1
|
Chen Y, Hu J, Zhang Y, Peng L, Li X, Li C, Wu X, Wang C. Epilepsy therapy beyond neurons: Unveiling astrocytes as cellular targets. Neural Regen Res 2026; 21:23-38. [PMID: 39819836 PMCID: PMC12094549 DOI: 10.4103/nrr.nrr-d-24-01035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 10/16/2024] [Accepted: 12/10/2024] [Indexed: 01/19/2025] Open
Abstract
Epilepsy is a leading cause of disability and mortality worldwide. However, despite the availability of more than 20 antiseizure medications, more than one-third of patients continue to experience seizures. Given the urgent need to explore new treatment strategies for epilepsy, recent research has highlighted the potential of targeting gliosis, metabolic disturbances, and neural circuit abnormalities as therapeutic strategies. Astrocytes, the largest group of nonneuronal cells in the central nervous system, play several crucial roles in maintaining ionic and energy metabolic homeostasis in neurons, regulating neurotransmitter levels, and modulating synaptic plasticity. This article briefly reviews the critical role of astrocytes in maintaining balance within the central nervous system. Building on previous research, we discuss how astrocyte dysfunction contributes to the onset and progression of epilepsy through four key aspects: the imbalance between excitatory and inhibitory neuronal signaling, dysregulation of metabolic homeostasis in the neuronal microenvironment, neuroinflammation, and the formation of abnormal neural circuits. We summarize relevant basic research conducted over the past 5 years that has focused on modulating astrocytes as a therapeutic approach for epilepsy. We categorize the therapeutic targets proposed by these studies into four areas: restoration of the excitation-inhibition balance, reestablishment of metabolic homeostasis, modulation of immune and inflammatory responses, and reconstruction of abnormal neural circuits. These targets correspond to the pathophysiological mechanisms by which astrocytes contribute to epilepsy. Additionally, we need to consider the potential challenges and limitations of translating these identified therapeutic targets into clinical treatments. These limitations arise from interspecies differences between humans and animal models, as well as the complex comorbidities associated with epilepsy in humans. We also highlight valuable future research directions worth exploring in the treatment of epilepsy and the regulation of astrocytes, such as gene therapy and imaging strategies. The findings presented in this review may help open new therapeutic avenues for patients with drug-resistant epilepsy and for those suffering from other central nervous system disorders associated with astrocytic dysfunction.
Collapse
Affiliation(s)
- Yuncan Chen
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiayi Hu
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Zhang
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lulu Peng
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoyu Li
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cong Li
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xunyi Wu
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Cong Wang
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Shanghai, China
- Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| |
Collapse
|
2
|
Liu X, Wu W, Li X, Wang C, Chai K, Yuan F, Zheng H, Yao Y, Li C, Ye ZC, Zha D. The compound (E)-2-(3,4-dihydroxystyryl)-3-hydroxy-4H-pyran-4-one alleviates neuroinflammation and cognitive impairment in a mouse model of Alzheimer's disease. Neural Regen Res 2025; 20:3330-3344. [PMID: 39715098 PMCID: PMC11881737 DOI: 10.4103/nrr.nrr-d-23-01890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 04/17/2024] [Accepted: 06/02/2024] [Indexed: 12/25/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202511000-00034/figure1/v/2024-12-20T164640Z/r/image-tiff Previous studies have shown that the compound (E)-2-(3,4-dihydroxystyryl)-3-hydroxy-4H-pyran-4-one (D30), a pyromeconic acid derivative, possesses antioxidant and anti-inflammatory properties, inhibits amyloid-β aggregation, and alleviates scopolamine-induced cognitive impairment, similar to the phase III clinical drug resveratrol. In this study, we established a mouse model of Alzheimer's disease via intracerebroventricular injection of fibrillar amyloid-β to investigate the effect of D30 on fibrillar amyloid-β-induced neuropathology. Our results showed that D30 alleviated fibrillar amyloid-β-induced cognitive impairment, promoted fibrillar amyloid-β clearance from the hippocampus and cortex, suppressed oxidative stress, and inhibited activation of microglia and astrocytes. D30 also reversed the fibrillar amyloid-β-induced loss of dendritic spines and synaptic protein expression. Notably, we demonstrated that exogenous fibrillar amyloid-β introduced by intracerebroventricular injection greatly increased galectin-3 expression levels in the brain, and this increase was blocked by D30. Considering the role of D30 in clearing amyloid-β, inhibiting neuroinflammation, protecting synapses, and improving cognition, this study highlights the potential of galectin-3 as a promising treatment target for patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Xueyan Liu
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Wei Wu
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Xuejuan Li
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Chengyan Wang
- Institute of Laboratory Animal Center, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ke Chai
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Fanru Yuan
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Huijuan Zheng
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yuxing Yao
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Chenlu Li
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Hyperbaric Oxygen, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Zu-Cheng Ye
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Daijun Zha
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
3
|
Ren J, Wang Y, Wang Y, Zhang Y, Xing M, Deng S, Tong S, Wang L, Zheng C, Yang J, Ni G, Ming D. Dynamic changes of hippocampal dendritic spines in Alzheimer's disease mice among the different stages. Exp Neurol 2025; 390:115266. [PMID: 40246009 DOI: 10.1016/j.expneurol.2025.115266] [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: 01/20/2025] [Revised: 03/16/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of amyloid-β (Aβ) peptides and a progressive decline in cognitive function. Hippocampus as a crucial brain area for learning and memory, is also adversely affected by AD's pathology. The accumulation of Aβ is often associated with the loss of dendritic spines of the hippocampus. However, the dynamic alterations in dendritic spines throughout AD progression are not fully understood. To investigate it, we conducted in-vivo imaging in two mouse models representing the early and late stages of AD pathology: young mice injected with Aβ1-42 oligomers and APP/PS1 transgenic mice. In the early-stage AD model, imaging was conducted at third- and fifth- weeks post-injection. In the late-stage AD model, a four-month imaging began at 14 months old. The imaging results showed spine elimination in both models. Notably, acute Aβ exposure was linked to heightened spine loss on secondary dendrites, while in the late stage the primary effect was on tertiary dendrites. Concurrently, with the metabolism of Aβ, cognition recovered to some extent by five weeks post Aβ1-42 exposure. These findings suggested that dendritic spine plasticity was impaired during the development of AD, as evidenced by increasing spine loss at different levels. However, the cognitive recovery observed in early-stage AD model mice may indicate a compensatory structural reorganization, highlighting the potential of early intervention to mitigate disease progression. Our results provide novel insights into the neurotoxic effects of Aβ1-42 and may contribute to the development of therapeutic strategies for AD.
Collapse
Affiliation(s)
- Jing Ren
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Yimeng Wang
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Yinuo Wang
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Yiping Zhang
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Mu Xing
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Shouzhe Deng
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Siyi Tong
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Ling Wang
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin 300072, China; Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin 300392, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin 300072, China
| | - Chenguang Zheng
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin 300072, China; Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin 300392, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin 300072, China
| | - Jiajia Yang
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin 300072, China; Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin 300392, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin 300072, China.
| | - Guangjian Ni
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin 300072, China; Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin 300392, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin 300072, China.
| | - Dong Ming
- Medical School of Tianjin University, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, Tianjin 300072, China; Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin 300392, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
4
|
Xin Q, Wang J, Zheng J, Tan Y, Jia X, Ni Z, Xu Z, Feng J, Wu Z, Li Y, Li XM, Ma H, Hu H. Neuron-astrocyte coupling in lateral habenula mediates depressive-like behaviors. Cell 2025; 188:3291-3309.e24. [PMID: 40280131 DOI: 10.1016/j.cell.2025.04.010] [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: 09/08/2024] [Revised: 01/08/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025]
Abstract
The lateral habenula (LHb) neurons and astrocytes have been strongly implicated in depression etiology, but it was not clear how the two dynamically interact during depression onset. Here, using multi-brain-region calcium photometry recording in freely moving mice, we discover that stress induces a most rapid astrocytic calcium rise and a bimodal neuronal response in the LHb. LHb astrocytic calcium requires the α1A-adrenergic receptor and depends on a recurrent neural network between the LHb and locus coeruleus (LC). Through the gliotransmitter glutamate and ATP/adenosine, LHb astrocytes mediate the second-wave LHb neuronal activation and norepinephrine (NE) release. Activation or inhibition of LHb astrocytic calcium signaling facilitates or prevents stress-induced depressive-like behaviors, respectively. These results identify a stress-induced positive feedback loop in the LHb-LC axis, with astrocytes being a critical signaling relay. The identification of this prominent neuron-glia interaction may shed light on stress management and depression prevention.
Collapse
Affiliation(s)
- Qianqian Xin
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Junying Wang
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Jinkun Zheng
- Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Yi Tan
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China; Department of Psychiatry and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Xiaoning Jia
- Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Zheyi Ni
- Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Zijie Xu
- Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Jiesi Feng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
| | - Zhaofa Wu
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
| | - Xiao-Ming Li
- Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Huan Ma
- Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Hailan Hu
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-Computer Interface Institute, Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou 311121, China; Department of Psychiatry and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China.
| |
Collapse
|
5
|
Zengeler KE, Hollis A, Deutsch TCJ, Samuels JD, Ennerfelt H, Moore KA, Steacy EJ, Sabapathy V, Sharma R, Patel MK, Lukens JR. Inflammasome signaling in astrocytes modulates hippocampal plasticity. Immunity 2025; 58:1519-1535.e11. [PMID: 40318630 PMCID: PMC12158643 DOI: 10.1016/j.immuni.2025.04.007] [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: 04/18/2024] [Revised: 12/13/2024] [Accepted: 04/08/2025] [Indexed: 05/07/2025]
Abstract
Emerging evidence indicates that a baseline level of controlled innate immune signaling is required to support proper brain function. However, little is known about the function of most innate immune pathways in homeostatic neurobiology. Here, we report a role for astrocyte-dependent inflammasome signaling in regulating hippocampal plasticity. Inflammasomes are multiprotein complexes that promote caspase-1-mediated interleukin (IL)-1 and IL-18 production in response to pathogens and tissue damage. We observed that inflammasome complex formation was regularly detected under homeostasis in hippocampal astrocytes and that its assembly is dynamically regulated in response to learning and regional activity. Conditional ablation of caspase-1 in astrocytes limited hyperexcitability in an acute seizure model and impacted hippocampal plasticity via modulation of synaptic protein density, neuronal activity, and perineuronal net coverage. Caspase-1 and IL-18 regulated hippocampal IL-33 production and related plasticity. These findings reveal a homeostatic function for astrocyte inflammasome activity in regulating hippocampal physiology in health and disease.
Collapse
Affiliation(s)
- Kristine E Zengeler
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA.
| | - Ava Hollis
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Tyler C J Deutsch
- Department of Anesthesiology, University of Virginia, Charlottesville, VA 22908, USA
| | - Joshua D Samuels
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Hannah Ennerfelt
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA 24304, USA
| | - Katelyn A Moore
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Eric J Steacy
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Vikram Sabapathy
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, VA 22908, USA
| | - Rahul Sharma
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, VA 22908, USA
| | - Manoj K Patel
- Department of Anesthesiology, University of Virginia, Charlottesville, VA 22908, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
| | - John R Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
6
|
Loera-Lopez AL, Lord MN, Noble EE. Astrocytes of the hippocampus and responses to periprandial neuroendocrine hormones. Physiol Behav 2025; 295:114913. [PMID: 40209869 PMCID: PMC12066093 DOI: 10.1016/j.physbeh.2025.114913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/15/2025] [Accepted: 04/08/2025] [Indexed: 04/12/2025]
Abstract
Astrocytes have risen as stars in the field of energy homeostasis and neurocognitive function, acting as a bridge of communication between the periphery and the brain, providing metabolic support, signaling via gliotransmitters, and altering synaptic communication. Dietary factors and energy state have a profound influence on hippocampal function, and the hippocampus is critical for appropriate behavioral responses associated with feeding and internal hunger cues (being in the fasted or full state), but how the hippocampus senses periprandial status and is impacted by diet is largely unknown. Periprandial hormones act within the hippocampus to modulate processes involved in hippocampal-dependent learning and memory function and astrocytes likely play an important role in modulating this signaling. In addition to periprandial hormones, astrocytes are positioned to respond to changes in circulating nutrients like glucose. Here, we review literature investigating how astrocytes mediate changes in hippocampal function, highlighting astrocyte location, morphology, and function in the context of integrating glucose metabolism, neuroendocrine hormone action, and/or cognitive function in the hippocampus. Specifically, we discuss research findings on the effects of insulin, ghrelin, leptin, and GLP-1 on glucose homeostasis, neural activity, astrocyte function, and behavior in the hippocampus. Because obesogenic diets impact neuroendocrine hormones, astrocytes, and cognitive function, we also discuss the effects of diet and diet-induced obesity on these parameters.
Collapse
Affiliation(s)
- Ana L Loera-Lopez
- Neuroscience Graduate Program, University of Georgia, Athens, GA, 30606, USA; Department of Nutritional Sciences, University of Georgia, Athens, GA, 30606, USA
| | - Magen N Lord
- Department of Nutritional Sciences, University of Georgia, Athens, GA, 30606, USA
| | - Emily E Noble
- Neuroscience Graduate Program, University of Georgia, Athens, GA, 30606, USA; Department of Nutritional Sciences, University of Georgia, Athens, GA, 30606, USA.
| |
Collapse
|
7
|
Li H, Zhao Y, Dai R, Geng P, Weng D, Wu W, Yu F, Lin R, Wu Z, Li Y, Luo M. Astrocytes release ATP/ADP and glutamate in flashes via vesicular exocytosis. Mol Psychiatry 2025; 30:2475-2489. [PMID: 39578520 DOI: 10.1038/s41380-024-02851-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Astrocytes regulate brain functions through gliotransmitters like ATP/ADP and glutamate, but their release patterns and mechanisms remain controversial. Here, we visualized ATP/ADP and glutamate response following astrocyte activation and investigated their mechanisms in vivo. Employing cOpn5-mediated optogenetic stimulation, genetically encoded fluorescent sensors, and two-photon imaging, we observed ATP/ADP released as temporally prolonged and spatially extended flashes that later converted to adenosine. This release occurs via Ca2+ and VNUT-dependent vesicular exocytosis. Additionally, astrocytes also release glutamate in flashes through TeNT-sensitive exocytosis, independent of ATP/ADP release. ATP/ADP released by astrocytes triggers further ATP/ADP release from microglia through P2Y12- and VNUT-dependent mechanisms. VNUT in astrocytes and microglia also contributes to ATP/ADP release under LPS-induced brain inflammation. These findings establish Ca2+-dependent vesicular exocytosis as a key mode of action, reveal intricate astrocyte-microglia interactions, and suggest a role for gliotransmission in brain inflammation. Furthermore, the methodologies may provide valuable tools for deciphering glial physiology and pathophysiology.
Collapse
Affiliation(s)
- Heng Li
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
- School of Life Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), 102206, Beijing, China
| | - Yuqing Zhao
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
| | - Ruicheng Dai
- Chinese Institute for Brain Research (CIBR), Beijing, 102206, Beijing, China
| | - Peiyao Geng
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), 102206, Beijing, China
| | - Danwei Weng
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
| | - Wenting Wu
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), 102206, Beijing, China
| | - Fengting Yu
- Chinese Institute for Brain Research (CIBR), Beijing, 102206, Beijing, China
| | - Rui Lin
- National Institute of Biological Sciences (NIBS), Beijing, 102206, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), 102206, Beijing, China
| | - Zhaofa Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, 100871, Beijing, China
- New Cornerstone Science Laboratory, 518054, Shenzhen, China
| | - Minmin Luo
- Chinese Institute for Brain Research (CIBR), Beijing, 102206, Beijing, China.
- New Cornerstone Science Laboratory, 518054, Shenzhen, China.
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, 100005, Beijing, China.
- Beijing Institute for Brain Research, Chinese Academy of Medical Sciences & Peking Union Medical College, 102206, Beijing, China.
| |
Collapse
|
8
|
Kozachkov L, Slotine JJ, Krotov D. Neuron-astrocyte associative memory. Proc Natl Acad Sci U S A 2025; 122:e2417788122. [PMID: 40408402 DOI: 10.1073/pnas.2417788122] [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: 09/11/2024] [Accepted: 04/07/2025] [Indexed: 05/25/2025] Open
Abstract
Astrocytes, the most abundant type of glial cell, play a fundamental role in memory. Despite most hippocampal synapses being contacted by an astrocyte, there are no current theories that explain how neurons, synapses, and astrocytes might collectively contribute to memory function. We demonstrate that fundamental aspects of astrocyte morphology and physiology naturally lead to a dynamic, high-capacity associative memory system. The neuron-astrocyte networks generated by our framework are closely related to popular machine learning architectures known as Dense Associative Memories. Adjusting the connectivity pattern, the model developed here leads to a family of associative memory networks that includes a Dense Associative Memory and a Transformer as two limiting cases. In the known biological implementations of Dense Associative Memories, the ratio of stored memories to the number of neurons remains constant, despite the growth of the network size. Our work demonstrates that neuron-astrocyte networks follow a superior memory scaling law, outperforming known biological implementations of Dense Associative Memory. Our model suggests an exciting and previously unnoticed possibility that memories could be stored, at least in part, within the network of astrocyte processes rather than solely in the synaptic weights between neurons.
Collapse
Affiliation(s)
- Leo Kozachkov
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
- Thomas J. Watson Research Center, International Business Machines Research, Yorktown Heights, NY 10598
| | - Jean-Jacques Slotine
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Dmitry Krotov
- Massachusetts Institute of Technology-International Business Machines, Watson Artificial Intelligence Laboratory, International Business Machines Research, Cambridge, MA 02142
| |
Collapse
|
9
|
Bellier F, Walter A, Lecoin L, Chauveau F, Rouach N, Rancillac A. Astrocytes at the heart of sleep: from genes to network dynamics. Cell Mol Life Sci 2025; 82:207. [PMID: 40397158 PMCID: PMC12095758 DOI: 10.1007/s00018-025-05671-3] [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: 12/14/2024] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 05/22/2025]
Abstract
Astrocytes have transcended their role from mere structural scaffolds to pivotal regulators of neural circuitry and sleep-wake dynamics. The strategic proximity of their fine processes to blood vessels and synapses positions them as key players in neurobiology, contributing to the tripartite synapse concept. Gap-junction proteins also enable astrocytes to form an extensive network interacting with neuronal assemblies to influence sleep physiology. Recent advances in genetic engineering, neuroimaging and molecular biology have deepened our understanding of astrocytic functions. This review highlights the different mechanisms by which astrocytes regulate sleep, notably through transcriptomic and morphological changes, as well as gliotransmission, whereby intracellular calcium (Ca2+) dynamics plays a significant role in modulating the sleep-wake cycle. In vivo optogenetic stimulation of astrocytes indeed induces ATP release, which is subsequently degraded into adenosine, modulating neuronal excitability in sleep-wake regulatory brain regions. Astrocytes also participate in synaptic plasticity, potentially modulating sleep-associated downscaling, a process essential for memory consolidation and preventing synaptic saturation. Although astrocytic involvement in synaptic maintenance is well supported, the precise molecular mechanisms linking these processes to sleep regulation remain to be elucidated. By highlighting astrocytes' multiple roles in sleep physiology, these insights deepen our understanding of sleep mechanisms and pave the way for improving sleep quality.
Collapse
Affiliation(s)
- Félix Bellier
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology-CIRB, Collège de France, CNRS UMR 7241/Inserm U1050, Université PSL, PSL-NEURO, 11, Place Marcelin Berthelot, 75005, Paris, France
- IRBA (Institut de Recherche Biomédicale Des Armées), Brétigny-sur-Orge, France
| | - Augustin Walter
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology-CIRB, Collège de France, CNRS UMR 7241/Inserm U1050, Université PSL, PSL-NEURO, 11, Place Marcelin Berthelot, 75005, Paris, France
| | - Laure Lecoin
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology-CIRB, Collège de France, CNRS UMR 7241/Inserm U1050, Université PSL, PSL-NEURO, 11, Place Marcelin Berthelot, 75005, Paris, France
| | - Fréderic Chauveau
- IRBA (Institut de Recherche Biomédicale Des Armées), Brétigny-sur-Orge, France
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology-CIRB, Collège de France, CNRS UMR 7241/Inserm U1050, Université PSL, PSL-NEURO, 11, Place Marcelin Berthelot, 75005, Paris, France
| | - Armelle Rancillac
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology-CIRB, Collège de France, CNRS UMR 7241/Inserm U1050, Université PSL, PSL-NEURO, 11, Place Marcelin Berthelot, 75005, Paris, France.
| |
Collapse
|
10
|
Li S, Xue Y, Sun Z, Wei H, Wu F, Mao L. A Chemistry-Informed Generative Deep Learning Approach for Enhancing Voltammetric Neurochemical Sensing in Living Mouse Brain. J Am Chem Soc 2025; 147:16804-16811. [PMID: 40358003 DOI: 10.1021/jacs.5c05393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Exploring the time-resolved dynamics of neurochemicals is essential for deciphering neuronal functions, intercellular communication, and neurophysiological or pathological mechanisms. However, the complex interplay among neurochemicals between neurocytes, coupled with extensive chemical signal crosstalk, puts simultaneous sensing of multiple neurochemicals into a longstanding challenge. Herein, we report a chemistry-informed generative neural network (CIGNN) model to separate the Faradaic and the non-Faradaic components from voltammetric currents, minimizing their mutual interference and enhancing quantitative accuracy. With the assistance of generative deep learning, we successfully establish a new platform for in vivo neurochemical sensing, which is validated by simultaneously monitoring the dynamics of dopamine (DA), ascorbic acid (AA), and ionic strength in a neuroinflammation mouse model. We observe that the stimulation with KCl solution triggers a significant enhancement of AA efflux on the model mice (300 ± 50 μM) compared with that from the control mice (170 ± 20 μM), as well as a significant decrease of ion influx (55 ± 7 mM) compared with that from the control mice (120 ± 16 mM), while not evoking a significant change in the DA release from the model mice (2.8 ± 0.3 μM) versus that from the control mice (3.0 ± 0.5 μM). This work provides a robust tool for studying multineurochemical signaling and elucidating the molecular mechanisms underlying various brain activities.
Collapse
Affiliation(s)
- Shuxin Li
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Yifei Xue
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Zhining Sun
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Huan Wei
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Fei Wu
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Lanqun Mao
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
11
|
Appleton E, Tao J, Liu S, Glass C, Fonseca G, Church G. Machine-guided cell-fate engineering. Cell Rep 2025; 44:115726. [PMID: 40382774 DOI: 10.1016/j.celrep.2025.115726] [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: 10/16/2024] [Revised: 03/06/2025] [Accepted: 04/30/2025] [Indexed: 05/20/2025] Open
Abstract
The creation of induced pluripotent stem cells (iPSCs) has enabled scientists to explore the function, mechanisms, and differentiation processes of many types of cells. One of the fastest and most efficient approaches is transcription factor (TF) over-expression. However, finding the right combination of TFs to over-express to differentiate iPSCs directly into other cell types is a difficult task. Here, we describe a machine-learning (ML) pipeline, called CellCartographer, that uses chromatin accessibility and transcriptomics data to design multiplex TF pooled-screening experiments for cell-type conversions that then may be iteratively refined. We validate this method by differentiating iPSCs into twelve cell types at low efficiency in preliminary screens and iteratively refine our TF combinations to achieve high-efficiency differentiation for six of these cell types in <6 days. Finally, we functionally characterize iPSC-derived cytotoxic T cells (iCytoTs), regulatory T cells (iTregs), type II astrocytes (iAstIIs), and hepatocytes (iHeps) to validate functionally accurate differentiation.
Collapse
Affiliation(s)
- Evan Appleton
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.
| | - Jenhan Tao
- Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA.
| | - Songlei Liu
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher Glass
- Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Gregory Fonseca
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, Montréal, QC H4A-3J1, Canada; Quantitative Life Sciences, McGill University, Montréal, QC H4A-3J1, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, QC H4A-3J1, Canada
| | - George Church
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
12
|
Liss A, Siddiqi MT, Marsland P, Varodayan FP. Neuroimmune regulation of the prefrontal cortex tetrapartite synapse. Neuropharmacology 2025; 269:110335. [PMID: 39904409 DOI: 10.1016/j.neuropharm.2025.110335] [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: 11/14/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
The prefrontal cortex (PFC) is an essential driver of cognitive, affective, and motivational behavior. There is clear evidence that the neuroimmune system directly influences PFC synapses, in addition to its role as the first line of defense against toxins and pathogens. In this review, we first describe the core structures that form the tetrapartite PFC synapse, focusing on the signaling microdomain created by astrocytic cradling of the synapse as well as the emerging role of the extracellular matrix in synaptic organization and plasticity. Neuroimmune signals (e.g. pro-inflammatory interleukin 1β) can impact the function of each core structure within the tetrapartite synapse, as well as promote intra-synaptic crosstalk, and we will provide an overview of recent advances in this field. Finally, evidence from post mortem human brain tissue and preclinical studies indicate that inflammation may be a key contributor to PFC dysfunction. Therefore, we conclude with a mechanistic discussion of neuroimmune-mediated maladaptive plasticity in neuropsychiatric disorders, with a focus on alcohol use disorder (AUD). Growing recognition of the neuroimmune system's role as a critical regulator of the PFC tetrapartite synapse provides strong support for targeting the neuroimmune system to develop new pharmacotherapeutics.
Collapse
Affiliation(s)
- Andrea Liss
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Mahum T Siddiqi
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Paige Marsland
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Florence P Varodayan
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA.
| |
Collapse
|
13
|
Mi X, Chen ABY, Duarte D, Carey E, Taylor CR, Braaker PN, Bright M, Almeida RG, Lim JX, Ruetten VMS, Wang Y, Wang M, Zhang W, Zheng W, Reitman ME, Huang Y, Wang X, Li L, Deng H, Shi SH, Poskanzer KE, Lyons DA, Nimmerjahn A, Ahrens MB, Yu G. Fast, accurate, and versatile data analysis platform for the quantification of molecular spatiotemporal signals. Cell 2025; 188:2794-2809.e21. [PMID: 40203826 DOI: 10.1016/j.cell.2025.03.012] [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: 05/31/2024] [Revised: 01/13/2025] [Accepted: 03/06/2025] [Indexed: 04/11/2025]
Abstract
Optical recording of intricate molecular dynamics is becoming an indispensable technique for biological studies, accelerated by the development of new or improved biosensors and microscopy technology. This creates major computational challenges to extract and quantify biologically meaningful spatiotemporal patterns embedded within complex and rich data sources, many of which cannot be captured with existing methods. Here, we introduce activity quantification and analysis (AQuA2), a fast, accurate, and versatile data analysis platform built upon advanced machine-learning techniques. It decomposes complex live-imaging-based datasets into elementary signaling events, allowing accurate and unbiased quantification of molecular activities and identification of consensus functional units. We demonstrate applications across a wide range of biosensors, cell types, organs, animal models, microscopy techniques, and imaging approaches. As exemplar findings, we show how AQuA2 identified drug-dependent interactions between neurons and astroglia, as well as distinct sensorimotor signal propagation patterns in the mouse spinal cord.
Collapse
Affiliation(s)
- Xuelong Mi
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Alex Bo-Yuan Chen
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Graduate Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| | - Daniela Duarte
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Erin Carey
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Charlotte R Taylor
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Philipp N Braaker
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4SB, UK
| | - Mark Bright
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Rafael G Almeida
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4SB, UK
| | - Jing-Xuan Lim
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Virginia M S Ruetten
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA; Gatsby Computational Neuroscience Unit, UCL, London W1T 4JG, UK
| | - Yizhi Wang
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Mengfan Wang
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Weizhan Zhang
- Department of Automation, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Wei Zheng
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Michael E Reitman
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Yongkang Huang
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; New Cornerstone Science Laboratory, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaoyu Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Lei Li
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; New Cornerstone Science Laboratory, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - HanFei Deng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Song-Hai Shi
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; New Cornerstone Science Laboratory, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Kira E Poskanzer
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA
| | - David A Lyons
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4SB, UK
| | - Axel Nimmerjahn
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Misha B Ahrens
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| | - Guoqiang Yu
- Department of Automation, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; Beijing National Research Center for Information Science and Technology, Beijing 100084, China.
| |
Collapse
|
14
|
Gao K, Cheung-Hoi Yu A. Glutamate, a Key for Astrocytes to Participate in Brain Function and Diseases. Neurochem Res 2025; 50:166. [PMID: 40372498 DOI: 10.1007/s11064-025-04418-7] [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: 02/19/2025] [Revised: 05/01/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025]
Abstract
Astrocytes support neurons by maintaining health, regulating the environment, and aiding synaptic transmission, while glutamate is vital for excitatory signaling in learning and memory. The "glutamate-glutamine cycle," verified by Hertz and Schousboe, illustrates the interaction between neurons and astrocytes in glutamate regulation. Recent studies show astrocytes not only manage glutamate levels but also influence synaptic activity through gliotransmission and contribute to brain energy via glutamate metabolism. Dysregulation of this signaling is linked to neurological disorders like epilepsy and Alzheimer's. This mini-review will explore research progress on astrocytes and glutamate, highlighting glutamate's role in brain function and disease.
Collapse
Affiliation(s)
- Kai Gao
- Children's Medical Center, Peking University First Hospital, No.5 Le Yuan Road, Daxing District, Beijing, 102627, China.
| | - Albert Cheung-Hoi Yu
- Hai Kang Life Corporation Ltd, Units 15-18, 16/F South Wing Delta House, 3 On Yiu Street, Shatin, N.T., Hong Kong, China.
- Neuroscience Research Institute, Peking University, Beijing, China.
| |
Collapse
|
15
|
Wu CH, Camelot L, Lecca S, Mameli M. Neuromodulatory signaling contributing to the encoding of aversion. Trends Neurosci 2025:S0166-2236(25)00078-5. [PMID: 40318995 DOI: 10.1016/j.tins.2025.04.003] [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: 02/05/2025] [Revised: 03/28/2025] [Accepted: 04/08/2025] [Indexed: 05/07/2025]
Abstract
The appropriate and rapid encoding of stimuli bearing a negative valence enables behaviors that are essential for survival. Recent advances in neuroscience using rodents as a model system highlight the relevance of cell type-specific neuronal activities in diverse brain networks for the encoding of aversion, as well as their importance for subsequent behavioral strategies. Within these networks, neuromodulators influence cell excitability, adjust fast synaptic neurotransmission, and affect plasticity, ultimately modulating behaviors. In this review we first discuss contemporary findings leveraging the use of cutting-edge neurotechnologies to define aversion-related neural circuits. The spatial and temporal dynamics of the release of neuromodulators and neuropeptides upon exposure to aversive stimuli are described within defined brain circuits. Together, these mechanistic insights update the present neural framework through which aversion drives motivated behaviors.
Collapse
Affiliation(s)
- Cheng-Hsi Wu
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| | - Léa Camelot
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| | - Salvatore Lecca
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| | - Manuel Mameli
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMRS) 839, 75005 Paris, France.
| |
Collapse
|
16
|
Kettenmann H, Ugursu B, Ransom BR, Steinhäuser C. The Concept of Neuroglia - the State of the Art Circa 1900. Glia 2025; 73:890-904. [PMID: 39902825 PMCID: PMC11920685 DOI: 10.1002/glia.24678] [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: 10/01/2024] [Revised: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Glial cells were first defined by Rudolf Virchow in 1856. About 40 years later, glial research had developed into a field distinct from the mainstream study of neurons as the central elements governing brain function. By that time, substantial knowledge about the properties of glial cells had accumulated, exemplified by five important publications by four distinguished investigators: Gustav Retzius, Michael von Lenhossek, Carl Weigert, and Hans Held. These treatises broadly summarized what was known about glial cells, comparing findings from leeches to humans. Practically speaking, these articles represent the foundation of our current knowledge. All five contributions were published in German, which at the time was one of the dominant languages for scientific exchange. This article summarizes and comments on their findings and thus provides insight into what was known about glial cells at that time. More importantly, in the Supporting Information, we provide English translations and original scans of these five publications, making them accessible to an international readership.
Collapse
Affiliation(s)
- Helmut Kettenmann
- Department of NeuroscienceShenzhen University of Advanced TechnologyShenzhenChina
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
| | - Bilge Ugursu
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
| | | | | |
Collapse
|
17
|
Delcourte S, Bouloufa A, Rovera R, Brunet E, Le HD, Williams AE, Panda S, Azmani R, Raineteau O, Dkhissi-Benyahya O, Haddjeri N. Lateral habenula astroglia modulate the potentiating antidepressant-like effects of bright light stimulation in intractable depression. Front Pharmacol 2025; 16:1592909. [PMID: 40337515 PMCID: PMC12055791 DOI: 10.3389/fphar.2025.1592909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/07/2025] [Indexed: 05/09/2025] Open
Abstract
Background Beside image vision, light plays a pivotal role in regulating diverse non-visual functions, including affective behaviors. Recently, bright light stimulation (BLS) was revealed to be beneficial for treating non-seasonal depression, although its mechanism of action is not fully understood. Methods We developed a novel mouse model of refractory depression, induced through social isolation and chronic despair during the active (dark) phase of the animal, and we have tested if antidepressant treatments, including BLS, could protect against anxio-depressive-like behavior. Results We report that anxiety- and depressive-like behaviors are resistant to BLS as well as to both conventional and new antidepressants, including ketamine. Remarkably, we unveil that BLS potentiates the effect of antidepressants, and this beneficial effect is mediated via rod retinal photoreceptors. Furthermore, we demonstrate that both chemogenetic activation of lateral habenula (LHb) astroglia and serotonin (5-HT) depletion prevent the potentiating effect of BLS on chronic despair. Conclusion These results reveal, for the first time, that BLS enhances the efficacy of antidepressants through an unexpectedly circuit involving rods, LHb astroglia and 5-HT.
Collapse
Affiliation(s)
- Sarah Delcourte
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm U1208, Stem Cell and Brain Research Institute, Bron, France
| | - Amel Bouloufa
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm U1208, Stem Cell and Brain Research Institute, Bron, France
| | - Renaud Rovera
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm U1208, Stem Cell and Brain Research Institute, Bron, France
| | - Elie Brunet
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm U1208, Stem Cell and Brain Research Institute, Bron, France
| | - Hiep D. Le
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, United States
| | - April E. Williams
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Satchidananda Panda
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Rihab Azmani
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm U1208, Stem Cell and Brain Research Institute, Bron, France
| | - Olivier Raineteau
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm U1208, Stem Cell and Brain Research Institute, Bron, France
| | - Ouria Dkhissi-Benyahya
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm U1208, Stem Cell and Brain Research Institute, Bron, France
| | - Nasser Haddjeri
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm U1208, Stem Cell and Brain Research Institute, Bron, France
| |
Collapse
|
18
|
Cobo CB, Robertson F, Kangwa T, Annandale J, Subramoney S, Narr K, Joshi S, Hoffman N, Zar H, Stein D, Donald K, Wedderburn C, Naudé P. Maternal and child immune profiles are associated with neurometabolite measures of early-life neuroinflammation in children who are HIV-exposed and uninfected: a South African birth cohort. RESEARCH SQUARE 2025:rs.3.rs-6169072. [PMID: 40195997 PMCID: PMC11975042 DOI: 10.21203/rs.3.rs-6169072/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Children who are HIV-exposed and uninfected (HEU) are at risk of neurodevelopmental delays, which may be partially due to maternal immune dysregulation during pregnancy. This study investigates associations between maternal and child immune profiles and early neurometabolite profiles in HEU and HIV-unexposed (HU) children from a South African birth cohort. A subgroup of 156 children (66 HEU, 90 HU) from the Drakenstein Child Health Study underwent magnetic resonance spectroscopy at age 2-3 years, and maternal and child serum markers were measured at multiple timepoints via immunoassays. In HEU children, serum concentrations of maternal pro-inflammatory cytokines IL-5 (β = 0.79, p = 0.005) and IL-8 (β = 0.64, p = 0.02) were associated with myo-inositol ratios in parietal grey and white matter regions, respectively, while child serum MMP-9 at two years was associated with myo-inositol ratios in the midline parietal grey matter (β = 1.30, p = 0.03). The association of maternal anti-inflammatory cytokine IL-13 with glutamate ratios in the midline parietal grey matter was negative in HEU (β=-0.41, p = 0.038) and positive in HU children (β = 0.42, p < 0.0001). These findings suggest maternal immune activation may affect neurometabolite profiles in HEU children.
Collapse
|
19
|
Ziebarth T, Pape N, Nelson JS, van Alphen FI, Kalia M, Meijer HG, Rose CR, Reiner A. Atypical plume-like events contribute to glutamate accumulation in metabolic stress conditions. iScience 2025; 28:112256. [PMID: 40241754 PMCID: PMC12002667 DOI: 10.1016/j.isci.2025.112256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 12/02/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Neural glutamate homeostasis is important for health and disease. Ischemic conditions, like stroke, cause imbalances in glutamate release and uptake due to energy depletion and depolarization. We here used the glutamate sensor SF-iGluSnFR(A184V) to probe how chemical ischemia affects the extracellular glutamate dynamics in slice cultures from mouse cortex. SF-iGluSnFR imaging showed spontaneous glutamate release indicating synchronous network activity, similar to calcium imaging with GCaMP6f. Glutamate imaging further revealed local, atypically large, and long-lasting plume-like release events. Plumes occurred with low frequency, independent of network activity, and persisted in tetrodotoxin (TTX). Blocking glutamate uptake with TFB-TBOA favored plumes, whereas blocking ionotropic glutamate receptors (iGluRs) suppressed plumes. During chemical ischemia plumes became more pronounced, overly abundant and contributed to large-scale glutamate accumulation. Similar plumes were previously observed in cortical spreading depression and migraine models, and they may thus be a more general consequence of glutamate uptake dysfunctions in neurological and neurodegenerative diseases.
Collapse
Affiliation(s)
- Tim Ziebarth
- Department of Biology and Biotechnology, Ruhr University Bochum, Universitätsstrasse 150, 44801 Bochum, Germany
| | - Nils Pape
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Joel S.E. Nelson
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Fleur I.M. van Alphen
- Department of Applied Mathematics, University of Twente, Drienerlolaan 5, 7522 NB Enschede, the Netherlands
| | - Manu Kalia
- Department of Applied Mathematics, University of Twente, Drienerlolaan 5, 7522 NB Enschede, the Netherlands
| | - Hil G.E. Meijer
- Department of Applied Mathematics, University of Twente, Drienerlolaan 5, 7522 NB Enschede, the Netherlands
| | - Christine R. Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Andreas Reiner
- Department of Biology and Biotechnology, Ruhr University Bochum, Universitätsstrasse 150, 44801 Bochum, Germany
| |
Collapse
|
20
|
Liu M, Li M, Du H, Xu D, Wang J, Ren Q, Wang R, Gong H, Liu Y, Qi K, Tao J, Xia S, Wang H, Li X, Liu Q. The alteration of glutamate involved in the brain of Parkinson's disease patients using glutamate chemical exchange saturation transfer (GluCEST). Behav Brain Res 2025; 483:115484. [PMID: 39955039 DOI: 10.1016/j.bbr.2025.115484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/03/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Increased levels of glutamate, a novel regulator of neuroinflammation, is involved in the pathogenesis of Parkinson's disease (PD). Although glutamate chemical exchange saturation transfer (GluCEST) is widely used in central nervous system (CNS) disorders, it has been less commonly used in clinical practice for PD. Here, to explore the clinical significance of variations in glutamate levels in the striatum and thalamus in PD, we included forty-nine PD patients and forty-four healthy controls (HCs). Glutamate levels were analyzed by performing magnetization transfer ratio asymmetry (MTRasym) using GluCEST data. Four regions of interest (ROIs) were manually outlined on GluCEST images, and MTRasym values were calculated for each. FreeSurfer was used to calculate the volumes. We found that MTRasym values in the striatum and thalamus were elevated in PD. Variations in MTRasym values were correlated with motor scores. It has been found that the volume of the left pallidal nucleus were reduced in PD. The glutamate levels in the striatum and thalamus were significantly different from those in HCs and associated with disease progression. Collectively, glutamate metabolic abnormalities may be present in PD pathophysiology and associated with disease progression. GluCEST imaging may have potential to become an imaging technology for measuring glutamate alterations in the striatum and thalamus in PD.
Collapse
Affiliation(s)
- Miaomiao Liu
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Minglong Li
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Hailing Du
- Department of Emergency, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou 256603, China
| | - Donghao Xu
- Department of Radiology, Shengli Oilfield Central Hospital, Dongying 247034, China
| | - Jing Wang
- Department of Radiology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou 256603, China
| | - Qingfa Ren
- Department of Radiology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou 256603, China
| | - Rui Wang
- Department of Neurology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou 256603, China
| | - He Gong
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Yuwei Liu
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Kai Qi
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Jin Tao
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Shuyuan Xia
- Department of Radiology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou 256603, China
| | - Hongcai Wang
- Department of Neurology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou 256603, China.
| | - Xianglin Li
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China.
| | - Quanyuan Liu
- Department of Radiology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou 256603, China.
| |
Collapse
|
21
|
Joyce MKP, Datta D, Arellano JI, Duque A, Morozov YM, Morrison JH, Arnsten AFT. Contrasting patterns of extrasynaptic NMDAR-GluN2B expression in macaque subgenual cingulate and dorsolateral prefrontal cortices. Front Neuroanat 2025; 19:1553056. [PMID: 40255911 PMCID: PMC12006084 DOI: 10.3389/fnana.2025.1553056] [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: 12/30/2024] [Accepted: 03/19/2025] [Indexed: 04/22/2025] Open
Abstract
Expression of the N-methyl-D-aspartate receptor, particularly when containing the GluN2B subunit (NMDAR-GluN2B), varies across the prefrontal cortex (PFC). In humans, the subgenual cingulate cortex (SGC) contains among the highest levels of NMDAR-GluN2B expression, while the dorsolateral prefrontal cortex (dlPFC) exhibits a more moderate level of NMDAR-GluN2B expression. NMDAR-GluN2B are commonly associated with ionotropic synaptic function and plasticity and are essential to the neurotransmission underlying working memory in the macaque dlPFC in the layer III circuits, which in humans are afflicted in schizophrenia. However, NMDAR-GluN2B can also be found at extrasynaptic sites, where they may trigger distinct events, including some linked to neurodegenerative processes. The SGC is an early site of tau pathology in sporadic Alzheimer's disease (sAD), which mirrors its high NMDAR-GluN2B expression. Additionally, the SGC is hyperactive in depression, which can be treated with NMDAR antagonists. Given the clinical relevance of NMDAR in the SGC and dlPFC, the current study used immunoelectron microscopy (immunoEM) to quantitatively compare the synaptic and extrasynaptic expression patterns of NMDAR-GluN2B across excitatory and inhibitory neuron dendrites in rhesus macaque layer III SGC and dlPFC. We found a larger population of extrasynaptic NMDAR-GluN2B in dendrites of putative pyramidal neurons in SGC as compared to the dlPFC, while the dlPFC had a higher proportion of synaptic NMDAR-GluN2B. In contrast, in putative inhibitory dendrites from both areas, extrasynaptic expression of NMDAR-GluN2B was far more frequently observed over synaptic expression. These findings may provide insight into varying cortical vulnerability to alterations in excitability and neurodegenerative forces.
Collapse
Affiliation(s)
- Mary Kate P. Joyce
- Department of Neuroscience, Yale Medical School, New Haven, CT, United States
| | - Dibyadeep Datta
- Department of Psychiatry, Yale Medical School, New Haven, CT, United States
| | - Jon I. Arellano
- Department of Neuroscience, Yale Medical School, New Haven, CT, United States
| | - Alvaro Duque
- Department of Neuroscience, Yale Medical School, New Haven, CT, United States
| | - Yury M. Morozov
- Department of Neuroscience, Yale Medical School, New Haven, CT, United States
| | - John H. Morrison
- Department of Neurology, University of California, Davis, Davis, CA, United States
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Amy F. T. Arnsten
- Department of Neuroscience, Yale Medical School, New Haven, CT, United States
| |
Collapse
|
22
|
Chen S, Bao Q, Xu W, Zhai X. Extracellular particles: emerging insights into central nervous system diseases. J Nanobiotechnology 2025; 23:263. [PMID: 40170148 PMCID: PMC11960037 DOI: 10.1186/s12951-025-03354-6] [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: 01/13/2025] [Accepted: 03/24/2025] [Indexed: 04/03/2025] Open
Abstract
Extracellular particles (EPs), including extracellular vesicles (EVs) and non-vesicular extracellular particles (NVEPs), are multimolecular biomaterials released by cells that play a crucial role in intercellular communication. Recently, new subtypes of EPs associated with central nervous system (CNS), such as exophers and supermeres have been identified. These EPs provide new perspectives for understanding the pathological progression of CNS disorders and confer potential diagnostic value for liquid biopsies in neurodegenerative diseases (NDs). Moreover, EPs have emerged as promising drug delivery vehicles and targeted platforms for CNS-specific therapies. In this review, we delineate the landscape of EP subtypes and their roles in the pathophysiology of CNS diseases. We also review the recent advances of EP-based diagnosis in NDs and highlight the importance of analytical platforms with single-particle resolution in the exploitation of potential biomarkers. Furthermore, we summarize the application of engineered EVs in the treatment of CNS diseases and outline the underexplored potential of NVEPs as novel therapeutic agents.
Collapse
Affiliation(s)
- Shenyuan Chen
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Suzhou, Jiangsu, 215600, China
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Qinghua Bao
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Suzhou, Jiangsu, 215600, China
| | - Wenrong Xu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Suzhou, Jiangsu, 215600, China.
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China.
| | - Xiao Zhai
- Department of Orthopedics, Shanghai Changhai Hospital, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
23
|
Wu Z, Xu L, Xie Y, Sambangi A, Swaminathan S, Pei Z, Ji W, Li Z, Guo Y, Li Z, Chen G. Brain-Wide Neuroregenerative Gene Therapy Improves Cognition in a Mouse Model of Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410080. [PMID: 39951299 PMCID: PMC11984881 DOI: 10.1002/advs.202410080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 01/09/2025] [Indexed: 04/12/2025]
Abstract
Alzheimer's disease (AD) is a progressive and irreversible brain disorder with extensive neuronal loss in the neocortex and hippocampus. Current therapeutic interventions focus on the early stage of AD but lack effective treatment for the late stage of AD, largely due to the inability to replenish the lost neurons and repair the broken neural circuits. In this study, by using engineered adeno-associated virus vectors that efficiently cross the blood-brain-barrier in the mouse brain, a brain-wide neuroregenerative gene therapy is developed to directly convert endogenous astrocytes into functional neurons in a mouse model of AD. It is found that ≈500 000 new neurons are regenerated and widely distributed in the cerebral cortex and hippocampus. Importantly, it is demonstrated that the converted neurons can integrate into pre-existing neural networks and improve various cognitive performances in AD mice. Chemogenetic inhibition of the converted neurons abolishes memory enhancement in AD mice, suggesting a pivotal role for the newly converted neurons in cognitive restoration. Together, brain-wide neuroregenerative gene therapy may provide a viable strategy for the treatment of AD and other brain disorders associated with massive neuronal loss.
Collapse
Affiliation(s)
- Zheng Wu
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
- Department of BiologyHuck Institutes of Life SciencesPennsylvania State UniversityUniversity ParkPA16802USA
| | - Liang Xu
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Yu Xie
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Abhijeet Sambangi
- Department of BiologyHuck Institutes of Life SciencesPennsylvania State UniversityUniversity ParkPA16802USA
| | - Shreya Swaminathan
- Department of BiologyHuck Institutes of Life SciencesPennsylvania State UniversityUniversity ParkPA16802USA
| | - Zifei Pei
- Department of BiologyHuck Institutes of Life SciencesPennsylvania State UniversityUniversity ParkPA16802USA
| | - Wenyu Ji
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Zeru Li
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Yaowei Guo
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Zhifei Li
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Gong Chen
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
- Department of BiologyHuck Institutes of Life SciencesPennsylvania State UniversityUniversity ParkPA16802USA
| |
Collapse
|
24
|
Wang Y, Zhang M, Zhang T, Zhang S, Ji F, Qin J, Li H, Jiao J. PD-L1/PD-1 checkpoint pathway regulates astrocyte morphogenesis and myelination during brain development. Mol Psychiatry 2025:10.1038/s41380-025-02969-3. [PMID: 40164696 DOI: 10.1038/s41380-025-02969-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/05/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Programmed cell death protein 1 (PD-1) and its primary ligand PD-L1 are integral components of a significant immune checkpoint pathway, widely recognized for its central role in cancer immunotherapy. However, emerging evidence highlights their broader involvement in both the central and peripheral nervous systems. In this study, we demonstrate that PD-L1/PD-1 signaling in astrocytes during mouse brain development regulates astrocyte maturation and morphogenesis via the MEK/ERK pathway by targeting the downstream effector cysteine and glycine rich protein 1 (CSRP1). This enhanced astrocyte morphological complexity results in increased end-foot coverage of blood vessels. Additionally, aberrant secretion of CSRP1 by astrocytes interacts with oligodendrocyte precursor cells (OPCs) membrane proteins annexin A1 (ANXA1) and annexin A2 (ANXA2), leading to the exclusion of migrating OPCs from blood vessels. This disruption in OPC migration and differentiation results in abnormal myelination and is associated with cognitive deficits in the mice. Our results provide critical insights into the function of PD-L1/PD-1 signaling in astrocyte-OPC interactions and underscore its relevance to glial cell development and pathogenesis in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yanyan Wang
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mengtian Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tianyu Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shukui Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fen Ji
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jie Qin
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hong Li
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jianwei Jiao
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
25
|
Hastings N, Rahman S, Stempor PA, Wayland MT, Kuan WL, Kotter MRN. Connexin 43 is downregulated in advanced Parkinson's disease in multiple brain regions which correlates with symptoms. Sci Rep 2025; 15:10250. [PMID: 40133513 PMCID: PMC11937269 DOI: 10.1038/s41598-025-94188-7] [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: 12/10/2024] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative condition with the greatest increase in disability globally. Dysfunction of dopaminergic neurons is a well-known PD hallmark; however, changes in astrocytes also accompany PD progression. One aspect of astrocyte biology not yet investigated in PD is their network coupling. To assess this, we focussed on the major astrocytic gap junctional protein connexin 43 (Cx43, GJA1). A dataset of 20 post-mortem late-stage PD brain tissue samples from the cortex and basal ganglia alongside 20 age-matched control sets was collected, accompanied by clinical histories and data on α-synuclein, tau, and amyloid-β pathology. Protein levels and intracellular distribution of Cx43 and other key markers were measured. Computational re-analysis of open-source mRNA sequencing datasets from the striatum and midbrain complemented the original findings. Two novel observations were made: first, profound Cx43 loss in late-stage PD, and second, differential manifestation of this pathology in different brain areas, including those outside of the midbrain substantia nigra-the region that is most commonly used in PD research. Cx43 downregulation in specific regions correlated with non-motor symptoms of PD such as depression and sleep disturbance. Astrocytic tree simplification in the frontal cortex was further observed. In conclusion, astrocytic network decoupling through Cx43 downregulation in PD may contribute to astrocytic dysfunction and PD symptom development.
Collapse
Affiliation(s)
- Nataly Hastings
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK.
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0AW, UK.
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, CB3 0FA, UK.
| | - Saifur Rahman
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0AW, UK
| | | | - Matthew T Wayland
- Department of Zoology, University of Cambridge, Cambridge, CB2 3EJ, UK
| | - Wei-Li Kuan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
- Alborada Drug Discovery Institute, University of Cambridge, Cambridge, CB2 0AH, UK
| | - Mark R N Kotter
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0AW, UK
| |
Collapse
|
26
|
Bertran-Cobo C, Robertson FC, Kangwa TS, Annandale J, Subramoney S, Narr KL, Joshi SH, Hoffman N, Zar HJ, Stein DJ, Donald KA, Wedderburn CJ, Naudé PJW. Maternal and child immune profiles are associated with neurometabolite measures of early-life neuroinflammation in children who are HIV-exposed and uninfected: a South African birth cohort. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.17.643628. [PMID: 40166324 PMCID: PMC11957058 DOI: 10.1101/2025.03.17.643628] [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: 04/02/2025]
Abstract
Children who are HIV-exposed and uninfected (HEU) are at risk of neurodevelopmental delays, which may be partially due to maternal immune dysregulation during pregnancy. This study investigates associations between maternal and child immune profiles and early neurometabolite profiles in HEU and HIV-unexposed (HU) children from a South African birth cohort. A subgroup of 156 children (66 HEU, 90 HU) from the Drakenstein Child Health Study underwent magnetic resonance spectroscopy at age 2-3 years, and maternal and child serum markers were measured at multiple timepoints via immunoassays. In HEU children, serum concentrations of maternal pro-inflammatory cytokines IL-5 (β=0.79, p=0.005) and IL-8 (β=0.64, p=0.02) were associated with myo-inositol ratios in parietal grey and white matter regions, respectively, while child serum MMP-9 at two years was associated with myo-inositol ratios in the midline parietal grey matter (β=1.30, p=0.03). The association of maternal anti-inflammatory cytokine IL-13 with glutamate ratios in the midline parietal grey matter was negative in HEU (β=-0.41, p=0.038) and positive in HU children (β=0.42, p<0.0001). These findings suggest maternal immune activation may affect neurometabolite profiles in HEU children.
Collapse
Affiliation(s)
- Cesc Bertran-Cobo
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Frances C Robertson
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Cape Universities Body Imaging Centre (CUBIC), Cape Town, South Africa
| | - Tusekile Sarah Kangwa
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Jenna Annandale
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Sivenesi Subramoney
- Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Cape Town, South Africa
| | - Katherine L Narr
- Departments of Neurology, Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, United States
| | - Shantanu H Joshi
- Departments of Neurology, Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nadia Hoffman
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Heather J Zar
- Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Cape Town, South Africa
- SAMRC Unit on Child & Adolescent Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Cape Town, South Africa
| | - Dan J Stein
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- SAMRC Unit on Risk and Resilience in Mental Disorders, University of Cape Town, Cape Town, South Africa
| | - Kirsten A Donald
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Cape Town, South Africa
| | - Catherine J Wedderburn
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Cape Town, South Africa
- Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Petrus J W Naudé
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
27
|
Koh ZM, Arceo RA, Hammer J, Chau K, Light SE, Dolojan A, Januszewski M, Svara F, Smith CJ. An ultrastructural map of a spinal sensorimotor circuit reveals the potential of astroglial modulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.05.641432. [PMID: 40093104 PMCID: PMC11908220 DOI: 10.1101/2025.03.05.641432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Information flow through circuits is dictated by the precise connectivity of neurons and glia. While a single astrocyte can contact many synapses, how glial-synaptic interactions are arranged within a single circuit to impact information flow remains understudied. Here, we use the local spinal sensorimotor circuit in zebrafish as a model to understand how neurons and astroglia are connected in a vertebrate circuit. With semi-automated cellular reconstructions and automated approaches to map all the synaptic connections, we identified the precise synaptic connections of the local sensorimotor circuit, from dorsal root ganglia neurons to spinal interneurons and finally to motor neurons. This revealed a complex network of interneurons that interact in the local sensorimotor circuit. We then mapped the glial processes within tripartite synapses in the circuit. We demonstrate that tripartite synapses are equally distributed across the circuit, supporting the idea that glia can modulate information flow through the circuit at different levels. We show that multiple astroglia, including bona fide astrocytes, contact synapses within a single sensory neuron's circuit and that each of these astroglia can contact multiple parts of the circuit. This detailed map reveals an extensive network of connected neurons and astroglia that process sensory stimuli in a vertebrate. We then utilized this ultrastructural map to model how synaptic thresholding and glial modulation could alter information flow in circuits. We validated this circuit map with GCaMP6s imaging of dorsal root ganglia, spinal neurons and astroglia. This work provides a foundational resource detailing the ultrastructural organization of neurons and glia in a vertebrate circuit, offering insights in how glia could influence information flow in complex neural networks.
Collapse
Affiliation(s)
- Zachary M. Koh
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine University of Notre Dame, Notre Dame, IN
| | - Ricky Avalos Arceo
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine University of Notre Dame, Notre Dame, IN
| | - Jacob Hammer
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine University of Notre Dame, Notre Dame, IN
| | - Khang Chau
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine University of Notre Dame, Notre Dame, IN
| | - Sarah E.W. Light
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine University of Notre Dame, Notre Dame, IN
| | - Antonio Dolojan
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine University of Notre Dame, Notre Dame, IN
| | | | - Fabian Svara
- Zürich, Switzerland. ariadne.ai ag, Buchrain, Switzerland
| | - Cody J. Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine University of Notre Dame, Notre Dame, IN
| |
Collapse
|
28
|
Yang MA, Kang S, Hong SI, Lee J, Bormann NL, Lee SW, Choi DS. Astrocytes in the External Globus Pallidus Selectively Represent Routine Formation During Repeated Reward-Seeking in Mice. eNeuro 2025; 12:ENEURO.0552-24.2025. [PMID: 40032533 PMCID: PMC11913404 DOI: 10.1523/eneuro.0552-24.2025] [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: 12/03/2024] [Revised: 02/03/2025] [Accepted: 02/24/2025] [Indexed: 03/05/2025] Open
Abstract
The external globus pallidus (GPe) is a central part of the basal ganglia indirect pathway implicated in movement and decision-making. As a hub connecting the dorsal striatum and subthalamic nucleus (STN), the GPe guides repetitive and routine behaviors. However, it remains unknown how diverse GPe cells engage in routine formation while learning action sequences in repetitive reward-seeking conditioning. Here, in male mice, we investigated the Ca2+ dynamics of two GPe cell types, astrocytes and parvalbumin-expressing neurons, during routine formation. Our findings show that the dynamics of GPe astrocytes may be involved in action sequence refinement, a characteristic potentially contributing to more efficient reward-seeking behavior.
Collapse
Affiliation(s)
- Minsu Abel Yang
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Program of Brain and Cognitive Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Shinwoo Kang
- Department of Clinical Pharmacology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Sa-Ik Hong
- Department of Pharmacy, Pohang SM Christianity Hospital, Pohang 37816, Republic of Korea
| | - Jeyeon Lee
- Departments of Radiology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota 55905
| | - Nicholas L Bormann
- Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota 55905
| | - Sang Wan Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Program of Brain and Cognitive Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Department of Brain & Cognitive Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Kim Jaechul Graduate School of AI, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Doo-Sup Choi
- Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota 55905
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, Minnesota 55905
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, Minnesota 55905
| |
Collapse
|
29
|
Tang Z, Lu H, Yang X, Wu M, Yang J, Li S, Liu H, Zhou J, Tang B, Du X, Xu F, Shao Y, Wang J. Single-cell RNA sequencing provides new insights into the interaction between astrocytes and neurons after spinal cord injury in mice. Biochem Biophys Rep 2025; 41:101917. [PMID: 39896108 PMCID: PMC11787598 DOI: 10.1016/j.bbrep.2025.101917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
Background Spinal cord injury (SCI) is a devastating neurological disease in which astrocytes play a central role. Understanding the relationship between different subtypes of astrocytes and neuron subtypes during the progression of SCI is critical to understanding the disease. Methods and results In this study, single-cell RNA sequencing (scRNA-seq) was used to analyze the transcriptome data of acute, subacute and intermediate stages of SCI in mice as well as normal tissues. Different subtypes of astrocytes and neuronal cells were identified and their dynamic changes and functionalities during the development of SCI. An intriguing discovery was the identification of a specific subtype of astrocytes characterized by unique expression of Gap43, Vim, Aldoc, and Mt1. This subtype of cells shows similarities in gene expression with neurons and potentially transitioned into neurons during the course of SCI. Furthermore, we have uncovered the important role of the glycolytic pathway in this cellular transformation process. Furthermore, through cellular interaction analysis, we validated pathways (mdk-ptprz1,ptn-ptprz1,ptn-sdc3) associated with the potential conversion of these specific cell subsets into neurons. Finally, these cells were observed by fluorescence microscopy and critical gene expressions were validated by Western blot. Conclusions The results of this study not only deepen our understanding of the mechanisms underlying SCI, but also provide new insights and opportunities for the development of novel therapeutic strategies and interventions.
Collapse
Affiliation(s)
- Zhi Tang
- Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Hengyang Lu
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
- The PRC Ministry of Education Engineering Research Center of Intelligent Technology for Healthcare, Wuxi, Jiangsu 214122, China
| | - Xiao Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Mao Wu
- Department of Orthopaedics and Traumatology, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Junfeng Yang
- Department of Orthopaedics and Traumatology, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Shaoshuo Li
- Department of Orthopaedics and Traumatology, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Heng Liu
- Wuxi City Binhu Traditional Chinese Medicine Hospital, China
| | - Junkang Zhou
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Bin Tang
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Xinyao Du
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Fei Xu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yang Shao
- Department of Orthopaedics and Traumatology, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Jianwei Wang
- Department of Orthopaedics and Traumatology, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
30
|
Zhu CC, Zheng YL, Gong C, Chen BL, Guo JB. Role of Exercise on Neuropathic Pain in Preclinical Models: Perspectives for Neuroglia. Mol Neurobiol 2025; 62:3684-3696. [PMID: 39316356 DOI: 10.1007/s12035-024-04511-y] [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: 11/03/2023] [Accepted: 09/15/2024] [Indexed: 09/25/2024]
Abstract
The benefits of exercise on neuropathic pain (NP) have been demonstrated in numerous studies. In recent studies, inflammation, neurotrophins, neurotransmitters, and endogenous opioids are considered as the main mechanisms. However, the role of exercise in alleviating NP remains unclear. Neuroglia, widely distributed in both the central and peripheral nervous systems, perform functions such as support, repair, immune response, and maintenance of normal neuronal activity. A large number of studies have shown that neuroglia play an important role in the occurrence and development of NP, and exercise can alleviate NP by regulating neuroglia. This article reviewed the involvement of neuroglia in the development of NP and their role in the exercise treatment of NP, intending to provide a theoretical basis for the exercise treatment strategy of NP.
Collapse
Affiliation(s)
- Chen-Chen Zhu
- The Second School of Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- School of Rehabilitation Medicine, Nanjing Medical University, Nanjing, China
| | - Yi-Li Zheng
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, 200438, China
| | - Chan Gong
- The Second School of Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- School of Rehabilitation Medicine, Nanjing Medical University, Nanjing, China
| | - Bing-Lin Chen
- The Second School of Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Jia-Bao Guo
- The Second School of Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
31
|
Sona C, Yeh YT, Li Y, Liu X, Ghosh A, Hinte LC, Ku MC, Rathjen T, Niendorf T, Yu G, Jia S, Kononenko NL, Hermann A, Luo J, Lin J, von Meyenn F, Yan X, Poy MN. Glutamatergic argonaute2 promotes the formation of the neurovascular unit in mice. Sci Signal 2025; 18:eadl6745. [PMID: 39999211 DOI: 10.1126/scisignal.adl6745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 11/08/2024] [Accepted: 01/28/2025] [Indexed: 02/27/2025]
Abstract
Proper formation of the complex neurovascular unit (NVU) along with the blood-brain barrier is critical for building and sustaining a healthy, functioning central nervous system. The RNA binding protein argonaute2 (Ago2) mediates microRNA (miRNA)-mediated gene silencing, which is critical for many facets of brain development, including NVU development. Here, we found that Ago2 in glutamatergic neurons was critical for NVU formation in the developing cortices of mice. Glutamatergic neuron-specific loss of Ago2 diminished synaptic formation, neuronal-to-endothelial cell contacts, and morphogenesis of the brain vasculature, ultimately compromising the integrity of the blood-brain barrier. Ago2 facilitated miRNA targeting of phosphatase and tensin homolog (Pten) mRNA, which encodes a phosphatase that modulates reelin-dependent phosphatidylinositol 3-kinase (PI3K)-Akt signaling within the glutamatergic subpopulation. Conditionally deleting Pten in Ago2-deficient neurons restored Akt2 phosphorylation as well as postnatal development and survival. Several mutations in AGO2 impair small RNA silencing and are associated with Lessel-Kreienkamp syndrome, a neurodevelopmental disorder. When expressed in a neuronal cell line, these human AGO2 loss-of-function variants failed to suppress PTEN, resulting in attenuated PI3K-Akt signaling, further indicating that dysregulation of Ago2 function may contribute to both impaired development and neurological disorders. Together, these results identify Ago2 as central to the engagement of neurons with blood vessels in the developing brain.
Collapse
Affiliation(s)
- Chandan Sona
- All Children's Hospital, Johns Hopkins University, St. Petersburg, FL 33701, USA
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Yu-Te Yeh
- All Children's Hospital, Johns Hopkins University, St. Petersburg, FL 33701, USA
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Yunxiao Li
- Translational Neurodegeneration Section, "Albrecht Kossel," University Medical Center Rostock, University of Rostock, Rostock 18147, Germany
| | - Xiaoxuan Liu
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Adhideb Ghosh
- Laboratory of Nutrition and Metabolic Epigenetics, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach 8603, Switzerland
| | - Laura C Hinte
- Laboratory of Nutrition and Metabolic Epigenetics, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach 8603, Switzerland
| | - Min-Chi Ku
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, Berlin 13125, Germany
| | - Thomas Rathjen
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, Berlin 13125, Germany
| | - Thoralf Niendorf
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, Berlin 13125, Germany
| | - Guoxing Yu
- Institutes of Biomedical Sciences, College of Life Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Shiqi Jia
- Institutes of Biomedical Sciences, College of Life Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Natalia L Kononenko
- CECAD Excellence Center & Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne 50931, Germany
| | - Andreas Hermann
- Translational Neurodegeneration Section, "Albrecht Kossel," University Medical Center Rostock, University of Rostock, Rostock 18147, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, Rostock 18147, Germany
- German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Rostock 17489, Germany
| | - Jiankai Luo
- Translational Neurodegeneration Section, "Albrecht Kossel," University Medical Center Rostock, University of Rostock, Rostock 18147, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, Rostock 18147, Germany
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Ferdinand von Meyenn
- Laboratory of Nutrition and Metabolic Epigenetics, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach 8603, Switzerland
| | - Xin Yan
- Translational Neurodegeneration Section, "Albrecht Kossel," University Medical Center Rostock, University of Rostock, Rostock 18147, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, Berlin 13125, Germany
| | - Matthew N Poy
- All Children's Hospital, Johns Hopkins University, St. Petersburg, FL 33701, USA
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University, Baltimore, MD 21287, USA
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, Berlin 13125, Germany
| |
Collapse
|
32
|
Shen W, Chen F, Tang Y, Zhao Y, Zhu L, Xiang L, Ning L, Zhou W, Chen Y, Wang L, Li J, Huang H, Zeng LH. mGluR5-mediated astrocytes hyperactivity in the anterior cingulate cortex contributes to neuropathic pain in male mice. Commun Biol 2025; 8:266. [PMID: 39979531 PMCID: PMC11842833 DOI: 10.1038/s42003-025-07733-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 02/13/2025] [Indexed: 02/22/2025] Open
Abstract
Astrocytes regulate synaptic transmission in healthy and pathological conditions, but their involvement in modulating synaptic transmission in chronic pain is unknown. Our study demonstrates that astrocytes in the anterior cingulate cortex (ACC) exhibit abnormal calcium signals and induce the release of glutamate in male mice. This leads to an elevation in extracellular glutamate concentration, activation of presynaptic kainate receptors, and an increase in synaptic transmission following neuropathic pain. We discovered that the abnormal calcium signals are caused by the reappearance of metabotropic glutamate receptor type 5 (mGluR5) in astrocytes in male mice. Importantly, when we specifically inhibit the Gq pathway using iβARK and reduce the expression of mGluR5 in astrocytes through shRNA, we observe a restoration of astrocytic calcium activity, normalization of synaptic transmission and extracellular concentration of glutamate, and improvement in mechanical allodynia in male mice. Furthermore, the activation of astrocytes through chemogenetics results in an overabundance of excitatory synaptic transmission, exacerbating mechanical allodynia in mice with neuropathic pain, but not in sham-operated male mice. In summary, our findings suggest that the abnormal calcium signaling in astrocytes, mediated by mGluR5, plays a crucial role in enhancing synaptic transmission in ACC and contributing to mechanical allodynia in male mice.
Collapse
Affiliation(s)
- Weida Shen
- Anji People's Hospital, Affiliated Anji Hospital, School of Medicine, Hangzhou City University, Hangzhou, China.
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
| | - Fujian Chen
- Anji People's Hospital, Affiliated Anji Hospital, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yejiao Tang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Yulu Zhao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Linjing Zhu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Liyang Xiang
- School of Medicine, Nankai University, Tianjin, China
| | - Li Ning
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wen Zhou
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yiran Chen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Liangxue Wang
- Anji People's Hospital, Affiliated Anji Hospital, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Jing Li
- Anji People's Hospital, Affiliated Anji Hospital, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Hui Huang
- Anji People's Hospital, Affiliated Anji Hospital, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Ling-Hui Zeng
- Anji People's Hospital, Affiliated Anji Hospital, School of Medicine, Hangzhou City University, Hangzhou, China.
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
| |
Collapse
|
33
|
Chen J, Yang L, Shen J, Lu J, Mo X, Huang L, Chen L, Yu C. Distinct Roles of Astrocytes and GABAergic Neurons in the Paraventricular Thalamic Nucleus in Modulating Diabetic Neuropathic Pain. J Neurosci 2025; 45:e1013242024. [PMID: 39622642 PMCID: PMC11841761 DOI: 10.1523/jneurosci.1013-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 02/21/2025] Open
Abstract
Diabetic neuropathic pain (DNP) is a common chronic complication of diabetes mellitus and a clinically common form of neuropathic pain. The thalamus is an important center for the conduction and modulation of nociceptive signals. The paraventricular thalamic nucleus (PVT) is an important midline nucleus of the thalamus involved in sensory processing, but the specific role of PVT astrocytes and GABAergic neurons in DNP remains unclear. Here, we examined the activity of PVT astrocytes and neurons at various time points during the development of DNP by fluorescence immunohistochemistry and found that the activity of PVT astrocytes was significantly increased while that of PVT neurons was significantly decreased 14 d after streptozotocin injection in male rats. The inhibition of PVT astrocytes by chemogenetic manipulation relieved mechanical allodynia in male DNP model rats, whereas the activation of PVT astrocytes induced mechanical allodynia in normal male rats. Interestingly, chemogenetic activation of GABAergic neurons in the PVT alleviated mechanical allodynia in male DNP model rats, whereas chemogenetic inhibition of GABAergic neurons in the PVT induced mechanical allodynia in normal male rats. These data demonstrate the distinct roles of PVT astrocytes and GABAergic neurons in modulating DNP, revealing the mechanism of DNP pathogenesis and the role of the PVT in pain modulation.
Collapse
Affiliation(s)
- Jian Chen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Lan Yang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Jinhuang Shen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Jingshan Lu
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou 350122, China
| | - Xiaona Mo
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Linyi Huang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Li Chen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Changxi Yu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| |
Collapse
|
34
|
Yang J, Agrawal K, Stanley J, Li R, Jacobs N, Wang H, Lu C, Qu R, Clarke D, Chen Y, Jiang Y, Bai D, Zheng S, Fox H, Ho YC, Huttner A, Gerstein M, Kluger Y, Zhang L, Spudich S. Multi-omic Characterization of HIV Effects at Single Cell Level across Human Brain Regions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636707. [PMID: 39975288 PMCID: PMC11839123 DOI: 10.1101/2025.02.05.636707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
HIV infection exerts profound and long-lasting neurodegenerative effects on the central nervous system (CNS) that can persist despite antiretroviral therapy (ART). Here, we used single-nucleus multiome sequencing to map the transcriptomic and epigenetic landscapes of postmortem human brains from 13 healthy individuals and 20 individuals with HIV who have a history of treatment with ART. Our study spanned three distinct regions-the prefrontal cortex, insular cortex, and ventral striatum-enabling a comprehensive exploration of region-specific and cross-regional perturbations. We found widespread and persistent HIV-associated transcriptional and epigenetic alterations across multiple cell types. Detailed analyses of microglia revealed state changes marked by immune activation and metabolic dysregulation, while integrative multiomic profiling of astrocytes identified multiple subpopulations, including a reactive subpopulation unique to HIV-infected brains. These findings suggest that cells from people with HIV exhibit molecular shifts that may underlie ongoing neuroinflammation and CNS dysfunction. Furthermore, cell-cell communication analyses uncovered dysregulated and pro-inflammatory interactions among glial populations, underscoring the multifaceted and enduring impact of HIV on the brain milieu. Collectively, our comprehensive atlas of HIV-associated brain changes reveals distinct glial cell states with signatures of proinflammatory signaling and metabolic dysregulation, providing a framework for developing targeted therapies for HIV-associated neurological dysfunction.
Collapse
Affiliation(s)
- Junchen Yang
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Kriti Agrawal
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Jay Stanley
- Program in Applied Mathematics, Yale University, New Haven, CT, USA
| | - Ruiqi Li
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Nicholas Jacobs
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Haowei Wang
- Department of Neurology, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale University, New Haven, CT, USA
| | - Chang Lu
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Rihao Qu
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Declan Clarke
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Yuhang Chen
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Yunzhe Jiang
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Donglu Bai
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Suchen Zheng
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Howard Fox
- Department of Neurological Sciences, University of Nebraska School of Medicine, Omaha, NB, USA
| | - Ya-chi Ho
- Department of Microbial Pathogenesis, Yale University, New Haven, CT, USA
| | - Anita Huttner
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Mark Gerstein
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Computer Science, Yale University, New Haven, CT, USA
- Department of Statistics and Data Science, Yale University, New Haven, CT, USA
- Department of Biomedical Informatics & Data Science, Yale University, New Haven, CT, USA
| | - Yuval Kluger
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Program in Applied Mathematics, Yale University, New Haven, CT, USA
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Le Zhang
- Department of Neurology, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale University, New Haven, CT, USA
| | - Serena Spudich
- Department of Neurology, Yale University, New Haven, CT, USA
- Center for Brain and Mind Health, Yale University, New Haven, CT, USA
| |
Collapse
|
35
|
Li S, Cai Y, Xia Z. Editorial: Function and regulation of non-neuronal cells in the nervous system. Front Cell Neurosci 2025; 19:1550903. [PMID: 39990969 PMCID: PMC11842420 DOI: 10.3389/fncel.2025.1550903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 01/10/2025] [Indexed: 02/25/2025] Open
Affiliation(s)
- Sisi Li
- Department of Anesthesiology, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Yin Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Zhengyuan Xia
- Department of Anesthesiology and Perioperative Medicine, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Doctoral Training Platform for Research and Translation, Shuanghe, Hubei, China
| |
Collapse
|
36
|
Joyce M, Datta D, Arellano J, Duque A, Morozov YM, Morrison JH, Arnsten A. Contrasting patterns of extrasynaptic NMDAR-GluN2B expression in macaque subgenual cingulate and dorsolateral prefrontal cortices. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636752. [PMID: 39975025 PMCID: PMC11839065 DOI: 10.1101/2025.02.05.636752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Expression of the N-methyl-D-aspartate receptor, particularly when containing the GluN2B subunit (NMDAR-GluN2B) varies across the prefrontal cortex (PFC). In humans, the subgenual cingulate cortex (SGC) contains among the highest levels of NMDAR-GluN2B expression, while the dorsolateral prefrontal cortex (dlPFC) exhibits a more moderate level of NMDAR-GluN2B expression. NMDAR-GluN2B are commonly associated with ionotropic synaptic function and plasticity, and are essential to the neurotransmission underlying working memory in the macaque dlPFC in the layer III circuits afflicted in schizophrenia. However, NMDAR-GluN2B can also be found at extrasynaptic sites, where they may trigger distinct events, including some linked to neurodegenerative processes. The SGC is an early site of tau pathology in sporadic Alzheimer's Disease (sAD), which mirrors its high NMDAR-GluN2B expression. Additionally, the SGC is hyperactive in depression, which is treated with NMDAR antagonists. Given the clinical relevance of NMDAR in the SGC and dlPFC, the current study used immunoelectron microscopy (immunoEM) to quantitatively compare the synaptic and extrasynaptic expression patterns of NMDAR-GluN2B across excitatory and inhibitory neuron dendrites in the rhesus macaque SGC and dlPFC. We found a larger population of extrasynaptic NMDAR-GluN2B in dendritic shafts and spines of putative pyramidal neurons in SGC as compared to the dlPFC, while the dlPFC had a higher proportion of synaptic NMDAR-GluN2B. In contrast, in putative inhibitory dendrites from both areas, extrasynaptic expression of NMDAR-GluN2B was far more frequently observed over synaptic expression. These findings may provide insight into varying cortical vulnerability to alterations in excitability and to neurodegenerative forces. Scope Statement NMDAR are ionotropic receptors that contribute to neurotransmission and second messenger signaling events. NMDAR can induce a diverse array of neuronal events, in part due to variation in subunit composition and subcellular localization of receptor expression. Expression of the GluN2B subunit varies across the prefrontal cortex in humans. This subunit is highly expressed in the subgenual cingulate, an area associated with mood and emotion, and more moderately expressed in the dorsolateral prefrontal cortex, an area associated with cognitive processes. Extrasynaptic NMDAR, which often contain with the GluN2B subunit, have been linked to detrimental cellular events like neurodegeneration. Here, using high resolution electron microscopy in rhesus macaques, we found evidence that extrasynaptic NMDAR-GluN2B expression may be more prominent in subgenual cortex than in the dorsolateral prefrontal cortex. Conversely, synaptic NMDAR-GluN2B may be more prominent in the dorsolateral prefrontal cortex, consistent with their essential contribution to neuronal firing during working memory. These findings may help to illuminate the propensity of the subgenual cortex to tonic hyperactivity in major depression and its vulnerability to neurodegeneration in Alzheimer's disease, and may help to explain how rapid acting antidepressants exert therapeutic action across diverse neural circuits.
Collapse
|
37
|
Liu Y, Wu L, Peng W, Mao X. Glial polarization in neurological diseases: Molecular mechanisms and therapeutic opportunities. Ageing Res Rev 2025; 104:102638. [PMID: 39672208 DOI: 10.1016/j.arr.2024.102638] [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: 09/28/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024]
Abstract
Glial cell polarization plays a pivotal role in various neurological disorders. In response to distinct stimuli, glial cells undergo polarization to either mitigate neurotoxicity or facilitate neural repair following injury, underscoring the importance of glial phenotypic polarization in modulating central nervous system function. This review presents an overview of glial cell polarization, focusing on astrocytes and microglia. It explores the involvement of glial polarization in neurological diseases such as Alzheimer's disease, Parkinson's disease, stroke, epilepsy, traumatic brain injury, amyotrophic lateral sclerosis, multiple sclerosis and meningoencephalitis. Specifically, it emphasizes the role of glial cell polarization in disease pathogenesis through mechanisms including neuroinflammation, neurodegeneration, calcium signaling dysregulation, synaptic dysfunction and immune response. Additionally, it summarizes various therapeutic strategies including pharmacological treatments, dietary supplements and cell-based therapies, aimed at modulating glial cell polarization to ameliorate brain dysfunction. Future research focused on the spatio-temporal manipulation of glial polarization holds promise for advancing precision diagnosis and treatment of neurological diseases.
Collapse
Affiliation(s)
- Yuqing Liu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Lei Wu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha 410008, China; Institute of Clinical Pharmacology and Engineering Research Center of Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | - Xiaoyuan Mao
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha 410008, China; Institute of Clinical Pharmacology and Engineering Research Center of Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China.
| |
Collapse
|
38
|
Akyuz E, Arulsamy A, Aslan FS, Sarisözen B, Guney B, Hekimoglu A, Yilmaz BN, Retinasamy T, Shaikh MF. An Expanded Narrative Review of Neurotransmitters on Alzheimer's Disease: The Role of Therapeutic Interventions on Neurotransmission. Mol Neurobiol 2025; 62:1631-1674. [PMID: 39012443 PMCID: PMC11772559 DOI: 10.1007/s12035-024-04333-y] [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: 07/06/2023] [Accepted: 06/24/2024] [Indexed: 07/17/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease. The accumulation of amyloid-β (Aβ) plaques and tau neurofibrillary tangles are the key players responsible for the pathogenesis of the disease. The accumulation of Aβ plaques and tau affect the balance in chemical neurotransmitters in the brain. Thus, the current review examined the role of neurotransmitters in the pathogenesis of Alzheimer's disease and discusses the alterations in the neurochemical activity and cross talk with their receptors and transporters. In the presence of Aβ plaques and neurofibrillary tangles, changes may occur in the expression of neuronal receptors which in turn triggers excessive release of glutamate into the synaptic cleft contributing to cell death and neuronal damage. The GABAergic system may also be affected by AD pathology in a similar way. In addition, decreased receptors in the cholinergic system and dysfunction in the dopamine neurotransmission of AD pathology may also contribute to the damage to cognitive function. Moreover, the presence of deficiencies in noradrenergic neurons within the locus coeruleus in AD suggests that noradrenergic stimulation could be useful in addressing its pathophysiology. The regulation of melatonin, known for its effectiveness in enhancing cognitive function and preventing Aβ accumulation, along with the involvement of the serotonergic system and histaminergic system in cognition and memory, becomes remarkable for promoting neurotransmission in AD. Additionally, nitric oxide and adenosine-based therapeutic approaches play a protective role in AD by preventing neuroinflammation. Overall, neurotransmitter-based therapeutic strategies emerge as pivotal for addressing neurotransmitter homeostasis and neurotransmission in the context of AD. This review discussed the potential for neurotransmitter-based drugs to be effective in slowing and correcting the neurodegenerative processes in AD by targeting the neurochemical imbalance in the brain. Therefore, neurotransmitter-based drugs could serve as a future therapeutic strategy to tackle AD.
Collapse
Affiliation(s)
- Enes Akyuz
- Department of Biophysics, International School of Medicine, University of Health Sciences, Istanbul, Turkey
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
| | | | - Bugra Sarisözen
- School of Medicine, Tekirdağ Namık Kemal University, Tekirdağ, Turkey
| | - Beyzanur Guney
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | | | - Beyza Nur Yilmaz
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange, New South Wales, 2800, Australia.
| |
Collapse
|
39
|
Marinelli S. BoNT/Action beyond neurons. Toxicon 2025; 255:108250. [PMID: 39862929 DOI: 10.1016/j.toxicon.2025.108250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025]
Abstract
Botulinum neurotoxin type A (BoNT/A) has expanded its therapeutic uses beyond neuromuscular disorders to include treatments for various pain syndromes and neurological conditions. Originally recognized for blocking acetylcholine release at neuromuscular junctions, BoNT/A's effects extend to both peripheral and central nervous systems. Its ability to undergo retrograde transport allows BoNT/A to modulate synaptic transmission and reduce pain centrally, influencing neurotransmitter systems beyond muscle control. BoNT/A also interacts with glial cells, such as Schwann cells, satellite glial cells, astrocytes, microglia, and oligodendrocytes. Schwann cells, key to peripheral nerve regeneration, are directly influenced by BoNT/A, which promotes their proliferation and enhances remyelination. Satellite glial cells, involved in sensory neuron regulation, show reduced glutamate release in response to BoNT/A, aiding in pain relief. In the CNS, BoNT/A modulates astrocyte activity, reducing excitotoxicity and inflammation, which is relevant in conditions like epilepsy. Microglia, the CNS's immune cells, shift from a pro-inflammatory to a neuroprotective state when treated with BoNT/A, enhancing tissue repair. Additionally, BoNT/A promotes oligodendrocyte survival and remyelination, especially after spinal cord injury. Overall, BoNT/A's ability to target both neurons and glial cells presents a multifaceted therapeutic strategy for neurological disorders, pain management, and CNS repair. Further research is necessary to fully elucidate its mechanisms and optimize its clinical application.
Collapse
Affiliation(s)
- Sara Marinelli
- National Research Council of Italy, Institute of Biochemistry and Cell Biology, 00015, Monterotondo, RM, Italy.
| |
Collapse
|
40
|
Huang Q, Lee HH, Volpe B, Zhang Q, Xue C, Liu BC, Abuhasan YR, Li L, Yang JS, Egholm J, Gutierrez-Vazquez C, Li A, Lee A, Tang S, Wong CW, Liu T, Huang Y, Ramos RL, Stout RF, El Ouaamari A, Quintana FJ, Lowell BB, Kahn CR, Pothos EN, Cai W. Deletion of murine astrocytic vesicular nucleotide transporter increases anxiety and depressive-like behavior and attenuates motivation for reward. Mol Psychiatry 2025; 30:506-520. [PMID: 39122778 PMCID: PMC11750621 DOI: 10.1038/s41380-024-02692-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
Astrocytes are multi-functional glial cells in the central nervous system that play critical roles in modulation of metabolism, extracellular ion and neurotransmitter levels, and synaptic plasticity. Astrocyte-derived signaling molecules mediate many of these modulatory functions of astrocytes, including vesicular release of ATP. In the present study, we used a unique genetic mouse model to investigate the functional significance of astrocytic exocytosis of ATP. Using primary cultured astrocytes, we show that loss of vesicular nucleotide transporter (Vnut), a primary transporter responsible for loading cytosolic ATP into the secretory vesicles, dramatically reduces ATP loading into secretory lysosomes and ATP release, without any change in the molecular machinery of exocytosis or total intracellular ATP content. Deletion of astrocytic Vnut in adult mice leads to increased anxiety, depressive-like behaviors, and decreased motivation for reward, especially in females, without significant impact on food intake, systemic glucose metabolism, cognition, or sociability. These behavioral alterations are associated with significant decreases in the basal extracellular dopamine levels in the nucleus accumbens. Likewise, ex vivo brain slices from these mice show a strong trend toward a reduction in evoked dopamine release in the nucleus accumbens. Mechanistically, the reduced dopamine signaling we observed is likely due to an increased expression of monoamine oxidases. Together, these data demonstrate a key modulatory role of astrocytic exocytosis of ATP in anxiety, depressive-like behavior, and motivation for reward, by regulating the mesolimbic dopamine circuitry.
Collapse
Affiliation(s)
- Qian Huang
- Department of Molecular and Cellular Biochemistry, the Barnstable Brown Diabetes and Obesity Center, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Hiu Ham Lee
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Bryan Volpe
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Qingchen Zhang
- Program in Pharmacology and Experimental Therapeutics and Pharmacology and Drug Development, Graduate School of Biomedical Sciences and Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Chang Xue
- Program in Pharmacology and Experimental Therapeutics and Pharmacology and Drug Development, Graduate School of Biomedical Sciences and Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Brian C Liu
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Yahia R Abuhasan
- Program in Pharmacology and Experimental Therapeutics and Pharmacology and Drug Development, Graduate School of Biomedical Sciences and Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Lingyun Li
- Program in Pharmacology and Experimental Therapeutics and Pharmacology and Drug Development, Graduate School of Biomedical Sciences and Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Jeremy S Yang
- Program in Pharmacology and Experimental Therapeutics and Pharmacology and Drug Development, Graduate School of Biomedical Sciences and Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Julie Egholm
- Program in Pharmacology and Experimental Therapeutics and Pharmacology and Drug Development, Graduate School of Biomedical Sciences and Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Cristina Gutierrez-Vazquez
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Allen Li
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Alyssa Lee
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Sharon Tang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Chun Wa Wong
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Tiemin Liu
- Key Laboratory of Genetic Engineering, Department of Endocrinology and Metabolism, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, PR China
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Yuan Huang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Raddy L Ramos
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Randy F Stout
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | | | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bradford B Lowell
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Emmanuel N Pothos
- Program in Pharmacology and Experimental Therapeutics and Pharmacology and Drug Development, Graduate School of Biomedical Sciences and Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Weikang Cai
- Department of Molecular and Cellular Biochemistry, the Barnstable Brown Diabetes and Obesity Center, University of Kentucky College of Medicine, Lexington, KY, USA.
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA.
| |
Collapse
|
41
|
Abstract
Almost every facet of our behavior and physiology varies predictably over the course of day and night, anticipating and adapting us to their associated opportunities and challenges. These rhythms are driven by endogenous biological clocks that, when deprived of environmental cues, can continue to oscillate within a period of approximately 1 day, hence circa-dian. Normally, retinal signals synchronize them to the cycle of light and darkness, but disruption of circadian organization, a common feature of modern lifestyles, carries considerable costs to health. Circadian timekeeping pivots around a cell-autonomous molecular clock, widely expressed across tissues. These cellular timers are in turn synchronized by the principal circadian clock of the brain: the hypothalamic suprachiasmatic nucleus (SCN). Intercellular signals make the SCN network a very powerful pacemaker. Previously, neurons were considered the sole SCN timekeepers, with glial cells playing supportive roles. New discoveries have revealed, however, that astrocytes are active partners in SCN network timekeeping, with their cell-autonomous clock regulating extracellular glutamate and GABA concentrations to control circadian cycles of SCN neuronal activity. Here, we introduce circadian timekeeping at the cellular and SCN network levels before focusing on the contributions of astrocytes and their mutual interaction with neurons in circadian control in the brain.
Collapse
Affiliation(s)
- Nicola J. Smyllie
- Medical Research Council Laboratory of Molecular Biology, Cambridge, U.K
| | | | - Andrew P. Patton
- Medical Research Council Laboratory of Molecular Biology, Cambridge, U.K
| |
Collapse
|
42
|
Veiga A, Abreu DS, Dias JD, Azenha P, Barsanti S, Oliveira JF. Calcium-Dependent Signaling in Astrocytes: Downstream Mechanisms and Implications for Cognition. J Neurochem 2025; 169:e70019. [PMID: 39992167 DOI: 10.1111/jnc.70019] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/25/2025]
Abstract
Astrocytes are glial cells recognized for their diverse roles in regulating brain circuit structure and function. They can sense and adapt to changes in the microenvironment due to their unique structural and biochemical properties. A key aspect of astrocytic function involves calcium (Ca2+)-dependent signaling, which serves as a fundamental mechanism for their interactions with neurons and other cells in the brain. However, while significant progress has been made in understanding the spatio-temporal properties of astrocytic Ca2+ signals, the downstream molecular pathways and exact mechanisms through which astrocytes decode these signals to regulate homeostatic and physiological processes remain poorly understood. To address this topic, we review here the available literature on the sources of intracellular Ca2+, as well as its downstream mechanisms and signaling pathways. We review the well-studied Ca2+-dependent exocytosis but draw attention to additional intracellular Ca2+-dependent mechanisms that are less understood and are, most likely, highly influential for many other cellular functions. Finally, we review how intracellular Ca2+ is thought to underlie neuron-astrocyte signaling in brain regions involved in cognitive processing.
Collapse
Affiliation(s)
- Alexandra Veiga
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Daniela Sofia Abreu
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - José Duarte Dias
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia Azenha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sara Barsanti
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João Filipe Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
43
|
Piacentini R, Grassi C. Interleukin 1β receptor and synaptic dysfunction in recurrent brain infection with Herpes simplex virus type-1. Neural Regen Res 2025; 20:416-423. [PMID: 38819045 PMCID: PMC11317954 DOI: 10.4103/nrr.nrr-d-23-01690] [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: 10/11/2023] [Revised: 02/21/2024] [Accepted: 03/21/2024] [Indexed: 06/01/2024] Open
Abstract
Several experimental evidence suggests a link between brain Herpes simplex virus type-1 infection and the occurrence of Alzheimer's disease. However, the molecular mechanisms underlying this association are not completely understood. Among the molecular mediators of synaptic and cognitive dysfunction occurring after Herpes simplex virus type-1 infection and reactivation in the brain neuroinflammatory cytokines seem to occupy a central role. Here, we specifically reviewed literature reports dealing with the impact of neuroinflammation on synaptic dysfunction observed after recurrent Herpes simplex virus type-1 reactivation in the brain, highlighting the role of interleukins and, in particular, interleukin 1β as a possible target against Herpes simplex virus type-1-induced neuronal dysfunctions.
Collapse
Affiliation(s)
- Roberto Piacentini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| |
Collapse
|
44
|
D'Gama PP, Jeong I, Nygård AM, Jamali A, Yaksi E, Jurisch-Yaksi N. Motile cilia modulate neuronal and astroglial activity in the zebrafish larval brain. Cell Rep 2025; 44:115195. [PMID: 39798091 DOI: 10.1016/j.celrep.2024.115195] [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: 02/09/2024] [Revised: 10/11/2024] [Accepted: 12/20/2024] [Indexed: 01/15/2025] Open
Abstract
The brain uses a specialized system to transport cerebrospinal fluid (CSF), consisting of interconnected ventricles lined by motile ciliated ependymal cells. These cells act jointly with CSF secretion and cardiac pressure gradients to regulate CSF dynamics. To date, the link between cilia-mediated CSF flow and brain function is poorly understood. Using zebrafish larvae as a model system, we identify that loss of ciliary motility does not alter progenitor proliferation, brain morphology, or spontaneous neural activity despite leading to an enlarged telencephalic ventricle. We observe altered neuronal responses to photic stimulations in the optic tectum and hindbrain and brain asymmetry defects in the habenula. Finally, we investigate astroglia since they contact CSF and regulate neuronal activity. Our analyses reveal a reduction in astroglial calcium signals during both spontaneous and light-evoked activity. Our findings highlight a role of motile cilia in regulating brain physiology through the modulation of neural and astroglial networks.
Collapse
Affiliation(s)
- Percival P D'Gama
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Erling Skjalgssons Gate 1, 7491 Trondheim, Norway
| | - Inyoung Jeong
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Erling Skjalgssons Gate 1, 7491 Trondheim, Norway
| | - Andreas Moe Nygård
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Erling Skjalgssons Gate 1, 7491 Trondheim, Norway
| | - Ahmed Jamali
- Kavli Institute for Systems Neuroscience and Centre for Algorithms in the Cortex, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030 Trondheim, Norway
| | - Emre Yaksi
- Kavli Institute for Systems Neuroscience and Centre for Algorithms in the Cortex, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030 Trondheim, Norway; Koç University Research Center for Translational Medicine, Koç University School of Medicine, Davutpaşa Caddesi, No:4, Topkapı, Istanbul 34010, Turkey
| | - Nathalie Jurisch-Yaksi
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Erling Skjalgssons Gate 1, 7491 Trondheim, Norway; Kavli Institute for Systems Neuroscience and Centre for Algorithms in the Cortex, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030 Trondheim, Norway.
| |
Collapse
|
45
|
Tizabi Y, Antonelli MC, Tizabi D, Aschner M. Role of Glial Cells and Receptors in Schizophrenia Pathogenesis. Neurochem Res 2025; 50:85. [PMID: 39869278 DOI: 10.1007/s11064-025-04336-8] [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: 09/30/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/28/2025]
Abstract
The specific pathogeneses of schizophrenia (SCZ) remain an enigma despite extensive research that has implicated both genetic and environmental factors. Recent revelations that dysregulated immune system caused by glial cell overactivation result in neuroinflammation, a key player in neurodegenerative as well as neuropsychiatric disorders including SCZ are providing novel clues on potential therapeutic interventions. Here, we review the roles of glial cells (Dr. Arne Schousboe's passion) and two of their most implicated receptors, toll-like receptors (TLRs), and nicotinic cholinergic receptors, in SCZ pathology with suggestions as potential targets in this devastating neuropsychiatric condition.
Collapse
Affiliation(s)
- Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA.
| | - Marta C Antonelli
- Facultad de Medicina, UBA, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", Buenos Aires, Argentina
| | - Daniela Tizabi
- Institute of Marine and Environmental Technology, University of Maryland Center for Environmental Science, Baltimore, MD, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
46
|
Doliwa M, Kuzniewska B, Nader K, Reniewicz P, Kaczmarek L, Michaluk P, Kalita K. Astrocyte-Secreted Lcn2 Modulates Dendritic Spine Morphology. Cells 2025; 14:159. [PMID: 39936951 PMCID: PMC11817088 DOI: 10.3390/cells14030159] [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: 10/30/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 02/13/2025] Open
Abstract
Learning and memory formation rely on synaptic plasticity, the process that changes synaptic strength in response to neuronal activity. In the tripartite synapse concept, molecular signals that affect synapse strength and morphology originate not only from the pre- and post-synaptic neuronal terminals but also from astrocytic processes ensheathing many synapses. Despite significant progress made in understanding astrocytic contribution to synaptic plasticity, only a few astrocytic plasticity-related proteins have been identified so far. In this study, we present evidence indicating the role of astrocyte-secreted Lipocalin-2 (Lcn2) in neuronal plasticity. We show that Lcn2 expression is induced in hippocampal astrocytes in a kainate-evoked aberrant plasticity model. Next, we demonstrate that chemically induced long-term potentiation (cLTP) similarly increases Lcn2 expression in astrocytes of neuronal-glial co-cultures, and that glutamate causes the immediate release of Lcn2 from these cultures. Additionally, through experiments in primary astrocytic cultures, we reveal that Lcn2 release is triggered by calcium signaling, and we demonstrate that a brief treatment of neuronal-glial co-cultures with Lcn2 alters the morphology of dendritic spines. Based on these findings, we propose Lcn2 as an activity-dependent molecule released by astrocytes that influences dendritic spine morphology.
Collapse
Affiliation(s)
| | | | | | | | | | - Piotr Michaluk
- Laboratory of Neurobiology, Nencki-EMBL Partnership for Neural Plasticity and Brain Disorders-BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Katarzyna Kalita
- Laboratory of Neurobiology, Nencki-EMBL Partnership for Neural Plasticity and Brain Disorders-BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| |
Collapse
|
47
|
Drouin E, Martínez Murillo R, Hautecoeur P. The brain in Spain: The legacy of Santiago Ramón y Cajal. Neuroscientist 2025:10738584241297663. [PMID: 39829152 DOI: 10.1177/10738584241297663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The legacy of Santiago Ramón y Cajal, Spain's first Nobel laureate neuroscientist recognized as the founding father of modern neuroscience, is to be preserved in a new museum in Madrid: the National Museum of Natural Sciences (MNCN), one of the most important scientific research institutes in the country sciences in the scope of natural sciences of the Spanish National Research Council. For a boy who dreamed of being an artist but started his career apprenticed to first a barber and then a cobbler, Santiago Ramón y Cajal made a distinguished mark in science. One of Cajal's most important contributions to our understanding of the brain was his discovery of the direction of the information flow within neurons and in neural circuits, which he called the "dynamic polarization law," without a doubt the founding principle of neurosciences. The exposition planned by the MNCN is a perfect occasion to show the academy and, it is hoped, the general public at large the beautiful organization of the nervous system as first acknowledged by modern science. With the highly motivated organizers of this well-planned initiative, neuroscientists at the Cajal Institute are confident that this sample of the Cajal legacy will also be taken as an esthetic experience for those who approach it for the first time. It might be that science and art often go together.
Collapse
Affiliation(s)
- Emmanuel Drouin
- Neurology Service, Lille Catholic Institute Hospital Group, (Groupe Hospitalier de l'Institut Catholique de Lille), GHICL, Lomme cedex, France
| | - Ricardo Martínez Murillo
- Grupo de Investigación Neurovascular, Departamento de Neurobiología Traslacional Instituto Cajal, CSIC, Madrid, Spain
| | - Patrick Hautecoeur
- Neurology Service, Lille Catholic Institute Hospital Group, (Groupe Hospitalier de l'Institut Catholique de Lille), GHICL, Lomme cedex, France
| |
Collapse
|
48
|
Kuang X, Chen S, Ye Q. The lactate metabolism and protein lactylation in epilepsy. Front Cell Neurosci 2025; 18:1464169. [PMID: 39876842 PMCID: PMC11772370 DOI: 10.3389/fncel.2024.1464169] [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: 07/13/2024] [Accepted: 12/17/2024] [Indexed: 01/31/2025] Open
Abstract
Protein lactylation is a new form of post-translational modification that has recently been proposed. Lactoyl groups, derived mainly from the glycolytic product lactate, have been linked to protein lactylation in brain tissue, which has been shown to correlate with increased neuronal excitability. Ischemic stroke may promote neuronal glycolysis, leading to lactate accumulation in brain tissue. This accumulation of lactate accumulation may heighten neuronal excitability by upregulating protein lactylation levels, potentially triggering post-stroke epilepsy. Although current clinical treatments for seizures have advanced significantly, approximately 30% of patients with epilepsy remain unresponsive to medication, and the prevalence of epilepsy continues to rise. This study explores the mechanisms of epilepsy-associated neuronal death mediated by lactate metabolism and protein lactylation. This study also examines the potential for histone deacetylase inhibitors to alleviate seizures by modifying lactylation levels, thereby offering fresh perspectives for future research into the pathogenesis and clinical treatment of epilepsy.
Collapse
Affiliation(s)
- Xi Kuang
- Hainan Health Vocational College, Haikou, China
| | - Shuang Chen
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Qingmei Ye
- Hainan General Hospital and Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
49
|
Ravera S, Farsetti E, Maura G, Marcoli M, Bozzo M, Cervetto C, Amaroli A. 810-nm Photobiomodulation Evokes Glutamate Release in Normal and Rotenone-Dysfunctional Cortical Nerve Terminals by Modulating Mitochondrial Energy Metabolism. Cells 2025; 14:67. [PMID: 39851493 PMCID: PMC11764165 DOI: 10.3390/cells14020067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
The dysfunction of mitochondria, the primary source of cellular energy and producer of reactive oxygen species (ROS), is associated with brain aging and neurodegenerative diseases. Scientific evidence indicates that light in the visible and near-infrared spectrum can modulate mitochondrial activity, a phenomenon known in medicine as photobiomodulation therapy (PBM-t). The beneficial effects of PBM-t on dementia and neurodegeneration have been reviewed in the literature. However, the molecular mechanisms underlying these findings have yet to be fully elucidated. This study investigates the mechanism behind dose-dependent glutamate release in nerve terminals after irradiation with 810 nm, 1 W for 60 s continuous, 1 cm2, 1 W/cm2, 60 J, 60 J/cm2 (810 nm-1 W) or 810 nm, 0.1 W for 60 s continuous, 1 cm2, 0.1 W/cm2, 6 J, 6 J/cm2 (810 nm-0.1 W), focusing on mitochondrial activities. The results show that PBM modulated the mitochondrial metabolism of cortical nerve terminals and supported a power-dependent increase in oxidative phosphorylation (OxPhos) activity when stimulated with pyruvate plus malate (P/M) or succinate (succ) as respiratory substrates. The PBM-induced increase in OxPhos was sensitive to adding rotenone (Complex I inhibitor) and antimycin A (Complex III inhibitor) when synaptosomes were stimulated with P/M, but only to antimycin A when stimulated with succ. This allowed us to observe that the glutamate efflux, disrupted in the presence of rotenone, was partially restored by PBM due to the increase in the OxPhos pathway led by Complex II. This evidence suggests that PBM, acting on mitochondria, could facilitate physiological communication within the neuron-astrocyte network through vesicular glutamate release, potentially regulating healthy brain function and brain dysfunction.
Collapse
Affiliation(s)
- Silvia Ravera
- Department of Experimental Medicine, University of Genova, 16132 Genova, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Elisa Farsetti
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, 16148 Genova, Italy;
| | - Guido Maura
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, 16132 Genova, Italy; (G.M.); (M.M.); (M.B.)
| | - Manuela Marcoli
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, 16132 Genova, Italy; (G.M.); (M.M.); (M.B.)
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), 56122 Pisa, Italy
| | - Matteo Bozzo
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, 16132 Genova, Italy; (G.M.); (M.M.); (M.B.)
| | - Chiara Cervetto
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, 16148 Genova, Italy;
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), 56122 Pisa, Italy
| | - Andrea Amaroli
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), 56122 Pisa, Italy
- BIO-Photonics Overarching Research Laboratory, Department of Earth, Environmental and Life Sciences (DISTAV), University of Genova, 16132 Genova, Italy
| |
Collapse
|
50
|
Lana D, Ugolini F, Iovino L, Attorre S, Giovannini MG. Astrocytes phenomics as new druggable targets in healthy aging and Alzheimer's disease progression. Front Cell Neurosci 2025; 18:1512985. [PMID: 39835288 PMCID: PMC11743640 DOI: 10.3389/fncel.2024.1512985] [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: 10/17/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
For over a century after their discovery astrocytes were regarded merely as cells located among other brain cells to hold and give support to neurons. Astrocytes activation, "astrocytosis" or A1 functional state, was considered a detrimental mechanism against neuronal survival. Recently, the scientific view on astrocytes has changed. Accumulating evidence indicate that astrocytes are not homogeneous, but rather encompass heterogeneous subpopulations of cells that differ from each other in terms of transcriptomics, molecular signature, function and response in physiological and pathological conditions. In this review, we report and discuss the recent literature on the phenomic differences of astrocytes in health and their modifications in disease conditions, focusing mainly on the hippocampus, a region involved in learning and memory encoding, in the age-related memory impairments, and in Alzheimer's disease (AD) dementia. The morphological and functional heterogeneity of astrocytes in different brain regions may be related to their different housekeeping functions. Astrocytes that express diverse transcriptomics and phenomics are present in strictly correlated brain regions and they are likely responsible for interactions essential for the formation of the specialized neural circuits that drive complex behaviors. In the contiguous and interconnected hippocampal areas CA1 and CA3, astrocytes show different, finely regulated, and region-specific heterogeneity. Heterogeneous astrocytes have specific activities in the healthy brain, and respond differently to physiological or pathological stimuli, such as inflammaging present in normal brain aging or beta-amyloid-dependent neuroinflammation typical of AD. To become reactive, astrocytes undergo transcriptional, functional, and morphological changes that transform them into cells with different properties and functions. Alterations of astrocytes affect the neurovascular unit, the blood-brain barrier and reverberate to other brain cell populations, favoring or dysregulating their activities. It will be of great interest to understand whether the differential phenomics of astrocytes in health and disease can explain the diverse vulnerability of the hippocampal areas to aging or to different damaging insults, in order to find new astrocyte-targeted therapies that might prevent or treat neurodegenerative disorders.
Collapse
Affiliation(s)
- Daniele Lana
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, Florence, Italy
| | - Ludovica Iovino
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| | - Selene Attorre
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, Florence, Italy
| | - Maria Grazia Giovannini
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|