cytometry readout for synaptic pruning to enable parallel CRISPRi-based screening of phagocytosis of beads, synaptosomes, and synaptic pruning. Our screen identified ADNP, a high-confidence ASD risk genes, as a modifier of microglial synaptic pruning. We found that microglia with ADNP loss have altered endocytic trafficking, remodeled proteomes, and increased motility in coculture.
Collapse
Affiliation(s)
- Olivia M Teter
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, San Francisco, San Francisco, CA, USA
| | - Amanda McQuade
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Venus Hagan
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Weiwei Liang
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Nina M Dräger
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Sydney M Sattler
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Brandon B Holmes
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Vincent Cele Castillo
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Vasileios Papakis
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Kun Leng
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA
| | - Steven Boggess
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Tomasz J Nowakowski
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, 94158, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - James Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
24
|
Karpova A, Hiesinger PR, Kuijpers M, Albrecht A, Kirstein J, Andres-Alonso M, Biermeier A, Eickholt BJ, Mikhaylova M, Maglione M, Montenegro-Venegas C, Sigrist SJ, Gundelfinger ED, Haucke V, Kreutz MR. Neuronal autophagy in the control of synapse function. Neuron 2025; 113:974-990. [PMID: 40010347 DOI: 10.1016/j.neuron.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/24/2024] [Accepted: 01/24/2025] [Indexed: 02/28/2025]
Abstract
Neurons are long-lived postmitotic cells that capitalize on autophagy to remove toxic or defective proteins and organelles to maintain neurotransmission and the integrity of their functional proteome. Mutations in autophagy genes cause congenital diseases, sharing prominent brain dysfunctions including epilepsy, intellectual disability, and neurodegeneration. Ablation of core autophagy genes in neurons or glia disrupts normal behavior, leading to motor deficits, memory impairment, altered sociability, and epilepsy, which are associated with defects in synapse maturation, plasticity, and neurotransmitter release. In spite of the importance of autophagy for brain physiology, the substrates of neuronal autophagy and the mechanisms by which defects in autophagy affect synaptic function in health and disease remain controversial. Here, we summarize the current state of knowledge on neuronal autophagy, address the existing controversies and inconsistencies in the field, and provide a roadmap for future research on the role of autophagy in the control of synaptic function.
Collapse
Affiliation(s)
- Anna Karpova
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - P Robin Hiesinger
- Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Marijn Kuijpers
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands; Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Anne Albrecht
- Institute of Anatomy, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto-von-Guericke-University, 39120 Magdeburg, Germany; German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Germany
| | - Janine Kirstein
- Leibniz Institute on Aging-Fritz-Lipmann-Institute, 07754 Jena, Germany; Friedrich-Schiller-Universität, Institute for Biochemistry & Biophysics, 07745 Jena, Germany
| | - Maria Andres-Alonso
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany; Leibniz Group "Dendritic Organelles and Synaptic Function", Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | | | - Britta J Eickholt
- Institute of Molecular Biology and Biochemistry, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Marina Mikhaylova
- Institute of Biology, Humboldt Universität zu Berlin, 10115 Berlin, Germany
| | - Marta Maglione
- Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Carolina Montenegro-Venegas
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany; Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Stephan J Sigrist
- Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany; Institute of Molecular Biology and Biochemistry, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Eckart D Gundelfinger
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto-von-Guericke-University, 39120 Magdeburg, Germany; Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Volker Haucke
- Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany; Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin, Germany; Institute of Molecular Biology and Biochemistry, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | - Michael R Kreutz
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto-von-Guericke-University, 39120 Magdeburg, Germany; Leibniz Group "Dendritic Organelles and Synaptic Function", Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; German Center for Neurodegenerative Diseases (DZNE), Site Magdeburg, 39120 Magdeburg, Germany.
| |
Collapse
|
25
|
Matuskey D, Yang Y, Naganawa M, Koohsari S, Toyonaga T, Gravel P, Pittman B, Torres K, Pisani L, Finn C, Cramer-Benjamin S, Herman N, Rosenthal LH, Franke CJ, Walicki BM, Esterlis I, Skosnik P, Radhakrishnan R, Wolf JM, Nabulsi N, Ropchan J, Huang Y, Carson RE, Naples AJ, McPartland JC. 11C-UCB-J PET imaging is consistent with lower synaptic density in autistic adults. Mol Psychiatry 2025; 30:1610-1616. [PMID: 39367053 DOI: 10.1038/s41380-024-02776-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Abstract
The neural bases of autism are poorly understood at the molecular level, but evidence from animal models, genetics, post-mortem studies, and single-gene disorders implicate synaptopathology. Here, we use positron emission tomography (PET) to assess the density of synapses with synaptic vesicle glycoprotein 2A (SV2A) in autistic adults using 11C-UCB-J. Twelve autistic (mean (SD) age 25 (4) years; six males), and twenty demographically matched non-autistic individuals (26 (3) years; eleven males) participated in a 11C-UCB-J PET scan. Binding potential, BPND, was the primary outcome measure and computed with the centrum semiovale as the reference region. Partial volume correction with Iterative Yang was applied to control for possible volumetric differences. Mixed-model statistics were calculated for between-group differences. Relationships to clinical characteristics were evaluated based on clinician ratings of autistic features. Whole cortex synaptic density was 17% lower in the autism group (p = 0.01). All brain regions in autism had lower 11C-UCB-J BPND compared to non-autistic participants. This effect was evident in all brain regions implicated in autism. Significant differences were observed across multiple individual regions, including the prefrontal cortex (-15%, p = 0.02), with differences most pronounced in gray matter (p < 0.0001). Synaptic density was significantly associated with clinical measures across the whole cortex (r = 0.67, p = 0.02) and multiple regions (rs = -0.58 to -0.82, ps = 0.05 to <0.01). The first in vivo investigation of synaptic density in autism with PET reveals pervasive and large-scale lower density in the cortex and across multiple brain areas. Synaptic density also correlated with clinical features, such that a greater number of autistic features were associated with lower synaptic density. These results indicate that brain-wide synaptic density may represent an as-yet-undiscovered molecular basis for the clinical phenotype of autism and associated pervasive alterations across a diversity of neural processes.
Collapse
Affiliation(s)
- David Matuskey
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA.
- Department of Psychiatry, Yale University, New Haven, CT, USA.
- Department of Neurology, Yale University, New Haven, CT, USA.
- Center for Brain and Mind Health, Yale University, New Haven, CT, USA.
| | - Yanghong Yang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Mika Naganawa
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Sheida Koohsari
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Paul Gravel
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Brian Pittman
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Kristen Torres
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Lauren Pisani
- Child Study Center, Yale University, New Haven, CT, USA
| | - Caroline Finn
- Child Study Center, Yale University, New Haven, CT, USA
| | | | - Nicole Herman
- Child Study Center, Yale University, New Haven, CT, USA
| | | | | | | | - Irina Esterlis
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Patrick Skosnik
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Rajiv Radhakrishnan
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Julie M Wolf
- Child Study Center, Yale University, New Haven, CT, USA
| | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Jim Ropchan
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Adam J Naples
- Center for Brain and Mind Health, Yale University, New Haven, CT, USA
- Child Study Center, Yale University, New Haven, CT, USA
| | - James C McPartland
- Center for Brain and Mind Health, Yale University, New Haven, CT, USA.
- Child Study Center, Yale University, New Haven, CT, USA.
| |
Collapse
|
26
|
Deri E, Kumar Ojha S, Kartawy M, Khaliulin I, Amal H. Multi-omics study reveals differential expression and phosphorylation of autophagy-related proteins in autism spectrum disorder. Sci Rep 2025; 15:10878. [PMID: 40158064 PMCID: PMC11954894 DOI: 10.1038/s41598-025-95860-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Our multi-omics study investigated the molecular mechanisms underlying autism spectrum disorder (ASD) using Shank3Δ4-22 and Cntnap2-/- mouse models. Through global- and phospho- proteomics of the mouse cortex, we focused on shared molecular changes and found that autophagy was particularly affected in both models. Global proteomics identified a small number of differentially expressed proteins that significantly impact postsynaptic components and synaptic function, including key pathways such as mTOR signaling. Phosphoproteomics revealed unique phosphorylation sites in autophagy-related proteins such as ULK2, RB1CC1, ATG16L1, and ATG9, suggesting that altered phosphorylation patterns contribute to impaired autophagic flux in ASD. SH-SY5Y cells with SHANK3 gene deletion showed elevated LC3-II and p62 levels, indicating autophagosome accumulation and autophagy initiation, while the reduced level of the lysosomal activity marker LAMP1 suggested impaired autophagosome-lysosome fusion. The study highlights the involvement of reactive nitrogen species and nitric oxide (NO) on autophagy disruption. Importantly, inhibition of neuronal NO synthase (nNOS) by 7-NI normalized autophagy markers levels in the SH-SY5Y cells and primary cultured neurons. We have previously shown that nNOS inhibition improved synaptic and behavioral phenotypes in Shank3Δ4-22 and Cntnap2-/- mouse models. Our multi-omics study reveals differential expression and phosphorylation of autophagy-related proteins in ASD but further investigation is needed to prove the full involvement of autophagy in ASD. Our study underscores the need for further examination into the functional consequences of the identified phosphorylation sites, which may offer potential novel therapeutic autophagy-related targets for ASD treatment.
Collapse
Affiliation(s)
- Eden Deri
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shashank Kumar Ojha
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA.
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
27
|
Barnes SA, Thomazeau A, Finnie PSB, Heinrich MJ, Heynen AJ, Komiyama NH, Grant SGN, Menniti FS, Osterweil EK, Bear MF. Non-ionotropic signaling through the NMDA receptor GluN2B carboxy-terminal domain drives dendritic spine plasticity and reverses fragile X phenotypes. Cell Rep 2025; 44:115311. [PMID: 39983718 PMCID: PMC12006837 DOI: 10.1016/j.celrep.2025.115311] [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: 07/12/2024] [Revised: 12/03/2024] [Accepted: 01/23/2025] [Indexed: 02/23/2025] Open
Abstract
N-methyl-D-aspartate (NMDA)-induced spine shrinkage proceeds independently of ion flux and requires the initiation of de novo protein synthesis. Using subtype-selective pharmacological and genetic tools, we find that structural plasticity is dependent on ligand binding to GluN2B-containing NMDA receptors (NMDARs) and signaling via the GluN2B carboxy-terminal domain (CTD). Disruption of non-ionotropic signaling by replacing the GluN2B CTD with the GluN2A CTD leads to an increase in spine density, dysregulated basal protein synthesis, exaggerated long-term depression mediated by G-protein-coupled metabotropic glutamate receptors (mGluR-LTD), and epileptiform activity reminiscent of phenotypes observed in the Fmr1 knockout (KO) model of fragile X syndrome. By crossing the Fmr1 KO mice with animals in which the GluN2A CTD has been replaced with the GluN2B CTD, we observe a correction of these core fragile X phenotypes. These findings suggest that non-ionotropic NMDAR signaling through GluN2B may represent a novel therapeutic target for the treatment of fragile X and related causes of intellectual disability and autism.
Collapse
Affiliation(s)
- Stephanie A Barnes
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Aurore Thomazeau
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Peter S B Finnie
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Maxwell J Heinrich
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arnold J Heynen
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Noburu H Komiyama
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK; The Patrick Wild Centre, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Seth G N Grant
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Frank S Menniti
- MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
| | - Emily K Osterweil
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; F.M. Kirby Center for Neurobiology, Translational Neuroscience Center, Department of Neurology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115, USA
| | - Mark F Bear
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
28
|
De Introna M, Krashia P, Sabetta A, La Barbera L, Nobili A, D'Amelio M, Cecconi F, Ammassari-Teule M, Pignataro A. Chemogenetic induction of CA1 hyperexcitability triggers indistinguishable autistic traits in asymptomatic mice differing in Ambra1 expression and sex. Transl Psychiatry 2025; 15:82. [PMID: 40097399 PMCID: PMC11914586 DOI: 10.1038/s41398-025-03271-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 12/18/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
Among the genomic alterations identified as risk factors in mice models of autism spectrum disorders (ASD), heterozygous deletion of Ambra1 (Activating Molecule in Beclin1-Regulated Autophagy) triggers an ASD phenotype associated with hippocampal hyperexcitability exclusively in the female sex although Ambra1 protein is comparably expressed in the hippocampus of symptomatic females and asymptomatic males. Given the intricate relationship between Ambra1 deficiency and sex in the etiology of ASD, we took advantage of asymptomatic mice including Ambra1+/- males and wild-type (Wt) mice of both sexes to investigate whether their non-pathogenic variations in Ambra1 levels could underlie a differential susceptibility to exhibit ASD-like traits in response to experimental elevation of hippocampal excitability. Here we report that selective activation of inhibitory DREADD in CA1 parvalbumin-positive interneurons (PV-IN) reduces GABAergic currents onto pyramidal neurons (PN), causes social and attentional deficits, and augments the proportion of immature/thin spines in CA1 PN dendrites to the same extent in Ambra1+/- males and Wt mice of both sexes. Our findings show that the substantial hippocampal variations in pro-autophagic Ambra1 gene product shown by asymptomatic mice differing in mutation and/or sex do not underlie a differential reactivity to chemogenetic induction of idiopathic ASD.
Collapse
Affiliation(s)
- Margherita De Introna
- IRCCS Santa Lucia Foundation, Centro Europeo di Ricerca sul Cervello CERC, Rome, Italy
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Paraskevi Krashia
- IRCCS Santa Lucia Foundation, Centro Europeo di Ricerca sul Cervello CERC, Rome, Italy
- Università Campus Bio-Medico di Roma, Rome, Italy
| | - Annamaria Sabetta
- IRCCS Santa Lucia Foundation, Centro Europeo di Ricerca sul Cervello CERC, Rome, Italy
- Dipartimento di Medicina Traslazionale e di Precisione, Sapienza Università di Roma, Rome, Italy
| | - Livia La Barbera
- IRCCS Santa Lucia Foundation, Centro Europeo di Ricerca sul Cervello CERC, Rome, Italy
- Università Campus Bio-Medico di Roma, Rome, Italy
| | - Annalisa Nobili
- IRCCS Santa Lucia Foundation, Centro Europeo di Ricerca sul Cervello CERC, Rome, Italy
- Università Campus Bio-Medico di Roma, Rome, Italy
| | - Marcello D'Amelio
- IRCCS Santa Lucia Foundation, Centro Europeo di Ricerca sul Cervello CERC, Rome, Italy
- Università Campus Bio-Medico di Roma, Rome, Italy
| | - Francesco Cecconi
- Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Annabella Pignataro
- IRCCS Santa Lucia Foundation, Centro Europeo di Ricerca sul Cervello CERC, Rome, Italy.
- Institute of Translational Pharmacology, National Research Council, CNR, Rome, Italy.
| |
Collapse
|
29
|
Alcaíno JM, Vera G, Almarza G, Lagos CF, Terraza CA, Del Campo A, Recabarren-Gajardo G. Novel 5-HT 6R modulators as mTOR-dependent neuronal autophagy inductors. Sci Rep 2025; 15:8380. [PMID: 40069248 PMCID: PMC11897353 DOI: 10.1038/s41598-025-92755-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Autophagy is a natural process in which the cell degrades substances through the lysosomal pathway. One of the most studied mechanisms for regulating autophagy is the mTOR signaling pathway. Recent research has shown that the 5-HT6 receptor is linked to the mTOR pathway and can affect cognition in various neurodevelopmental models. Therefore, developing 5-HT6 receptor antagonists could improve cognition by inducing autophagy through the inhibition of the mTOR pathway. Our study reports two in-house-designed 5-HT6R antagonists, PUC-10 and its indazole analogue PUC-55, that induce mTOR-dependent autophagy. PUC-10, an indole-based 5-HT6 receptor antagonist with high binding affinity (Ki = 14.6 nM) and antagonist potency (IC50 = 32 nM), demonstrated more than 90% at 25 µM cellular viability and a high capacity to induce autophagy in the neuroblastoma SH-SY5Y cell line. Similarly, its indazole analogue, PUC-55 (Ki = 37.5 nM), exhibited high cellular viability and potent autophagy-inducing activity. Both compounds induced overexpression of the 5-HT6 receptor after 24 h of stimulation, contrasting with the effects observed with Rapamycin (100 nM), a well-known mTOR inhibitor. Additionally, the signaling pathway was characterized, showing that both PUC-10 and PUC-55 induce autophagy by inhibiting the mTOR pathway, suggesting their potential therapeutic applications for neurological disorders.
Collapse
Affiliation(s)
- José Miguel Alcaíno
- Laboratorio de Fisiología y Bioenergética Celular, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, 7810000, Chile
| | - Gonzalo Vera
- Bioactive Heterocycles Synthesis Laboratory (BHSL), Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Avenida Vicuña Mackenna, Macul, Santiago, 4860, 7820436, Chile
| | - Gonzalo Almarza
- Laboratorio de Fisiología y Bioenergética Celular, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, 7810000, Chile
| | - Carlos F Lagos
- Chemical Biology & Drug Discovery Lab, Escuela de Química y Farmacia, Facultad de Medicina y Ciencia, Universidad San Sebastián, Campus Los Leones, Providencia, Lota, Santiago, 2465, 7510157, Chile
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Av. del Valle Norte 725, Huechuraba, Santiago, 8580702, Chile
| | - Claudio A Terraza
- Research Laboratory for Organic Polymers (RLOP), Faculty of Chemistry and of Pharmacy, Pontificia Universidad Católica de Chile, Post 22, P.O. Box. 306, Santiago, Chile
| | - Andrea Del Campo
- Laboratorio de Fisiología y Bioenergética Celular, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, 7810000, Chile.
| | - Gonzalo Recabarren-Gajardo
- Bioactive Heterocycles Synthesis Laboratory (BHSL), Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Avenida Vicuña Mackenna, Macul, Santiago, 4860, 7820436, Chile.
- Centro Interdisciplinario de Neurociencias, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, 8330024, Chile.
| |
Collapse
|
30
|
Shin YS, Christensen D, Wang J, Shirley DJ, Orlando AM, Romero RA, Vaillancourt DE, Wilkes BJ, Coombes SA, Wang Z. Transcallosal white matter and cortical gray matter variations in autistic adults aged 30-73 years. Mol Autism 2025; 16:16. [PMID: 40050930 PMCID: PMC11884179 DOI: 10.1186/s13229-025-00652-6] [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: 07/18/2024] [Accepted: 02/17/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a lifelong condition that profoundly impacts health, independence, and quality of life. However, research on brain aging in autistic adults is limited, and microstructural variations in white and gray matter remain poorly understood. To address this critical gap, we assessed novel diffusion MRI (dMRI) biomarkers, free water, and free water corrected fractional anisotropy (fwcFA), and mean diffusivity (fwcMD) across 32 transcallosal tracts and their corresponding homotopic grey matter origin/endpoint regions of interest (ROIs) in middle and old aged autistic adults. METHODS Forty-three autistic adults aged 30-73 and 43 age-, sex-, and IQ-matched neurotypical controls underwent dMRI scans. We examined free water, fwcFA, fwcMD differences between the two groups and age-related pattern of each dMRI metric across the whole brain for each group. The relationships between clinical measures of ASD and free water in regions that significantly differentiated autistic adults from neurotypical controls were also explored. In supplementary analyses, we also assessed free water uncorrected FA and MD using conventional single tensor modeling. RESULTS Autistic adults exhibited significantly elevated free water in seven frontal transcallosal tracts compared to controls. In controls, age-related increases in free water and decreases in fwcFA were observed across most transcallosal tracts. However, these age-associated patterns were entirely absent in autistic adults. In gray matter, autistic adults showed elevated free water in the calcarine cortices and lower fwcMD in the dorsal premotor cortices compared to controls. Lastly, age-related increases in free water were found across all white matter and gray matter ROIs in neurotypical controls, whereas no age-related associations were detected in any dMRI metrics for autistic adults. LIMITATIONS We only recruited cognitively capable autistic adults, which limits the generalizability of our findings across the full autism spectrum. The cross-sectional design precludes inferences about microstructural changes over time in middle and old aged autistic adults. CONCLUSIONS Our findings revealed increased free water load in frontal white matter in autistic adults and identified distinct age-associated microstructural variations between the two groups. These findings highlight more heterogeneous brain aging profiles in autistic adults. Our study also demonstrated the importance of quantifying free water in dMRI studies of ASD.
Collapse
Affiliation(s)
- Young Seon Shin
- Laboratory for Rehabilitation Neuroscience, Department of Applied Physiology and Kinesiology, University of Florida, PO Box 118206, Gainesville, FL, 32611-8205, USA
| | - Danielle Christensen
- Neurocognitive and Behavioral Development Laboratory, Department of Applied Physiology and Kinesiology, University of Florida, PO Box 118206, Gainesville, FL, 32611-8205, USA
| | - Jingying Wang
- Neurocognitive and Behavioral Development Laboratory, Department of Applied Physiology and Kinesiology, University of Florida, PO Box 118206, Gainesville, FL, 32611-8205, USA
| | - Desirae J Shirley
- Neurocognitive and Behavioral Development Laboratory, Department of Applied Physiology and Kinesiology, University of Florida, PO Box 118206, Gainesville, FL, 32611-8205, USA
| | - Ann-Marie Orlando
- Center for Autism and Related Disabilities (CARD), University of Florida, Gainesville, FL, 32606, USA
- UF Health Center for Autism and Neurodevelopment (UF Health CAN), University of Florida, Gainesville, FL, 32606, USA
- Department of Psychiatry, University of Florida, Gainesville, FL, 32606, USA
| | - Regilda A Romero
- UF Health Center for Autism and Neurodevelopment (UF Health CAN), University of Florida, Gainesville, FL, 32606, USA
- Department of Psychiatry, University of Florida, Gainesville, FL, 32606, USA
| | - David E Vaillancourt
- Laboratory for Rehabilitation Neuroscience, Department of Applied Physiology and Kinesiology, University of Florida, PO Box 118206, Gainesville, FL, 32611-8205, USA
- Department of Neurology and McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
- Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Bradley J Wilkes
- Laboratory for Rehabilitation Neuroscience, Department of Applied Physiology and Kinesiology, University of Florida, PO Box 118206, Gainesville, FL, 32611-8205, USA
| | - Stephen A Coombes
- Laboratory for Rehabilitation Neuroscience, Department of Applied Physiology and Kinesiology, University of Florida, PO Box 118206, Gainesville, FL, 32611-8205, USA
- Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Zheng Wang
- Neurocognitive and Behavioral Development Laboratory, Department of Applied Physiology and Kinesiology, University of Florida, PO Box 118206, Gainesville, FL, 32611-8205, USA.
- University of Florida, PO Box 118205, Gainesville, FL, 32611-8205, USA.
| |
Collapse
|
31
|
Pagani M, Zerbi V, Gini S, Alvino F, Banerjee A, Barberis A, Basson MA, Bozzi Y, Galbusera A, Ellegood J, Fagiolini M, Lerch J, Matteoli M, Montani C, Pozzi D, Provenzano G, Scattoni ML, Wenderoth N, Xu T, Lombardo M, Milham MP, Martino AD, Gozzi A. Biological subtyping of autism via cross-species fMRI. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.04.641400. [PMID: 40093106 PMCID: PMC11908180 DOI: 10.1101/2025.03.04.641400] [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: 03/19/2025]
Abstract
It is frequently assumed that the phenotypic heterogeneity in autism spectrum disorder reflects underlying pathobiological variation. However, direct evidence in support of this hypothesis is lacking. Here, we leverage cross-species functional neuroimaging to examine whether variability in brain functional connectivity reflects distinct biological mechanisms. We find that fMRI connectivity alterations in 20 distinct mouse models of autism (n=549 individual mice) can be clustered into two prominent hypo- and hyperconnectivity subtypes. We show that these connectivity profiles are linked to distinct signaling pathways, with hypoconnectivity being associated with synaptic dysfunction, and hyperconnectivity reflecting transcriptional and immune-related alterations. Extending these findings to humans, we identify analogous hypo- and hyperconnectivity subtypes in a large, multicenter resting state fMRI dataset of n=940 autistic and n=1036 neurotypical individuals. Remarkably, hypo- and hyperconnectivity autism subtypes are replicable across independent cohorts (accounting for 25.1% of all autism data), exhibit distinct functional network architecture, are behaviorally dissociable, and recapitulate synaptic and immune mechanisms identified in corresponding mouse subtypes. Our cross-species investigation, thus, decodes the heterogeneity of fMRI connectivity in autism into distinct pathway-specific etiologies, offering a new empirical framework for targeted subtyping of autism.
Collapse
Affiliation(s)
- Marco Pagani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
- Autism Center, Child Mind Institute, New York, NY, USA
- IMT School for Advanced Studies, Lucca, Italy
| | - Valerio Zerbi
- Department of Psychiatry, University of Geneva, Switzerland
- Department of Basic Neurosciences, University of Geneva, Switzerland
| | - Silvia Gini
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
- Center for Mind and Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
| | - Filomena Alvino
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | | | - Andrea Barberis
- Synaptic Plasticity of Inhibitory Networks, Istituto Italiano di Tecnologia, Genova, Italy
| | - M. Albert Basson
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| | - Yuri Bozzi
- Center for Mind and Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
| | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Jacob Ellegood
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
| | | | - Jason Lerch
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Michela Matteoli
- Humanitas University, Milan, Italy
- CNR Institute of Neuroscience c/o Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Caterina Montani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Davide Pozzi
- CNR Institute of Neuroscience c/o Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Giovanni Provenzano
- Department of Cellular, Computational and Integrative Biology. University of Trento, Trento, Italy
| | - Maria Luisa Scattoni
- Research Coordination and Support Service, Istituto Superiore di Sanità, Rome, Italy
| | | | - Ting Xu
- Center for Integrative Developing Brain, Child Mind Institute, New York, NY, USA
| | - Michael Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy
| | - Michael P Milham
- Center for the Integrative Developmental Neuroscience, Child Mind Institute, New York, NY, USA
| | | | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| |
Collapse
|
32
|
Tan B, Hedbacker K, Kelly L, Zhang Z, Moura-Assis A, Luo JD, Rabinowitz JD, Friedman JM. A cellular and molecular basis of leptin resistance. Cell Metab 2025; 37:723-741.e6. [PMID: 40043692 DOI: 10.1016/j.cmet.2025.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 09/09/2024] [Accepted: 01/02/2025] [Indexed: 05/13/2025]
Abstract
Similar to most humans with obesity, diet-induced obese (DIO) mice have high leptin levels and fail to respond to the exogenous hormone, suggesting that their obesity is caused by leptin resistance, the pathogenesis of which is unknown. We found that leptin treatment reduced plasma levels of leucine and methionine, mTOR-activating ligands, leading us to hypothesize that chronic mTOR activation might reduce leptin signaling. Rapamycin, an mTOR inhibitor, reduced fat mass and increased leptin sensitivity in DIO mice but not in mice with defects in leptin signaling. Rapamycin restored leptin's actions on POMC neurons and failed to reduce the weight of mice with defects in melanocortin signaling. mTOR activation in POMC neurons caused leptin resistance, whereas POMC-specific mutations in mTOR activators decreased weight gain of DIO mice. Thus, increased mTOR activity in POMC neurons is necessary and sufficient for the development of leptin resistance in DIO mice, establishing a key pathogenic mechanism leading to obesity.
Collapse
Affiliation(s)
- Bowen Tan
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Kristina Hedbacker
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Leah Kelly
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Zhaoyue Zhang
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Alexandre Moura-Assis
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Ji-Dung Luo
- Bioinformatics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Joshua D Rabinowitz
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Jeffrey M Friedman
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
33
|
Kaladiyil AP, Kukkle PL. Bassoon, Presynaptic Scaffolding Protein: Narrative Review in Health and Disease. Eur J Neurosci 2025; 61:e70066. [PMID: 40074701 DOI: 10.1111/ejn.70066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/31/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
The release of synaptic vesicles (SVs) at the synaptic junction is a complex process involving various specialized proteins that work in unison. Among these, Bassoon has emerged as a significant protein, particularly noted for its association with various neurological and aging-related diseases. Due to its structural and functional roles, Bassoon has become a focus of recent research, especially in understanding its implications in neurodegenerative and psychiatric disorders. In this narrative review, we explore Bassoon's structure, function, and its role across a spectrum of neurological disorders. Neurotransmission is a tightly regulated process that relies on specialized structures within the presynaptic terminal, such as the presynaptic active zone (AZ), to precisely control SV release in response to incoming signals. The AZ comprises a complex network of large, multidomain proteins, with Bassoon playing a crucial role in this arrangement. Bassoon facilitates the tethering and reloading of SVs, ensuring responsiveness to high-frequency signals, while also maintaining proteostasis at the presynapse. This involves orchestrating the localization of proteins essential for neuronal development and plasticity. Bassoon's large size and unique structural features enable it to interact with and regulate the function of multiple proteins, making it integral to presynaptic functioning. Variants in the Bassoon gene have been linked to a variety of neurodegenerative and psychiatric conditions, including Progressive Supranuclear Palsy, multiple system atrophy (MSA), epilepsy, schizophrenia, bipolar disorder, and Parkinson's disease. This review delves into Bassoon's pivotal role in preserving presynaptic integrity and how disruptions in its functions may contribute to these disorders.
Collapse
Affiliation(s)
- Ahamed P Kaladiyil
- Center for Brain Research, Indian Institute of Science, Bangalore, India
- Parkinson's Disease and Movement Disorders Clinic, Bangalore, India
| | | |
Collapse
|
34
|
James D, Lam VT, Jo B, Fung LK. Sex Differences in the Relationship Between Cortical Thickness and Sensory Motor Symptoms in Adults on the Autism Spectrum. Behav Neurol 2025; 2025:2951294. [PMID: 40041925 PMCID: PMC11879536 DOI: 10.1155/bn/2951294] [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: 01/18/2024] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 05/12/2025] Open
Abstract
Background: Autism spectrum disorder (ASD) involves alterations in both cortical morphology and sensory processing. These structural and perceptual changes may lie on a continuum with typically developing (TD) individuals. However, investigations on possible links between these two factors are lacking, and it remains to be seen if their relationship differs by sex. We hypothesized that cortical thickness in the postcentral gyrus (a somatosensory processing hub) would correlate with sensory processing symptoms in a combined cohort of autistic and TD individuals. We also hypothesized that these correlations would differ based on sex. Methods: We studied 23 autistic adults and 27 TD adults using magnetic resonance imaging to measure the cortical thickness of the postcentral gyrus and the Ritvo Autism Asperger Diagnostic Scale-Revised (RAADS-R) to measure autism characteristics, with a particular focus on the sensory motor subscale. Results: The left postcentral gyrus (PCG) was found to be thicker in the autism group than in the TD group (d = 0.946, p = 0.003), particularly in autistic males compared to TD males and TD females. The RAADS-R sensory motor subscale and bilateral PCG cortical thickness were positively correlated across both autistic and TD males (Spearman's rho = 0.481, p = 0.008) but not females. These correlations were specific to the sensory motor subscale, as no correlations were found for RAADS-R total score or any of the other subscales. Conclusions: These results demonstrate sex-specific differences in the relationship between cortical thickness at the PCG and sensory processing in autistic individuals and that these differences exist along a continuum that extends into the TD population. Our findings contribute to furthering our understanding of sex-specific neuroanatomical differences in people on the autism spectrum. The left PCG thickness could be a potential sex-specific biomarker for sensorimotor function that is generally applicable in both neurotypical and autism populations. With further validations, this biomarker could be used to track responses to interventions targeting sensorimotor challenges in people on the autism spectrum.
Collapse
Affiliation(s)
- David James
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, California, USA
| | - Vicky T. Lam
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, California, USA
| | - Booil Jo
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, California, USA
| | - Lawrence K. Fung
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, California, USA
| |
Collapse
|
35
|
Lazzeri G, Lenzi P, Signorini G, Raffaelli S, Giammattei E, Natale G, Ruffoli R, Fornai F, Ferrucci M. Retinoic Acid Promotes Neuronal Differentiation While Increasing Proteins and Organelles Related to Autophagy. Int J Mol Sci 2025; 26:1691. [PMID: 40004155 PMCID: PMC11855701 DOI: 10.3390/ijms26041691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/12/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
Retinoic acid (RA) is commonly used to differentiate SH-SY5Y neuroblastoma cells. This effect is sustained by a specific modulation of gene transcription, leading to marked changes in cellular proteins. In this scenario, autophagy may be pivotal in balancing protein synthesis and degradation. The present study analyzes whether some autophagy-related proteins and organelles are modified during RA-induced differentiation of SH-SY5Y cells. RA-induced effects were compared to those induced by starvation. SH-SY5Y cells were treated with a single dose of 10 µM RA or grown in starvation, for 3 days or 7 days. After treatments, cells were analyzed at light microscopy and transmission electron microscopy to assess cell morphology and immunostaining for specific markers (nestin, βIII-tubulin, NeuN) and some autophagy-related proteins (Beclin 1, LC3). We found that both RA and starvation differentiate SH-SY5Y cells. Specifically, cell differentiation was concomitant with an increase in autophagy proteins and autophagy-related organelles. However, the effects of a single dose of 10 μM RA persist for at least 7 days, while prolonged starvation produces cell degeneration and cell loss. Remarkably, the effects of RA are modulated in the presence of autophagy inhibitors or stimulators. The present data indicate that RA-induced differentiation is concomitant with an increased autophagy.
Collapse
Affiliation(s)
- Gloria Lazzeri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (G.L.); (P.L.); (G.S.); (S.R.); (E.G.); (G.N.); (R.R.); (F.F.)
| | - Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (G.L.); (P.L.); (G.S.); (S.R.); (E.G.); (G.N.); (R.R.); (F.F.)
| | - Giulia Signorini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (G.L.); (P.L.); (G.S.); (S.R.); (E.G.); (G.N.); (R.R.); (F.F.)
| | - Sara Raffaelli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (G.L.); (P.L.); (G.S.); (S.R.); (E.G.); (G.N.); (R.R.); (F.F.)
| | - Elisa Giammattei
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (G.L.); (P.L.); (G.S.); (S.R.); (E.G.); (G.N.); (R.R.); (F.F.)
| | - Gianfranco Natale
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (G.L.); (P.L.); (G.S.); (S.R.); (E.G.); (G.N.); (R.R.); (F.F.)
| | - Riccardo Ruffoli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (G.L.); (P.L.); (G.S.); (S.R.); (E.G.); (G.N.); (R.R.); (F.F.)
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (G.L.); (P.L.); (G.S.); (S.R.); (E.G.); (G.N.); (R.R.); (F.F.)
- IRCCS-Istituto di Ricovero e Cura a Carattere Scientifico, Neuromed, 86077 Pozzilli, Italy
| | - Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (G.L.); (P.L.); (G.S.); (S.R.); (E.G.); (G.N.); (R.R.); (F.F.)
| |
Collapse
|
36
|
Smith EM, Coughlan ML, Maday S. Turning garbage into gold: Autophagy in synaptic function. Curr Opin Neurobiol 2025; 90:102937. [PMID: 39667255 PMCID: PMC11903044 DOI: 10.1016/j.conb.2024.102937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 10/26/2024] [Accepted: 11/13/2024] [Indexed: 12/14/2024]
Abstract
Trillions of synapses in the human brain enable thought and behavior. Synaptic connections must be established and maintained, while retaining dynamic flexibility to respond to experiences. These processes require active remodeling of the synapse to control the composition and integrity of proteins and organelles. Macroautophagy (hereafter, autophagy) provides a mechanism to edit and prune the synaptic proteome. Canonically, autophagy has been viewed as a homeostatic process, which eliminates aged and damaged proteins to maintain neuronal survival. However, accumulating evidence suggests that autophagy also degrades specific cargoes in response to neuronal activity to impact neuronal transmission, excitability, and synaptic plasticity. Here, we will discuss the diverse roles, regulation, and mechanisms of neuronal autophagy in synaptic function and contributions from glial autophagy in these processes.
Collapse
Affiliation(s)
- Erin Marie Smith
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maeve Louise Coughlan
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sandra Maday
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
37
|
Khaliulin I, Hamoudi W, Amal H. The multifaceted role of mitochondria in autism spectrum disorder. Mol Psychiatry 2025; 30:629-650. [PMID: 39223276 PMCID: PMC11753362 DOI: 10.1038/s41380-024-02725-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: 03/10/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Normal brain functioning relies on high aerobic energy production provided by mitochondria. Failure to supply a sufficient amount of energy, seen in different brain disorders, including autism spectrum disorder (ASD), may have a significant negative impact on brain development and support of different brain functions. Mitochondrial dysfunction, manifested in the abnormal activities of the electron transport chain and impaired energy metabolism, greatly contributes to ASD. The aberrant functioning of this organelle is of such high importance that ASD has been proposed as a mitochondrial disease. It should be noted that aerobic energy production is not the only function of the mitochondria. In particular, these organelles are involved in the regulation of Ca2+ homeostasis, different mechanisms of programmed cell death, autophagy, and reactive oxygen and nitrogen species (ROS and RNS) production. Several syndromes originated from mitochondria-related mutations display ASD phenotype. Abnormalities in Ca2+ handling and ATP production in the brain mitochondria affect synaptic transmission, plasticity, and synaptic development, contributing to ASD. ROS and Ca2+ regulate the activity of the mitochondrial permeability transition pore (mPTP). The prolonged opening of this pore affects the redox state of the mitochondria, impairs oxidative phosphorylation, and activates apoptosis, ultimately leading to cell death. A dysregulation between the enhanced mitochondria-related processes of apoptosis and the inhibited autophagy leads to the accumulation of toxic products in the brains of individuals with ASD. Although many mitochondria-related mechanisms still have to be investigated, and whether they are the cause or consequence of this disorder is still unknown, the accumulating data show that the breakdown of any of the mitochondrial functions may contribute to abnormal brain development leading to ASD. In this review, we discuss the multifaceted role of mitochondria in ASD from the various aspects of neuroscience.
Collapse
Affiliation(s)
- Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
38
|
Ham A, Chang AY, Li H, Bain JM, Goldman JE, Sulzer D, Veenstra-VanderWeele J, Tang G. Impaired macroautophagy confers substantial risk for intellectual disability in children with autism spectrum disorders. Mol Psychiatry 2025; 30:810-824. [PMID: 39237724 DOI: 10.1038/s41380-024-02741-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Autism spectrum disorder (ASD) represents a complex of neurological and developmental disabilities characterized by clinical and genetic heterogeneity. While the causes of ASD are still unknown, many ASD risk factors are found to converge on intracellular quality control mechanisms that are essential for cellular homeostasis, including the autophagy-lysosomal degradation pathway. Studies have reported impaired autophagy in ASD human brain and ASD-like synapse pathology and behaviors in mouse models of brain autophagy deficiency, highlighting an essential role for defective autophagy in ASD pathogenesis. To determine whether altered autophagy in the brain may also occur in peripheral cells that might provide useful biomarkers, we assessed activities of autophagy in lympoblasts from ASD and control subjects. We find that lymphoblast autophagy is compromised in a subset of ASD participants due to impaired autophagy induction. Similar changes in autophagy are detected in postmortem human brains from ASD individuals and in brain and peripheral blood mononuclear cells from syndromic ASD mouse models. Remarkably, we find a strong correlation between impaired autophagy and intellectual disability in ASD participants. By depleting the key autophagy gene Atg7 from different brain cells, we provide further evidence that autophagy deficiency causes cognitive impairment in mice. Together, our findings suggest autophagy dysfunction as a convergent mechanism that can be detected in peripheral blood cells from a subset of autistic individuals, and that lymphoblast autophagy may serve as a biomarker to stratify ASD patients for the development of targeted interventions.
Collapse
Affiliation(s)
- Ahrom Ham
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Audrey Yuen Chang
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Hongyu Li
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jennifer M Bain
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - James E Goldman
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - David Sulzer
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Pharmacology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Jeremy Veenstra-VanderWeele
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, 10032, USA
- New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Guomei Tang
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
39
|
Mishra AK, Tripathi MK, Kumar D, Gupta SP. Neurons Specialize in Presynaptic Autophagy: A Perspective to Ameliorate Neurodegeneration. Mol Neurobiol 2025; 62:2626-2640. [PMID: 39141193 DOI: 10.1007/s12035-024-04399-8] [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: 05/06/2024] [Accepted: 07/24/2024] [Indexed: 08/15/2024]
Abstract
The efficient and prolonged neurotransmission is reliant on the coordinated action of numerous synaptic proteins in the presynaptic compartment that remodels synaptic vesicles for neurotransmitter packaging and facilitates their exocytosis. Once a cycle of neurotransmission is completed, membranes and associated proteins are endocytosed into the cytoplasm for recycling or degradation. Both exocytosis and endocytosis are closely regulated in a timely and spatially constrained manner. Recent research demonstrated the impact of dysfunctional synaptic vesicle retrieval in causing retrograde degeneration of midbrain neurons and has highlighted the importance of such endocytic proteins, including auxilin, synaptojanin1 (SJ1), and endophilin A (EndoA) in neurodegenerative diseases. Additionally, the role of other associated proteins, including leucine-rich repeat kinase 2 (LRRK2), adaptor proteins, and retromer proteins, is being investigated for their roles in regulating synaptic vesicle recycling. Research suggests that the degradation of defective vesicles via presynaptic autophagy, followed by their recycling, not only revitalizes them in the active zone but also contributes to strengthening synaptic plasticity. The presynaptic autophagy rejuvenating terminals and maintaining neuroplasticity is unique in autophagosome formation. It involves several synaptic proteins to support autophagosome construction in tiny compartments and their retrograde trafficking toward the cell bodies. Despite having a comprehensive understanding of ATG proteins in autophagy, we still lack a framework to explain how autophagy is triggered and potentiated in compact presynaptic compartments. Here, we reviewed synaptic proteins' involvement in forming presynaptic autophagosomes and in retrograde trafficking of terminal cargos. The review also discusses the status of endocytic proteins and endocytosis-regulating proteins in neurodegenerative diseases and strategies to combat neurodegeneration.
Collapse
Affiliation(s)
- Abhishek Kumar Mishra
- Department of Zoology, Government Shaheed Gendsingh College, Charama, Uttar Bastar Kanker, 494 337, Chhattisgarh, India.
| | - Manish Kumar Tripathi
- School of Pharmacy, Faculty of Medicine, Institute for Drug Research, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Dipak Kumar
- Department of Zoology, Munger University, Munger, Bihar, India
| | - Satya Prakash Gupta
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221 005, India
| |
Collapse
|
40
|
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
|
41
|
O’Neill E, Curham L, Ní Chasaide C, O’Brien S, McManus G, Moran B, Rubin K, Glazer S, Lynch MA, Mills KH. Neonatal infection with Bordetella pertussis promotes autism-like phenotypes in mice. iScience 2025; 28:111548. [PMID: 39897939 PMCID: PMC11784780 DOI: 10.1016/j.isci.2024.111548] [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: 04/11/2024] [Revised: 09/09/2024] [Accepted: 12/04/2024] [Indexed: 02/04/2025] Open
Abstract
Autism spectrum disorder (ASD) has been linked with infections early in life. Here we demonstrate that the infection of neonatal mice with the respiratory pathogen Bordetella pertussis leads to neuroinflammation, neurodevelopmental defects, and ASD-like behaviors. Following the respiratory challenge of neonatal mice with B. pertussis, multiple atypical CNS findings were observed, including blood-brain barrier disruption, dissemination of live B. pertussis bacteria to the brain with the concomitant infiltration of inflammatory monocytes, neutrophils, and activated IL-17A- and IFN-γ-producing CD4 T cells. Microglia from infected mice were activated, with impaired phagocytic function, resulting in defective synaptic pruning and disrupted neuronal circuit formation. Impaired neurodevelopment in B. pertussis-infected post-natal mice was associated with ASD-like behavioral abnormalities in young adulthood. Our data indicate that infection with virulent B. pertussis during infancy increases the risk of autism-like behavior in young adult mice. A study into the potential role of B. pertussis in human ASD is warranted.
Collapse
Affiliation(s)
- Eoin O’Neill
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, D02PD91 Dublin, Ireland
| | - Lucy Curham
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Caitlín Ní Chasaide
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Síofra O’Brien
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Gavin McManus
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Barry Moran
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Keith Rubin
- ILiAD Biotechnologies, Weston, FL 33331, USA
| | | | - Marina A. Lynch
- Trinity College Institute of Neuroscience, Trinity College Dublin, D02PD91 Dublin, Ireland
| | - Kingston H.G. Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| |
Collapse
|
42
|
Marin-Castañeda LA, Pacheco Aispuro G, Gonzalez-Garibay G, Martínez Zamora CA, Romo-Parra H, Rubio-Osornio M, Rubio C. Interplay of epilepsy and long-term potentiation: implications for memory. Front Neurosci 2025; 18:1451740. [PMID: 39867454 PMCID: PMC11760605 DOI: 10.3389/fnins.2024.1451740] [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: 09/17/2024] [Accepted: 12/20/2024] [Indexed: 01/28/2025] Open
Abstract
The interplay between long-term potentiation (LTP) and epilepsy represents a crucial facet in understanding synaptic plasticity and memory within neuroscience. LTP, a phenomenon characterized by a sustained increase in synaptic strength, is pivotal in learning and memory processes, particularly in the hippocampus. This review delves into the intricate relationship between LTP and epilepsy, exploring how alterations in synaptic plasticity mechanisms akin to those seen in LTP contribute to the hyperexcitable state of epilepsy. This state is conceptualized as a dysregulation between LTP and LTD (Long-term depression), leading to pathologically enhanced synaptic efficacy. Additionally, the role of neuroinflammation in both LTP and epilepsy is examined, highlighting how inflammatory mediators can influence synaptic plasticity. The dual role of neuroinflammatory pathways, enhancing or inhibiting LTP, is a focal area of ongoing research. The significance of various signaling pathways, including the MAPK, mTOR, and WNT/β-catenin pathways, in the modulation of synaptic plasticity and their relevance in both LTP and epilepsy. These pathways are instrumental in memory formation, consolidation, and epileptogenesis, illustrating a complex interaction between cellular mechanisms in the nervous system. Lastly, the role of calcium signaling in the relationship between LTP and epilepsy is scrutinized. Aberrant calcium signaling in epilepsy leads to an enhanced, yet pathologically altered, LTP. This dysregulation disrupts normal neural pathways, potentially leading to cognitive dysfunction, particularly in memory encoding and retrieval. The review emphasizes the need for targeted interventions in epilepsy that address cognitive functions alongside seizure control.
Collapse
Affiliation(s)
- Luis A. Marin-Castañeda
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| | | | - Guillermo Gonzalez-Garibay
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
- Anahuac University, Mexico City, Mexico
| | - Carlos Alejandro Martínez Zamora
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
- School of Medicine, Saint Luke, Mexico City, Mexico
| | - Hector Romo-Parra
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
- Universidad Iberoamericana, Mexico City, Mexico
| | - Moisés Rubio-Osornio
- Department of Neurochemistry, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| | - Carmen Rubio
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| |
Collapse
|
43
|
Chu MC, Wu HF, Lee CW, Wu CC, Chi H, Ko CY, Lee YC, Tang CW, Chen PS, Lin HC. Soluble epoxide hydrolase deletion rescues behavioral and synaptic deficits by AMPK-mTOR pathway in autism animals. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111190. [PMID: 39510156 DOI: 10.1016/j.pnpbp.2024.111190] [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: 06/13/2024] [Revised: 10/25/2024] [Accepted: 11/03/2024] [Indexed: 11/15/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social defects often accompanied with emotional comorbidities. Aberrations in synaptic function and plasticity are the core feature in the pathophysiology of ASD. Targeting soluble epoxide hydrolase (sEH) has been found to exert protection in a wide-range of pathological conditions. However, the regulation of sEH deficiency on the synaptic deficits of ASD and the underlying mechanisms remain unclear. The valproate (VPA)-treated ASD animal model with genetic sEH knockout was applied in the present study. The results showed that the sEH expression was significantly increased in the prefrontal cortex of VPA-treated animals. Although no effect was found on tail malformation and body weight loss, genetic sEH deletion alleviated social deficits, and fear learning and memory extinction in the VPA-treated mice. After a series of electrophysiological assessments, we found that the beneficial effects of sEH deletion focused on the long-term synaptic plasticity, rather than presynaptic efficiency, in the VPA-treated mice. Furthermore, we observed that the dysregulated AMPK-mTOR pathway was restored under genetic sEH deletion in VPA-treated mice. Taken together, these findings uncovered an important role of sEH deficiency in the synaptic dysfunctions of ASD mediated by AMPK-mTOR pathway, providing a novel therapeutic target for ASD.
Collapse
Affiliation(s)
- Ming-Chia Chu
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Han-Fang Wu
- Department of Optometry, MacKay Medical College, New Taipei City, Taiwan
| | - Chi-Wei Lee
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chi-Chun Wu
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiang Chi
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chiung-Yuan Ko
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan; Institute of Biomedical Sciences, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Biomedical Science and Environment Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan; Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chao Lee
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chih-Wei Tang
- Department of Neurology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Po See Chen
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hui-Ching Lin
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Brain Research Center and Membrane Protein Structural Biology Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
44
|
Mohamed AF, El-Gammal MA, El-Yamany MF, Khodeir AE. Sigma-1 receptor modulation by fluvoxamine ameliorates valproic acid-induced autistic behavior in rats: Involvement of chronic ER stress modulation, enhanced autophagy and M1/M2 microglia polarization. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111192. [PMID: 39510157 DOI: 10.1016/j.pnpbp.2024.111192] [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: 06/11/2024] [Revised: 10/21/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder. While, fluvoxamine (FVX) is an antidepressant and widely prescribed to ASD patients, clinical results are inconclusive and the mechanism of FVX in the management of ASD is unclear. This study determined the potential therapeutic impact of FVX, a sigma-1 receptor (S1R) agonist, against the valproic acid (VPA)-induced model of autism. On gestational day 12.5, Wistar pregnant rats were given a single intraperitoneal (i.p.) injection of either VPA (600 mg/kg) or normal saline (10 mL/kg, vehicle-control). Starting on postnatal day (PND) 21 to PND 50, FVX (30 mg/kg, P·O. daily) and NE-100, (S1R) antagonist, (1 mg/kg, i.p. daily) were given to male pups. Behavior tests and histopathological changes were identified at the end of the experiment. In addition, the cerebellum biomarkers of endoplasmic reticulum (ER) stress and autophagy were assessed. Microglial cell polarization to M1 and M2 phenotypes was also assessed. FVX effectively mitigated the histopathological alterations in the cerebellum caused by VPA. FVX enhanced sociability and stereotypic behaviors in addition to its noteworthy impact on autophagy enhancement, ER stress deterioration, and controlling microglial cell polarization. The current investigation confirmed that the S1R agonist, FVX, can lessen behavioral and neurochemical alterations in the VPA-induced rat model of autism.
Collapse
Affiliation(s)
- Ahmed F Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Governorate, Giza 11562, Egypt; Faculty of Pharmacy, King Salman International University (KSIU), South Sinai 46612, Egypt.
| | - Mohamad A El-Gammal
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University, New Damietta 34518, Egypt.
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Governorate, Giza 11562, Egypt.
| | - Ahmed E Khodeir
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University, New Damietta 34518, Egypt.
| |
Collapse
|
45
|
Bose R, Posada-Pérez M, Karvela E, Skandik M, Keane L, Falk A, Spulber S, Joseph B, Ceccatelli S. Bi-allelic NRXN1α deletion in microglia derived from iPSC of an autistic patient increases interleukin-6 production and impairs supporting function on neuronal networking. Brain Behav Immun 2025; 123:28-42. [PMID: 39243986 DOI: 10.1016/j.bbi.2024.09.001] [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: 01/19/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Abstract
Autism spectrum disorder (ASD) is a set of heterogeneous neurodevelopmental conditions, with a highly diverse genetic hereditary component, including altered neuronal circuits, that has an impact on communication skills and behaviours of the affected individuals. Beside the recognised role of neuronal alterations, perturbations of microglia and the associated neuroinflammatory processes have emerged as credible contributors to aetiology and physiopathology of ASD. Mutations in NRXN1, a member of the neurexin family of cell-surface receptors that bind neuroligin, have been associated to ASD. NRXN1 is known to be expressed by neurons where it facilitates synaptic contacts, but it has also been identified in glial cells including microglia. Asserting the impact of ASD-related genes on neuronal versus microglia functions has been challenging. Here, we present an ASD subject-derived induced pluripotent stem cells (iPSC)-based in vitro system to characterise the effects of the ASD-associated NRXN1 gene deletion on neurons and microglia, as well as on the ability of microglia to support neuronal circuit formation and function. Using this approach, we demonstrated that NRXN1 deletion, impacting on the expression of the alpha isoform (NRXN1α), in microglia leads to microglial alterations and release of IL6, a pro-inflammatory interleukin associated with ASD. Moreover, microglia bearing the NRXN1α-deletion, lost the ability to support the formation of functional neuronal networks. The use of recombinant IL6 protein on control microglia-neuron co-cultures or neutralizing antibody to IL6 on their NRXN1α-deficient counterparts, supported a direct contribution of IL6 to the observed neuronal phenotype. Altogether, our data suggest that, in addition to neurons, microglia are also negatively affected by NRXN1α-deletion, and this significantly contributes to the observed neuronal circuit aberrations.
Collapse
Affiliation(s)
- Raj Bose
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; Center for Neuromusculoskeletal Restorative Medicine, Shui On Centre, Wan Chai, Hong Kong
| | - Mercedes Posada-Pérez
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, 17177 Stockholm, Sweden; Center for Neuromusculoskeletal Restorative Medicine, Shui On Centre, Wan Chai, Hong Kong
| | - Eleni Karvela
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Martin Skandik
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Lily Keane
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; Center for Neuromusculoskeletal Restorative Medicine, Shui On Centre, Wan Chai, Hong Kong; Lund Stem Cell Center, Lund University, 22100 Lund, Sweden
| | - Stefan Spulber
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; Center for Neuromusculoskeletal Restorative Medicine, Shui On Centre, Wan Chai, Hong Kong
| | - Bertrand Joseph
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, 17177 Stockholm, Sweden; Center for Neuromusculoskeletal Restorative Medicine, Shui On Centre, Wan Chai, Hong Kong
| | - Sandra Ceccatelli
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; Center for Neuromusculoskeletal Restorative Medicine, Shui On Centre, Wan Chai, Hong Kong.
| |
Collapse
|
46
|
Prapiadou S, Mayerhofer E, Georgakis MK, Kals M, Radmanesh F, Izzy S, Richardson S, Okonkwo D, Puccio A, Temkin N, Palotie A, Ripatti S, Diaz-Arrastia R, Stein MB, Manley G, Menon DK, Rosand J, Parodi L, Anderson CD, on behalf of The Genetic Associations In Neurotrauma (GAIN) Consortium (with contribution from the CENTER-TBI, TRACK-TBI, CABI, MGB, and TBIcare studies). Exploring Synaptic Pathways in Traumatic Brain Injury: A Cross-Phenotype Genomics Approach. J Neurotrauma 2025; 42:131-142. [PMID: 39264867 PMCID: PMC12056582 DOI: 10.1089/neu.2024.0153] [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: 09/14/2024] Open
Abstract
Traumatic brain injury (TBI), a global leading cause of mortality and disability, lacks effective treatments to enhance recovery. Synaptic remodeling has been postulated as one mechanism that influences outcomes after TBI. We sought to investigate whether common mechanisms affecting synapse maintenance are shared between TBI and other neuropsychiatric conditions using pathway enrichment tools and genome-wide genotype data, with the goal of highlighting novel treatment targets. We leveraged an integrative approach, combining data from genome-wide association studies with pathway and gene-set enrichment analyses. Literature review-based and Reactome database-driven approaches were combined to identify synapse-related pathways of interest in TBI outcome and to assess for shared associations with conditions in which synapse-related pathobiological mechanisms have been implicated, including Alzheimer's disease, schizophrenia (SCZ), major depressive disorder, post-traumatic stress disorder, attention-deficit hyperactivity disorder, and autism spectrum disorder. Gene and pathway-level enrichment analyses were conducted using MAGMA and its extensions, e- and H-MAGMA, followed by Mendelian randomization to investigate potential causal associations. Of the 98 pathways tested, 32 were significantly enriched in the included conditions. In TBI outcome, we identified significant enrichment in five pathways: "Serotonin clearance from the synaptic cleft" (p = 0.0001), "Presynaptic nicotinic acetylcholine receptors" (p = 0.0003), "Postsynaptic nicotinic acetylcholine receptors" (p = 0.0003), "Highly sodium permeable postsynaptic acetylcholine nicotinic receptors" (p = 0.0001), and "Acetylcholine binding and downstream events" pathways (p = 0.0003). These associations highlight potential involvement of the cholinergic and serotonergic systems in post-TBI recovery. Three of those pathways were shared between TBI and SCZ, suggesting possible pathophysiologic commonalities. In this study, we utilize comparative and integrative genomic approaches across brain conditions that share synaptic mechanisms to explore the pathophysiology of TBI outcomes. Our results implicate associations between TBI outcome and synaptic pathways as well as pathobiological overlap with other neuropsychiatric diseases.
Collapse
Affiliation(s)
| | | | - Marios K. Georgakis
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Mart Kals
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Farid Radmanesh
- Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Division of Neurocritical Care, Department of Neurology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Saef Izzy
- Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Sylvia Richardson
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | - David Okonkwo
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ava Puccio
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nancy Temkin
- Departments of Neurological Surgery and Biostatistics, University of Washington, Seattle, Washington, USA
| | - Aarno Palotie
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Samuli Ripatti
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Murray B. Stein
- Department of Psychiatry, School of Medicine, and School of Public Health, University of California, La Jolla, California, USA
| | - Geoff Manley
- Department of Neurosurgery, University of California, San Francisco, California, USA
| | - David K. Menon
- Division of Anaesthesia, University of Cambridge, Cambridge, UK
| | | | - Livia Parodi
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | | | - on behalf of The Genetic Associations In Neurotrauma (GAIN) Consortium (with contribution from the CENTER-TBI, TRACK-TBI, CABI, MGB, and TBIcare studies)
- University of Patras Medical School, Patras, Greece
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, Massachusetts, USA
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Division of Neurocritical Care, Department of Neurology, University of New Mexico, Albuquerque, New Mexico, USA
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Departments of Neurological Surgery and Biostatistics, University of Washington, Seattle, Washington, USA
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, School of Medicine, and School of Public Health, University of California, La Jolla, California, USA
- Department of Neurosurgery, University of California, San Francisco, California, USA
- Division of Anaesthesia, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Wang X, Li Q, Lyu Z, Wu Y. Supplementing with Vitamin D during Pregnancy Reduces Inflammation and Prevents Autism-Related Behaviors in Offspring Caused by Maternal Immune Activation. Biol Pharm Bull 2025; 48:632-640. [PMID: 40383635 DOI: 10.1248/bpb.b25-00008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Autism spectrum disorder (ASD), a neurodevelopmental disorder of unknown etiology with limited treatment options, has emerged as a significant public health concern. Studies have demonstrated that prenatal vitamin D deficiency is a risk factor for ASD development in offspring; however, the underlying mechanism remains unclear. In this project, vitamin D was administered orally to pregnant mice with/without the subsequent administration of polyriboinosinic polyribocytidylic acid (Poly(I:C)), which induced the maternal immune activation (MIA). Our results showed that vitamin D supplementation during pregnancy alleviated MIA-induced ASD-like behaviors in offspring. Moreover, vitamin D supplementation reduced the MIA-induced elevation of interleukin-6 (IL-6) and IL-17a levels in both the maternal ileum and fetal brains. It also suppressed signal transducer and activator of transcription 3 (Stat3) activation and the elevated expression of serum amyloid A1 and A2 (SAA1/2) in the ileum of MIA-affected pregnant mice. This study revealed that vitamin D may reduce the expression of IL-17a by inhibiting the IL-6/Stat3/SAA signaling pathway, thereby improving ASD-like behavior in offspring mice, and provide a new theoretical support for the prevention and treatment of ASD by scientific dietary interventions and nutritional supplement during pregnancy.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Qingqing Li
- Department of Human Anatomy, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Zhihong Lyu
- Department of Psychiatry, Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Yingying Wu
- Department of Human Anatomy, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| |
Collapse
|
48
|
Heine VM, Dooves S. Neuroglia in autism spectrum disorders. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:303-311. [PMID: 40148051 DOI: 10.1016/b978-0-443-19102-2.00006-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Autism spectrum disorder (ASD) is characterized by difficulties in social interaction, communication, and repetitive behavior, typically diagnosed during early childhood and attributed to altered neuronal network connectivity. Several genetic and environmental risk factors contribute to ASD, including pre- or early life immune activation, which can trigger microglial and astroglial reactivity, impacting early neurodevelopment. In ASD, astrocytes show altered glutamate metabolism, directly influencing neuronal network activity, while microglia display impaired synaptic pruning, an essential developmental process for the refinement of neuronal connections. Additionally, reduced myelination in specific cortical and subcortical regions may affect brain connectivity in ASD, with white matter integrity correlating with the severity of the disorder, suggesting an important role for oligodendrocytes and myelin in ASD. This chapter provides an overview of current literature on the role of neuroglia cells in ASD, with a focus on immune activation, glutamate signaling, synaptic pruning, and myelination.
Collapse
Affiliation(s)
- Vivi M Heine
- Department of Child and Adolescence Psychiatry, Emma Center for Personalized Medicine, Amsterdam Neuroscience, Emma Children's Hospital, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| | - Stephanie Dooves
- Department of Child and Adolescence Psychiatry, Emma Center for Personalized Medicine, Amsterdam Neuroscience, Emma Children's Hospital, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
49
|
Kong C, Bing Z, Yang L, Huang Z, Wang W, Grebogi C. Transcriptomic Evidence Reveals the Dysfunctional Mechanism of Synaptic Plasticity Control in ASD. Genes (Basel) 2024; 16:11. [PMID: 39858558 PMCID: PMC11764921 DOI: 10.3390/genes16010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND/OBJECTIVES A prominent endophenotype in Autism Spectrum Disorder (ASD) is the synaptic plasticity dysfunction, yet the molecular mechanism remains elusive. As a prototype, we investigate the postsynaptic signal transduction network in glutamatergic neurons and integrate single-cell nucleus transcriptomics data from the Prefrontal Cortex (PFC) to unveil the malfunction of translation control. METHODS We devise an innovative and highly dependable pipeline to transform our acquired signal transduction network into an mRNA Signaling-Regulatory Network (mSiReN) and analyze it at the RNA level. We employ Cell-Specific Network Inference via Integer Value Programming and Causal Reasoning (CS-NIVaCaR) to identify core modules and Cell-Specific Probabilistic Contextualization for mRNA Regulatory Networks (CS-ProComReN) to quantitatively reveal activated sub-pathways involving MAPK1, MKNK1, RPS6KA5, and MTOR across different cell types in ASD. RESULTS The results indicate that specific pivotal molecules, such as EIF4EBP1 and EIF4E, lacking Differential Expression (DE) characteristics and responsible for protein translation with long-term potentiation (LTP) or long-term depression (LTD), are dysregulated. We further uncover distinct activation patterns causally linked to the EIF4EBP1-EIF4E module in excitatory and inhibitory neurons. CONCLUSIONS Importantly, our work introduces a methodology for leveraging extensive transcriptomics data to parse the signal transduction network, transforming it into mSiReN, and mapping it back to the protein level. These algorithms can serve as potent tools in systems biology to analyze other omics and regulatory networks. Furthermore, the biomarkers within the activated sub-pathways, revealed by identifying convergent dysregulation, illuminate potential diagnostic and prognostic factors in ASD.
Collapse
Affiliation(s)
- Chao Kong
- School of Systems Science, Beijing Normal University, Beijing 100875, China;
| | - Zhitong Bing
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Lei Yang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Zigang Huang
- School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Wenxu Wang
- School of Systems Science, Beijing Normal University, Beijing 100875, China;
| | - Celso Grebogi
- Institute for Complex Systems and Mathematical Biology, King’s College, University of Aberdeen, Old Aberdeen AB24 3UE, UK
| |
Collapse
|
50
|
Carbonell-Roig J, Aaltonen A, Wilson K, Molinari M, Cartocci V, McGuirt A, Mosharov E, Kehr J, Lieberman OJ, Sulzer D, Borgkvist A, Santini E. Dysregulated acetylcholine-mediated dopamine neurotransmission in the eIF4E Tg mouse model of autism spectrum disorders. Cell Rep 2024; 43:114997. [PMID: 39607825 DOI: 10.1016/j.celrep.2024.114997] [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: 02/05/2024] [Revised: 09/18/2024] [Accepted: 11/05/2024] [Indexed: 11/30/2024] Open
Abstract
Autism spectrum disorder (ASD) consists of diverse neurodevelopmental conditions where core behavioral symptoms are critical for diagnosis. Altered dopamine (DA) neurotransmission in the striatum has been suggested to contribute to the behavioral features of ASD. Here, we examine DA neurotransmission in a mouse model of ASD characterized by elevated expression of eukaryotic initiation factor 4E (eIF4E), a key regulator of cap-dependent translation, using a comprehensive approach that encompasses genetics, behavior, synaptic physiology, and imaging. The results indicate that increased eIF4E expression leads to behavioral inflexibility and impaired striatal DA release. The loss of normal DA neurotransmission is due to a defect in nicotinic receptor signaling that regulates calcium dynamics in dopaminergic axons. These findings provide a mechanistic understanding of ASD symptoms and offer a foundation for targeted therapeutic interventions by revealing the intricate interplay between eIF4E, DA neurotransmission, and behavioral flexibility.
Collapse
Affiliation(s)
| | - Alina Aaltonen
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
| | - Karin Wilson
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
| | - Maya Molinari
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
| | - Veronica Cartocci
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
| | - Avery McGuirt
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA
| | - Eugene Mosharov
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA
| | - Jan Kehr
- Pronexus Analytical AB, 16733 Stockholm-Bromma, Sweden
| | - Ori J Lieberman
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA; Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA 94143, USA
| | - David Sulzer
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA
| | - Anders Borgkvist
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden.
| | - Emanuela Santini
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden.
| |
Collapse
|