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
|
Janik K, Jin LQ, Kyzy KZ, Kaminski R, Smith GM, Krynska B. Neural tube defects induce abnormal astrocyte development by activation and epigenetic permissiveness of STAT3. Exp Neurol 2025; 389:115231. [PMID: 40169106 DOI: 10.1016/j.expneurol.2025.115231] [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: 11/16/2024] [Revised: 03/14/2025] [Accepted: 03/28/2025] [Indexed: 04/03/2025]
Abstract
The open neural tube defect, myelomeningocele (MMC), is a debilitating congenital defect of the central nervous system, characterized by impaired spinal cord development. The pathological development of MMC spinal cord involves enhanced astrogenesis. However, the mechanisms underlying the abnormal generation of astrocytes in MMC spinal cord are poorly understood. Using a fetal rat model of this defect, we discovered that neural progenitor cells (NPCs) in the developing spinal cord undergo the accelerated acquisition of astrocytic competency and the abnormal induction of GFAP expression, resulting in the enhanced generation of astrocytes. We identified that the activation of signal transducer and activator of transcription 3 (STAT3) in NPCs, accompanied by the early epigenetic modifications at the consensus STAT3 binding site within the Gfap promoter, induces this pathological process. Furthermore, we demonstrated that the amniotic fluid exposure is the stimulator of STAT3 activation in MMC spinal cord NPCs, promoting GFAP expression and differentiation of NPCs into astrocytes. To validate this mechanism, we showed that elimination of Stat3 expression from NPCs using CRISPR/Cas9 inhibited GFAP expression and the emergence of astrocytes. Thus, our study provides a novel mechanism that drives abnormal astrogenesis in the developing MMC spinal cord.
Collapse
Affiliation(s)
- Karolina Janik
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA; Center for Neural Development and Repair, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Li-Qing Jin
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Kuralai Zholdosh Kyzy
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Rafal Kaminski
- Department of Microbiology, Immunology, and Inflammation, Center for NeuroVirology and Gene Editing, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - George M Smith
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA; Center for Neural Development and Repair, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Barbara Krynska
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA; Center for Neural Development and Repair, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA.
| |
Collapse
|
3
|
Zhou C, Hardin EJ, Zimmer TS, Jackvony S, Barnett D, Khobrekar N, Giacomelli E, Studer L, Orr AL, Orr AG. Neuroimmune signaling mediates astrocytic nucleocytoplasmic disruptions and stress granule formation associated with TDP-43 pathology. Neurobiol Dis 2025; 211:106939. [PMID: 40339618 DOI: 10.1016/j.nbd.2025.106939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Accepted: 04/30/2025] [Indexed: 05/10/2025] Open
Abstract
Alterations in transactivating response region DNA-binding protein 43 (TDP-43) are prevalent in amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and other neurological disorders. TDP-43 influences neuronal functions and might also affect glial cells. However, specific intracellular effects of TDP-43 alterations on glial cells and underlying mechanisms are not clear. We report that TDP-43 dysregulation in mouse and human cortical astrocytes causes nucleoporin mislocalization, nuclear envelope remodeling, and changes in nucleocytoplasmic protein transport. These effects are dependent on interleukin-1 (IL-1) receptor activity and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling and are associated with the formation of cytoplasmic stress granules. Stimulation of IL-1 receptors and NF-κB signaling are necessary and sufficient to induce astrocytic stress granules and rapid nucleocytoplasmic changes, which are broadly alleviated by inhibition of the integrated stress response. These findings establish that TDP-43 alterations and neuroimmune factors can induce nucleocytoplasmic changes through NF-κB signaling, revealing mechanistic convergence of proteinopathy and neuroimmune pathways onto glial nucleocytoplasmic disruptions that may occur in diverse neurological conditions.
Collapse
Affiliation(s)
- Constance Zhou
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA; Helen and Robert Appel Alzheimer's Disease Research Institute, New York, NY, USA; Feil Family Brain and Mind Research Institute, New York, NY, USA
| | - Evelyn J Hardin
- Helen and Robert Appel Alzheimer's Disease Research Institute, New York, NY, USA; Feil Family Brain and Mind Research Institute, New York, NY, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Till S Zimmer
- Helen and Robert Appel Alzheimer's Disease Research Institute, New York, NY, USA; Feil Family Brain and Mind Research Institute, New York, NY, USA
| | - Stephanie Jackvony
- Helen and Robert Appel Alzheimer's Disease Research Institute, New York, NY, USA; Feil Family Brain and Mind Research Institute, New York, NY, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Daniel Barnett
- Helen and Robert Appel Alzheimer's Disease Research Institute, New York, NY, USA; Feil Family Brain and Mind Research Institute, New York, NY, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Noopur Khobrekar
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA; Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Elisa Giacomelli
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA; Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA; Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Adam L Orr
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA; Helen and Robert Appel Alzheimer's Disease Research Institute, New York, NY, USA; Feil Family Brain and Mind Research Institute, New York, NY, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Anna G Orr
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA; Helen and Robert Appel Alzheimer's Disease Research Institute, New York, NY, USA; Feil Family Brain and Mind Research Institute, New York, NY, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
4
|
Meng Y, Zhang J, Fan R, Pang W, Zeng W, Guo Q, Han X, Liu Y, Luo G. AAV-mediated GBA1 and GDNF rescue neurological defects in a murine model of neuronopathic Gaucher disease. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102506. [PMID: 40206656 PMCID: PMC11979523 DOI: 10.1016/j.omtn.2025.102506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Neuropathic Gaucher disease (nGD) is a life-threatening disease that progresses rapidly and is caused by a glucosylceramidase beta 1 (GBA1) mutation, which encodes the lysosomal hydrolase β-glucocerebrosidase (GCase). Nerve damage in nGD, associated with stunted growth and development, arises from the degeneration and death of nervous system cells, which is often irreversible. Approved therapies effectively reduce the substrate burden outside the central nervous system (CNS) through augmenting mutant enzyme activity with pharmacologic recombinant GCase or by inhibiting glucocerebroside synthesis. However, these therapies do not provide neuroprotection. In this study, we developed a novel double-gene therapy based on adeno-associated virus (AAV), AAV9-GBA1-GDNF, which stably expresses human GBA1 and glial derived neurotrophic factor (GDNF) over the long term. Pathological, molecular, and proteomic tests in the nGD model confirmed that the early stages of the disease are characterized by GBA1 deficiency, loss of neuronal function, and even neuronal death. After treatment with AAV9-GBA1-GDNF, the lifespan of nGD mice was extended, and weight, brain development, and motor ability were recovered. Additionally, GBA1 and GDNF additively prevented irreversible neuronal death by activating the AKT/GSK3β pathway. These findings offer potential therapeutic strategies for nGD and other neurodegenerative diseases associated with lysosomal dysfunction.
Collapse
Affiliation(s)
- Yuan Meng
- Institute of Health Sciences, China Medical University, Shenyang 110122, China
| | - Jiale Zhang
- Institute of Health Sciences, China Medical University, Shenyang 110122, China
| | - Ruoyue Fan
- Bionce Biotechnology, Ltd., Nanjing 210061, China
| | - Wei Pang
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang 110122, China
| | - Wanyang Zeng
- Institute of Health Sciences, China Medical University, Shenyang 110122, China
| | - Qingguo Guo
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang 110122, China
| | - Xuefei Han
- Bionce Biotechnology, Ltd., Nanjing 210061, China
| | - Ying Liu
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang 110122, China
| | - Guangzuo Luo
- Institute of Health Sciences, China Medical University, Shenyang 110122, China
- Bionce Biotechnology, Ltd., Nanjing 210061, China
| |
Collapse
|
5
|
Leisawitz JP, He J, Baggeroer C, Hewett SJ. Arsenite-Mediated Transcriptional Regulation of Glutathione Synthesis in Mammalian Primary Cortical Astrocytes. Int J Mol Sci 2025; 26:5375. [PMID: 40508184 PMCID: PMC12155450 DOI: 10.3390/ijms26115375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 05/21/2025] [Accepted: 05/30/2025] [Indexed: 06/16/2025] Open
Abstract
Arsenic, a potent metalloid contaminant of drinking water, is known for its ability to act as an initiator and modulator of disease in a variety of human tissues. Upon ingestion, arsenic is bio-transformed in the liver into a variety of metabolites, including arsenite. Arsenite permeates the blood-brain barrier (BBB), inducing oxidative stress that can be detrimental to brain neurons. As the primary glial cell at the BBB interface, astrocytes play a pivotal role in detoxifying xenobiotics such as arsenite via the production of the tripeptide antioxidant γ-glutamylcysteine, or glutathione (GSH). In this study, we assessed the mRNA levels of key components of the GSH synthetic pathway in astrocytes exposed to arsenite compared to vehicle controls. These components included xCT [substrate-specific light chain of the substrate importing transporter, system xc- (Sxc-)], glutamate-cysteine ligase [both catalytic (GCLC) and modifying (GCLM) subunits], and glutathione synthetase (GS). Additionally, we analyzed protein levels of some components by Western blotting and evaluated functional activity of Sxc- using a fluorescence-based cystine uptake assay. Finally, we utilized a luminescence-based glutathione assay to determine the intracellular and extracellular GSH content in arsenite-treated cells. Arsenite significantly increased xCT, GCLC, GCLM, and GS mRNA levels, an effect blocked by the transcriptional inhibitor actinomycin D (ActD). A corresponding increase in Sxc- activity was also observed in the arsenite treatment groups, along with significant increases in GCLC and GCLM protein expression. However, no increase in GS protein expression was detected. Finally, arsenite treatment significantly increased extracellular GSH levels, an effect which was also prevented by the inclusion of ActD. Overall, our study provides evidence that arsenite transcriptionally regulates several cellular processes necessary for GSH synthesis in primary cortical astrocyte cultures, thereby contributing to a better understanding of how this environmental toxicant influences antioxidant defenses in the brain. However, these results should be interpreted with caution regarding their applicability to vivo systems.
Collapse
Affiliation(s)
| | | | | | - Sandra J. Hewett
- Interdisciplinary Neuroscience Program, Department of Biology, Syracuse University, Syracuse, NY 13210, USA; (J.P.L.); (J.H.); (C.B.)
| |
Collapse
|
6
|
Xu X, Xuan S, Chen S, Liu D, Xiao Q, Tu J. Increased excitatory amino acid transporter 2 levels in basolateral amygdala astrocytes mediate chronic stress-induced anxiety-like behavior. Neural Regen Res 2025; 20:1721-1734. [PMID: 39104111 PMCID: PMC11688569 DOI: 10.4103/nrr.nrr-d-23-01411] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/03/2024] [Accepted: 02/20/2024] [Indexed: 08/07/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202506000-00024/figure1/v/2024-08-05T133530Z/r/image-tiff The conventional perception of astrocytes as mere supportive cells within the brain has recently been called into question by empirical evidence, which has revealed their active involvement in regulating brain function and encoding behaviors associated with emotions. Specifically, astrocytes in the basolateral amygdala have been found to play a role in the modulation of anxiety-like behaviors triggered by chronic stress. Nevertheless, the precise molecular mechanisms by which basolateral amygdala astrocytes regulate chronic stress-induced anxiety-like behaviors remain to be fully elucidated. In this study, we found that in a mouse model of anxiety triggered by unpredictable chronic mild stress, the expression of excitatory amino acid transporter 2 was upregulated in the basolateral amygdala. Interestingly, our findings indicate that the targeted knockdown of excitatory amino acid transporter 2 specifically within the basolateral amygdala astrocytes was able to rescue the anxiety-like behavior in mice subjected to stress. Furthermore, we found that the overexpression of excitatory amino acid transporter 2 in the basolateral amygdala, whether achieved through intracranial administration of excitatory amino acid transporter 2 agonists or through injection of excitatory amino acid transporter 2-overexpressing viruses with GfaABC1D promoters, evoked anxiety-like behavior in mice. Our single-nucleus RNA sequencing analysis further confirmed that chronic stress induced an upregulation of excitatory amino acid transporter 2 specifically in astrocytes in the basolateral amygdala. Moreover, through in vivo calcium signal recordings, we found that the frequency of calcium activity in the basolateral amygdala of mice subjected to chronic stress was higher compared with normal mice. After knocking down the expression of excitatory amino acid transporter 2 in the basolateral amygdala, the frequency of calcium activity was not significantly increased, and anxiety-like behavior was obviously mitigated. Additionally, administration of an excitatory amino acid transporter 2 inhibitor in the basolateral amygdala yielded a notable reduction in anxiety level among mice subjected to stress. These results suggest that basolateral amygdala astrocytic excitatory amino acid transporter 2 plays a role in in the regulation of unpredictable chronic mild stress-induced anxiety-like behavior by impacting the activity of local glutamatergic neurons, and targeting excitatory amino acid transporter 2 in the basolateral amygdala holds therapeutic promise for addressing anxiety disorders.
Collapse
Affiliation(s)
- Xirong Xu
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- University of Chinese of Academy of Sciences, Beijing, China
| | - Shoumin Xuan
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Shuai Chen
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- University of Chinese of Academy of Sciences, Beijing, China
| | - Dan Liu
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Qian Xiao
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Jie Tu
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- University of Chinese of Academy of Sciences, Beijing, China
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| |
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
|
Agnello L, Gambino CM, Ciaccio AM, Salemi G, Brighina F, Ragonese P, Piccoli T, Blandino V, Di Stefano V, Cacciabaudo F, Masucci A, Vassallo R, Scazzone C, Del Ben F, Ciaccio M. The value of serum glial fibrillary acidic protein as a biomarker of astrogliosis in different neurological diseases. Clin Chim Acta 2025; 572:120248. [PMID: 40113024 DOI: 10.1016/j.cca.2025.120248] [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: 02/27/2025] [Revised: 03/16/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Glial Fibrillary Acidic Protein (GFAP) is a well-established biomarker of astrocytes and astrogliosis, a pathological response observed in various neurological diseases. This study aimed to evaluate the diagnostic performance of serum GFAP in Alzheimer's disease (AD), multiple sclerosis (MS), and transthyretin amyloidosis (ATTR) polyneuropathy. METHODS We performed a retrospective observational study, including 498 participants (337 healthy controls and 161 patients with AD, MS, or ATTR amyloidosis). Serum GFAP levels were measured using the Lumipulse G1200 platform, and statistical analyses were performed to compare levels across disease groups and assess their diagnostic accuracy. RESULTS GFAP levels were significantly elevated in all neurological disease groups compared to age-matched controls, with the highest levels found in AD (79.4 pg/mL vs. 39.5 pg/mL, p = 2.55 × 10-12). ROC curve analysis revealed that GFAP had strong diagnostic performance for AD (AUC = 0.86), moderate performance for ATTR amyloidosis (AUC = 0.67), and poor performance for MS (AUC = 0.61). CONCLUSIONS These findings suggest that GFAP is a promising biomarker for AD, reflecting astrocytic activation and neuroinflammatory processes. Its diagnostic utility in ATTR amyloidosis is moderate, while its role in MS remains limited.
Collapse
Affiliation(s)
- Luisa Agnello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo, Palermo, Italy
| | - Caterina Maria Gambino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo, Palermo, Italy; Department of Laboratory Medicine, University Hospital Paolo Giaccone, Palermo, Italy
| | - Anna Maria Ciaccio
- Department of Health Promotion, Maternal and Infant Care, Internal Medicine and Medical Specialties "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Giuseppe Salemi
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Unit of Neurology, University of Palermo, Palermo, Italy
| | - Filippo Brighina
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Unit of Neurology, University of Palermo, Palermo, Italy
| | - Paolo Ragonese
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Unit of Neurology, University of Palermo, Palermo, Italy
| | - Tommaso Piccoli
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Unit of Neurology, University of Palermo, Palermo, Italy
| | - Valeria Blandino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Unit of Neurology, University of Palermo, Palermo, Italy
| | - Vincenzo Di Stefano
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Unit of Neurology, University of Palermo, Palermo, Italy
| | - Francesco Cacciabaudo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo, Palermo, Italy
| | - Anna Masucci
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo, Palermo, Italy
| | - Roberta Vassallo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo, Palermo, Italy
| | - Concetta Scazzone
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo, Palermo, Italy
| | - Fabio Del Ben
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico (CRO)-IRCCS, Aviano, Italy
| | - Marcello Ciaccio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo, Palermo, Italy; Department of Laboratory Medicine, University Hospital Paolo Giaccone, Palermo, Italy.
| |
Collapse
|
9
|
Zhang Y, Liu Y, Wan H, Deng H, Xu P, Cao L, Yang X, Li H. Silencing HMGB1 secretion inhibited EV71-induced blood-brain barrier dysfunction and neural inflammation by depressing astrocyte activation via sHh signal blockage. Int J Biochem Cell Biol 2025; 185:106797. [PMID: 40349912 DOI: 10.1016/j.biocel.2025.106797] [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: 12/23/2024] [Revised: 05/06/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
It is unclear whether high mobility group protein B1 (HMGB1) is associated with the malignant characterization of hand, foot, and mouth disease (HFMD), and whether it plays a key regulatory role in the process of enterovirus 71 (EV71)-induced brain damage. Firstly, we analyzed the correlation between clinical information and HMGB1 concentrations in patients with mild and severe HFMD. Immunofluorescence was used to determine the expression level of HMGB1 in astrocytes. The levels of cellular inflammatory factors (IL-1β, IL-4, IL-6, TNF-α and TGF-β1), chemokines (CCL2, CXCL10 and CXCL12) and adhesion factors (integrin β, P-gp, VCAM-1 and ICAM-1) were detected by ELISA kits. Western blot was used to measure the levels of blood-brain barrier (BBB) stability related factors (retinoic acid (RA), ANG1, ApoE and IGF-1) in astrocytes and BBB structure related proteins (occluding, claudin, PTCH-1 and ZO-1) in endothelial cells. Clinical studies found that the expression of HMGB1 was closely related to the HFMD severity. Knockdown of HMGB1 alleviated EV71-induced neuron damage and inhibited cellular inflammation and apoptosis. Importantly, silencing HMGB1 depressed excessive proliferation and the inflammation response of astrocytes caused by EV71 infection. Furthermore, knockdown of HMGB1 enhanced BBB stability by improving astrocyte adhesion and endothelial tight junctions. Mechanistically, HMGB1 regulated the stability of BBB by regulating sHh signaling and secretion in astrocytes. In conclusion, the level of HMGB1 is closely related to the clinical symptoms of patients with HFMD, and inhibiting the expression of HMGB1 promotes BBB stability by promoting sHh signaling in astrocytes.
Collapse
Affiliation(s)
- Yufeng Zhang
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, China
| | - Yanfang Liu
- Department of Ophthalmology, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, China
| | - Han Wan
- Department of General Surgery, Xi'an No.3 Hospital, Xi'an, Shaanxi 710021, China
| | - Huiling Deng
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, China
| | - Pengfei Xu
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, China
| | - Lu Cao
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, China
| | - Xiru Yang
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, China
| | - Hui Li
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Department of Neonatology, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, China.
| |
Collapse
|
10
|
Chen J, Xu S, Wang L, Liu X, Liu G, Tan Q, Li W, Zhang S, Du Y. Refining the interactions between microglia and astrocytes in Alzheimer's disease pathology. Neuroscience 2025; 573:183-197. [PMID: 40120713 DOI: 10.1016/j.neuroscience.2025.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/03/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Microglia and astrocytes are central to the pathogenesis and progression of Alzheimer's Disease (AD), working both independently and collaboratively to regulate key pathological processes such as β-amyloid protein (Aβ) deposition, tau aggregation, neuroinflammation, and synapse loss. These glial cells interact through complex molecular pathways, including IL-3/IL-3Ra and C3/C3aR, which influence disease progression and cognitive decline. Emerging research suggests that modulating these pathways could offer therapeutic benefits. For instance, recombinant IL-3 administration in mice reduced Aβ plaques and improved cognitive functions, while C3aR inhibition alleviated Aβ and tau pathologies, restored synaptic function, and corrected immune dysregulation. However, the effects of these interactions are context-dependent. Acute C3/C3aR activation enhances microglial Aβ clearance, whereas chronic activation impairs it, highlighting the dual roles of glial signaling in AD. Furthermore, C3/C3aR signaling not only impacts Aβ clearance but also modulates tau pathology and synaptic integrity. Given AD's multifactorial nature, understanding the specific pathological environment is crucial when investigating glial cell contributions. The interplay between microglia and astrocytes can be both neuroprotective and neurotoxic, depending on the disease stage and brain region. This complexity underscores the need for targeted therapies that modulate glial cell activity in a context-specific manner. By elucidating the molecular mechanisms underlying microglia-astrocyte interactions, this research advances our understanding of AD and paves the way for novel therapeutic strategies aimed at mitigating neurodegeneration and cognitive decline in AD and related disorders.
Collapse
Affiliation(s)
- Jiangmin Chen
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Shuyu Xu
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Li Wang
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Xinyuan Liu
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Guangya Liu
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Qian Tan
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Weixian Li
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Shuai Zhang
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Yanjun Du
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China; Hubei Shizhen Laboratory, China; Hubei International Science and Technology Cooperation Base of Preventive Treatment by Acupuncture and Moxibustion, China; Hubei Provincial Hospital of Traditional Chinese Medicine, China.
| |
Collapse
|
11
|
Wang S, Yang Y, Lin J, Zhang W, Yang C, Zhang R, Zhou C, Zhang L, Wang X, Liu J, Jin X, Ma Y. Astragalin actives autophagy and inhibits apoptosis of astrocytes in AD mice via down-regulating Fas/Fasl-VDAC1 pathway. Free Radic Biol Med 2025; 232:72-85. [PMID: 40032030 DOI: 10.1016/j.freeradbiomed.2025.02.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025]
Abstract
Alzheimer's disease (AD) as a common neurodegenerative disease, which characterized by amyloid Aβ deposition and neurofibrillary tangles. Astragalin (AST), a natural flavonoid, has anti-inflammatory, antioxidant, anti-cancer, and other pharmacological effects. Astrocytes can phagocytize and degrade Aβ in their vicinity. In this study, we used the AD mice model established by injecting the mixture of Aβ1-42 and Aβ25-35 into the CA1 region of the hippocampus, and C8D1A cells injured by Aβ1-42 to explore the neuroprotective effects of AST. Our findings showed that AST enhances learning and cognitive ability of AD mice, reduces Aβ deposition and neurofibrillary tangles in the brain, and improves the structural morphology of hippocampal nerve cells. Furthermore, AST promoted autophagy and suppressed apoptosis of astrocytes in the AD model. Additionally, AST inhibited the expression of proteins associated with the Fas/Fasl-VDAC1 signaling pathway, while autophagy inhibitor chloroquine (CQ) or apoptosis agonist phenoxodiol reversed above change. Interestingly, consistent with the action of pathway Fas inhibitor KR-33493, AST could activate autophagy of Aβ1-42 injured C8D1A cells while inhibit their apoptosis. In conclusion, AST activated autophagy and inhibited apoptosis of hippocampal astrocytes in AD mice, ameliorating animal cognitive deficits by down-regulating Fas/Fasl-VDAC1 signaling pathway. Thus, this study provided a new perspective and experimental foundation for developing AD treatment drugs.
Collapse
Affiliation(s)
- Shuhan Wang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yaqi Yang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiahong Lin
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Weishan Zhang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Cuizhu Yang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Runheng Zhang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chang Zhou
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Li Zhang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xin Wang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jing Liu
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaobao Jin
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuxin Ma
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
12
|
Zhai Y, Lu K, Yuan Y, Zhang Z, Xue L, Zhao F, Xu X, Wang H. Semaglutide improves cognitive function and neuroinflammation in APP/PS1 transgenic mice by activating AMPK and inhibiting TLR4/NF-κB pathway. J Alzheimers Dis 2025; 105:416-432. [PMID: 40151913 DOI: 10.1177/13872877251329439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
BackgroundAlzheimer's disease (AD) causes cognitive function disorder and has become the preeminent cause of dementia. Glucagon-like peptide-1 (GLP-1) receptor agonists, semaglutide, have shown positive effects on promoting the cognitive function. However, research about the mechanism of semaglutide as a therapeutic intervention in AD is sparse.ObjectiveThis study was to investigate the therapeutic efficacy of semaglutide in a transgenic mouse model of AD pathology and explored the detailed mechanism by semaglutide modulated neuroinflammatory processes.MethodsMale amyloid precursor protein/presenilin 1 (APP/PS1) transgenic mice were treated with semaglutide or vehicle for 8 weeks. Morris water maze test was used to assess the therapeutic efficacy of semaglutide on recognition function. Pathology analysis was performed to detect the deposition of amyloid plaques. High-throughput sequencing analysis was applied to specify the mechanism. Microglia and astrocyte activation were assessed with immunofluorescent staining. Inflammation cytokine levels were evaluated with enzyme-linked immunosorbent assay (ELISA). Related proteins and pathway were evaluated with western blot.ResultsSemaglutide treatment attenuated Aβ accumulation and enhanced cognitive function in APP/PS1 transgenic mice. Through transcriptomic profiling, immunohistochemical staining, and ELISA, semaglutide was substantiated to inhibit the overactivation of microglia and astrocytes, as well as to curtail the secretion of inflammatory mediators. Furthermore, semaglutide robustly activated AMP-activated protein kinase (AMPK) and suppressed the toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) signaling cascade, thus reducing the Aβ deposition and dampening the inflammatory cascade.ConclusionsThe results demonstrated that semaglutide mitigated neuroinflammation and decelerated the advance of AD in APP/PS1 transgenic mice.
Collapse
Affiliation(s)
- Yanyu Zhai
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
| | - Kaili Lu
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
| | - Yuan Yuan
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ziyao Zhang
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
| | - Lixia Xue
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
| | - Fei Zhao
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
| | - Xiaofeng Xu
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
| | - Hongmei Wang
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
| |
Collapse
|
13
|
Zhao Y, Xiong C, Wang B, Li D, Liu J, Wei S, Hou Y, Zhou Y, Zheng R. The Discovery of Phages in the Substantia Nigra and Its Implication for Parkinson's Disease. RESEARCH (WASHINGTON, D.C.) 2025; 8:0657. [PMID: 40308709 PMCID: PMC12041648 DOI: 10.34133/research.0657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/28/2025] [Accepted: 03/10/2025] [Indexed: 05/02/2025]
Abstract
Background: A century ago, a mystery between a virus and Parkinson's disease (PD) was described. Owing to the limitation of human brain biopsy and the challenge of electron microscopy in observing virions in human brain tissue, it has been difficult to study the viral etiology of PD. Recent discovery of virobiota reveals that viruses coexist with humans as symbionts. Newly developed transcriptomic sequencing and novel bioinformatic approaches for mining the encrypted virome in human transcriptome make it possible to study the relationship between symbiotic viruses and PD. Nevertheless, whether viruses exist in the human substantia nigra (SN) and whether symbiotic viruses underlie PD pathogenesis remain unknown. Methods: We collected current worldwide human SN transcriptomic datasets from the United States, the United Kingdom, the Netherlands, and Switzerland. We used bioinformatic approaches including viruSITE and the Viral-Track to identify the existence of viruses in the SN of patients. The comprehensive RNA sequencing-based virome analysis pipeline was used to characterize the virobiota in the SN. The Pearson's correlation analysis was used to examine the association between the viral RNA fragment counts (VRFCs) and PD-related human gene sequencing reads in the SN. The differentially expressed genes (DEGs) in the SN between PD patients and non-PD individuals were used to examine the molecular signatures of PD and also evaluate the impact of symbiotic viruses on the SN. Findings: We observed the existence of viruses in the human SN. A dysbiosis of virobiota was found in the SN of PD patients. A marked correlation between VRFC and PD-related human gene expression was detected in the SN of PD patients. These PD-related human genes correlated to VRFC were named as the virus-correlated PD-related genes (VPGs). We identified 3 bacteriophages (phages), including the Proteus phage VB_PmiS-Isfahan, the Escherichia phage phiX174, and the Lactobacillus phage Sha1, that might impair the gene expression of neural cells in the SN of PD patients. The Proteus phage VB_PmiS-Isfahan was a common virus in the SN of patients from the United Kingdom, the Netherlands, and Switzerland. VPGs and DEGs together highlighted that the phages might dampen dopamine biosynthesis and weaken the cGAS-STING function. Interpretation: This is the first study to discover the involvement of phages in PD pathogenesis. A lifelong low symbiotic viral load in the SN may be a contributor to PD pathogenesis. Our findings unlocked the black box between brain virobiota and PD, providing a novel insight into PD etiology from the perspective of phage-human symbiosis.
Collapse
Affiliation(s)
- Yun Zhao
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Changxian Xiong
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Bingwei Wang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Daotong Li
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Jiarui Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Shizhang Wei
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Yujia Hou
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Yuan Zhou
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences,
Peking University, Beijing, China
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences,
Peking University, Beijing, China
- Neuroscience Research Institute,
Peking University, Beijing, China
- Key Laboratory for Neuroscience of Ministry of Education,
Peking University, Beijing, China
- Key Laboratory for Neuroscience of National Health Commission,
Peking University, Beijing, China
- Beijing Life Science Academy, Beijing, China
| |
Collapse
|
14
|
Wang S, Baumert R, Séjourné G, Sivadasan Bindu D, Dimond K, Sakers K, Vazquez L, Moore JL, Tan CX, Takano T, Rodriguez MP, Brose N, Bradley L, Lessing R, Soderling SH, La Spada AR, Eroglu C. PD-linked LRRK2 G2019S mutation impairs astrocyte morphology and synapse maintenance via ERM hyperphosphorylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.04.09.536178. [PMID: 39253496 PMCID: PMC11383028 DOI: 10.1101/2023.04.09.536178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Astrocytes are highly complex cells that mediate critical roles in synapse formation and maintenance by establishing thousands of direct contacts with synapses through their perisynaptic processes. Here, we found that the most common Parkinsonism gene mutation, LRRK2 G2019S, enhances the phosphorylation of the ERM proteins (Ezrin, Radixin, and Moesin), components of the perisynaptic astrocyte processes in a subset of cortical astrocytes. The ERM hyperphosphorylation was accompanied by decreased astrocyte morphological complexity and reduced excitatory synapse density and function. Dampening ERM phosphorylation levels in LRRK2 G2019S mouse astrocytes restored both their morphology and the excitatory synapse density in the anterior cingulate cortex. To determine how LRRK2 mutation impacts Ezrin interactome, we used an in vivo BioID proteomic approach, and we found that astrocytic Ezrin interacts with Atg7, a master regulator of autophagy. The Ezrin/Atg7 interaction is inhibited by Ezrin phosphorylation, thus diminished in LRRK2 G2019S astrocytes. Importantly, the Atg7 function is required to maintain proper astrocyte morphology. Our data provide a molecular pathway through which the LRRK2 G2019S mutation alters astrocyte morphology and synaptic density in a brain-region-specific manner.
Collapse
|
15
|
Kopalli SR, Behl T, Baldaniya L, Ballal S, Joshi KK, Arya R, Chaturvedi B, Chauhan AS, Verma R, Patel M, Jain SK, Wal A, Gulati M, Koppula S. Neuroadaptation in neurodegenerative diseases: compensatory mechanisms and therapeutic approaches. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111375. [PMID: 40280271 DOI: 10.1016/j.pnpbp.2025.111375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Progressive neuronal loss is a hallmark of neurodegenerative diseases including Alzheimer's, Parkinson's, Huntington's, and Amyotrophic Lateral Sclerosis (ALS), which cause cognitive and motor impairment. Delaying the onset and course of symptoms is largely dependent on neuroadaptation, the brain's ability to restructure in response to damage. The molecular, cellular, and systemic processes that underlie neuroadaptation are examined in this study. These mechanisms include gliosis, neurogenesis, synaptic plasticity, and changes in neurotrophic factors. Axonal sprouting, dendritic remodelling, and compensatory alterations in neurotransmitter systems are important adaptations observed in NDDs; nevertheless, these processes may shift to maladaptive plasticity, which would aid in the advancement of the illness. Amyloid and tau pathology-induced synaptic alterations in Alzheimer's disease emphasize compensatory network reconfiguration. Dopamine depletion causes a major remodelling of the basal ganglia in Parkinson's disease, and non-dopaminergic systems compensate. Both ALS and Huntington's disease rely on motor circuit rearrangement and transcriptional dysregulation to slow down functional deterioration. Neuroadaptation is, however, constrained by oxidative stress, compromised autophagy, and neuroinflammation, particularly in elderly populations. The goal of emerging therapy strategies is to improve neuroadaptation by pharmacologically modifying neurotrophic factors, neuroinflammation, and synaptic plasticity. Neurostimulation, cognitive training, and physical rehabilitation are instances of non-pharmacological therapies that support neuroplasticity. Restoring compensating systems may be possible with the use of stem cell techniques and new gene treatments. The goal of future research is to combine biomarkers and individualized medicines to maximize neuroadaptive responses and decrease the course of illness. In order to reduce neurodegeneration and enhance patient outcomes, this review highlights the dual function of neuroadaptation in NDDs and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Punjab-140306, India
| | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot 360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, India; Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Bhumi Chaturvedi
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of research and innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Rakesh Verma
- Department of Pharmacology, Institute of Medical Science, BHU, Varanasi, India
| | - Minesh Patel
- Department of Pharmacology & Pharmacy Practice, Saraswati Institute of Pharmaceutical Sciences, Dhanap, Gandhinagar, Gujarat, India
| | - Sanmati Kumar Jain
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Koni, Bilaspur, India, 495009
| | - Ankita Wal
- Pranveer Singh Institute of Technology, Pharmacy, NH-19, Bhauti Road, Kanpur, UP, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
16
|
Vellucci L, Mazza B, Barone A, Nasti A, De Simone G, Iasevoli F, de Bartolomeis A. The Role of Astrocytes in the Molecular Pathophysiology of Schizophrenia: Between Neurodevelopment and Neurodegeneration. Biomolecules 2025; 15:615. [PMID: 40427508 PMCID: PMC12109222 DOI: 10.3390/biom15050615] [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: 02/28/2025] [Revised: 04/05/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Schizophrenia is a chronic and severe psychiatric disorder affecting approximately 1% of the global population, characterized by disrupted synaptic plasticity and brain connectivity. While substantial evidence supports its classification as a neurodevelopmental disorder, non-canonical neurodegenerative features have also been reported, with increasing attention given to astrocytic dysfunction. Overall, in this study, we explore the role of astrocytes as a structural and functional link between neurodevelopment and neurodegeneration in schizophrenia. Specifically, we examine how astrocytes contribute to forming an aberrant substrate during early neurodevelopment, potentially predisposing individuals to later neurodegeneration. Astrocytes regulate neurotransmitter homeostasis and synaptic plasticity, influencing early vulnerability and disease progression through their involvement in Ca2⁺ signaling and dopamine-glutamate interaction-key pathways implicated in schizophrenia pathophysiology. Astrocytes differentiate via nuclear factor I-A, Sox9, and Notch pathways, occurring within a neuronal environment that may already be compromised in the early stages due to the genetic factors associated with the 'two-hits' model of schizophrenia. As a result, astrocytes may contribute to the development of an altered neural matrix, disrupting neuronal signaling, exacerbating the dopamine-glutamate imbalance, and causing excessive synaptic pruning and demyelination. These processes may underlie both the core symptoms of schizophrenia and the increased susceptibility to cognitive decline-clinically resembling neurodegeneration but driven by a distinct, poorly understood molecular substrate. Finally, astrocytes are emerging as potential pharmacological targets for antipsychotics such as clozapine, which may modulate their function by regulating glutamate clearance, redox balance, and synaptic remodeling.
Collapse
Affiliation(s)
- Licia Vellucci
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy
| | - Benedetta Mazza
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Anita Nasti
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Giuseppe De Simone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona (UB), c. Casanova, 143, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), c. Villarroel, 170, 08036 Barcelona, Spain
- Bipolar and Depressive Disorders Unit, Hospìtal Clinic de Barcelona. c. Villarroel, 170, 08036 Barcelona, Spain
| | - Felice Iasevoli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
17
|
Huang L, Zeng F, Wei H, Su T, Su Y, Lin Y, Niu Q, Xu Q. SOAT1 dysregulation in astrocytes drives Blood-Brain barrier dysfunction and neuroinflammation in Alzheimer's disease. Brain Behav Immun 2025; 128:497-509. [PMID: 40274003 DOI: 10.1016/j.bbi.2025.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/02/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that leads to memory loss and cognitive decline, in which blood-brain barrier (BBB) and astrocyte dysfunction are significantly involved. Recent evidence suggests that dysregulation of lipid metabolism in astrocytes contributes to BBB disruption and neuroinflammation in AD. Sterol O-acyltransferase 1 (SOAT1), an enzyme involved in cholesterol esterification, has been implicated in BBB disruption and neuroinflammation, but its specific role in AD remains unclear. This study aimed to investigate the impact of SOAT1 on lipid metabolism, BBB integrity, and neuroinflammation in AD. Using Oil Red O staining of human autopsy brain tissue and reanalysis of publicly available single-nucleus RNA sequencing (snRNA-seq) data, we identified a significant increase in lipid droplet accumulation and lipid metabolism gene expression, particularly in astrocytes, in the brains of AD patients. Furthermore, in vitro BBB models and the 5 × FAD mouse model were used to explore how SOAT1 expression influences BBB function. Our results demonstrated that elevated SOAT1 expression in astrocytes was positively correlated with increased lipid droplet accumulation and compromised BBB integrity. Knockdown of SOAT1 using siRNA or treatment with the SOAT1 inhibitor K604 restored BBB function, reduced neuroinflammation, and improved cognitive function in 5 × FAD mice. These findings suggest that SOAT1 plays a critical role in astrocytic lipid metabolism and BBB dysfunction in AD. Targeting SOAT1 may be a promising therapeutic approach to alleviate neuroinflammation and restore cognitive function in AD patients.
Collapse
Affiliation(s)
- Lian Huang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, PR China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, PR China
| | - Fu Zeng
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, PR China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, PR China
| | - Hui Wei
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, PR China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, PR China
| | - Tong Su
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, PR China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, PR China
| | - Yuwen Su
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, PR China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, PR China
| | - Yarong Lin
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, PR China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, PR China
| | - Qi Niu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, PR China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, PR China
| | - Qi Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, PR China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, PR China.
| |
Collapse
|
18
|
Zagare A, Sauter T, Barmpa K, Pacheco M, Krüger R, Schwamborn JC, Saraiva C. MIRO1 mutation leads to metabolic maladaptation resulting in Parkinson's disease-associated dopaminergic neuron loss. NPJ Syst Biol Appl 2025; 11:37. [PMID: 40246848 PMCID: PMC12006346 DOI: 10.1038/s41540-025-00509-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 03/16/2025] [Indexed: 04/19/2025] Open
Abstract
MIRO1 is a mitochondrial outer membrane protein important for mitochondrial distribution, dynamics and bioenergetics. Over the last decade, evidence has pointed to a link between MIRO1 and Parkinson's disease (PD) pathogenesis. Moreover, a heterozygous MIRO1 mutation (p.R272Q) was identified in a PD patient, from which an iPSC-derived midbrain organoid model was derived, showing MIRO1 mutant-dependent selective loss of dopaminergic neurons. Herein, we use patient-specific iPSC-derived midbrain organoids carrying the MIRO1 p.R272Q mutation to further explore the cellular and molecular mechanisms involved in dopaminergic neuron degeneration. Using single-cell RNA sequencing (scRNAseq) analysis and metabolic modeling we show that the MIRO1 p.R272Q mutation affects the dopaminergic neuron developmental path leading to metabolic deficits and disrupted neuron-astrocyte metabolic crosstalk, which might represent an important pathogenic mechanism leading to their loss.
Collapse
Affiliation(s)
- Alise Zagare
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 2, place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg
| | - Thomas Sauter
- Systems Biology and Epigenetics Group, Department of Life Sciences and Medicine, University of Luxembourg, 2, place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg
| | - Kyriaki Barmpa
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 2, place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg
| | - Maria Pacheco
- Systems Biology and Epigenetics Group, Department of Life Sciences and Medicine, University of Luxembourg, 2, place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg
| | - Rejko Krüger
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 2, place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), 1 A-B rue Thomas Edison, L-1445, Strassen, Luxembourg
- Parkinson Research Clinic, Centre Hospitalier de Luxembourg, 4, rue Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Jens Christian Schwamborn
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 2, place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg.
| | - Claudia Saraiva
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 2, place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
19
|
Gaweda-Walerych K, Aragona V, Lodato S, Sitek EJ, Narożańska E, Buratti E. Progranulin deficiency in the brain: the interplay between neuronal and non-neuronal cells. Transl Neurodegener 2025; 14:18. [PMID: 40234992 PMCID: PMC12001433 DOI: 10.1186/s40035-025-00475-8] [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: 09/20/2024] [Accepted: 02/21/2025] [Indexed: 04/17/2025] Open
Abstract
Heterozygous mutations in GRN gene lead to insufficient levels of the progranulin (PGRN) protein, resulting in frontotemporal dementia (FTD) with TAR DNA-binding protein 43 (TDP-43) inclusions, classified pathologically as frontotemporal lobar degeneration (FTLD-TDP). Homozygous GRN mutations are exceedingly rare and cause neuronal ceroid lipofuscinosis 11, a lysosomal storage disease with onset in young adulthood, or an FTD syndrome with late-onset manifestations. In this review, we highlight the broad spectrum of clinical phenotypes associated with PGRN deficiency, including primary progressive aphasia and behavioral variant of frontotemporal dementia. We explore these phenotypes alongside relevant rodent and in vitro human models, ranging from the induced pluripotent stem cell-derived neural progenitors, neurons, microglia, and astrocytes to genetically engineered heterotypic organoids containing both neurons and astrocytes. We summarize advantages and limitations of these models in recapitulating the main FTLD-GRN hallmarks, highlighting the role of non-cell-autonomous mechanisms in the formation of TDP-43 pathology, neuroinflammation, and neurodegeneration. Data obtained from patients' brain tissues and biofluids, in parallel with single-cell transcriptomics, demonstrate the complexity of interactions among the highly heterogeneous cellular clusters present in the brain, including neurons, astrocytes, microglia, oligodendroglia, endothelial cells, and pericytes. Emerging evidence has revealed that PGRN deficiency is associated with cell cluster-specific, often conserved, genetic and molecular phenotypes in the central nervous system. In this review, we focus on how these distinct cellular populations and their dysfunctional crosstalk contribute to neurodegeneration and neuroinflammation in FTD-GRN. Specifically, we characterize the phenotypes of lipid droplet-accumulating microglia and alterations of myelin lipid content resulting from lysosomal dysfunction caused by PGRN deficiency. Additionally, we consider how the deregulation of glia-neuron communication affects the exchange of organelles such as mitochondria, and the removal of excess toxic products such as protein aggregates, in PGRN-related neurodegeneration.
Collapse
Affiliation(s)
- Katarzyna Gaweda-Walerych
- Department of Neurogenetics and Functional Genomics, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland.
| | - Vanessa Aragona
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Emilia J Sitek
- Division of Neurological and Psychiatric Nursing, Laboratory of Clinical Neuropsychology, Neurolinguistics, and Neuropsychotherapy, Faculty of Health Sciences, Medical University of Gdansk, 80-210, Gdansk, Poland.
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland.
| | - Ewa Narożańska
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland
| | - Emanuele Buratti
- Molecular Pathology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, 34149, Trieste, Italy
| |
Collapse
|
20
|
Wen X, Cao W, Ding H, Chen A, Sun Z, Wang Y, Xi Y, Wu S. Extracellular Vesicles Derived from FGF2-Primed Astrocytes Against Mitochondrial and Synaptic Toxicities in Parkinson's Disease. Int J Nanomedicine 2025; 20:4627-4644. [PMID: 40248029 PMCID: PMC12005207 DOI: 10.2147/ijn.s511474] [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: 12/24/2024] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
Purpose Mitochondrial dysfunction associated with neuronal degeneration and subsequent synaptic disconnection are essential for the development of Parkinson's disease (PD). Considering that astrocytes play key roles in synaptogenesis during development, we hypothesized that fibroblast growth factor - 2 (FGF2), a key factor for astrocyte development, could reverse the toxic phenotype of reactive astrocytes, and the extracellular vesicles (EVs) derived from FGF2-primed astrocytes would enhance synaptogenesis in PD model. The present study was to test this hypothesis. Methods EVs isolated from FGF2-primed astrocytes (FGF2-EVs) were characterized by transmission electron microscopy and nanoparticle tracking analysis. FGF2-EVs were applied to both in vitro and in vivo models of PD. EVs derived from naïve astrocytes (CON-EV) were used as control. Mitochondrial alterations, neuronal survival, synaptogenesis, and mice behavior were subsequently evaluated by quantitative real-time polymerase chain reaction, Western-blotting, immunohistochemistry, and CatWalk gait analysis. To dissect the underlying mechanisms, proteomic analysis and small interfering RNA (siRNA) mediated gene silencing were adopted. Results FGF2 treatment restored the expression of neural progenitor markers and suppressed the levels of A1 astrocytic markers in MPP+ pretreated astrocytes. FGF2-EVs, in comparison with that of CON-EVs, effectively protected neurons from mitochondrial fragmentation and stimulated synaptogenesis, as evidenced by expression of Mitofusin 2 (Mfn2), postsynaptic density protein 95 (PSD-95) and synaptophysin (SYP). Proteomic analysis revealed high enrichment of neural cell adhesion molecule 1 (NCAM1) in FGF2-EVs. Knocking down NCAM1 severely influenced the expression of mitochondrial and synaptic proteins. Furthermore, delivery of FGF2-EVs significantly enhanced the survival of TH+ neurons, the levels of NCAM1 and synaptogenesis in the substantia nigra of PD mice, as well as the locomotion of PD mice. Conclusion EVs from FGF2-primed astrocytes are superior in protecting PD mice against mitochondrial and synaptic toxicities, possibly through NCAM1, which could be used as a therapeutic strategy for PD.
Collapse
Affiliation(s)
- Xiaomin Wen
- College of Life Sciences, Northwest University, Xi’an, Shaanxi, 710069, People’s Republic of China
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Wanjun Cao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Hui Ding
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Andi Chen
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Zhichuan Sun
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Yazhou Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Ye Xi
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| |
Collapse
|
21
|
Ziar R, Tesar PJ, Clayton BLL. Astrocyte and oligodendrocyte pathology in Alzheimer's disease. Neurotherapeutics 2025; 22:e00540. [PMID: 39939240 PMCID: PMC12047399 DOI: 10.1016/j.neurot.2025.e00540] [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: 09/16/2024] [Revised: 01/10/2025] [Accepted: 01/24/2025] [Indexed: 02/14/2025] Open
Abstract
Astrocytes and oligodendrocytes, once considered passive support cells, are now recognized as active participants in the pathogenesis of Alzheimer's disease. Emerging evidence highlights the critical role that these glial cells play in the pathological features of Alzheimer's, including neuroinflammation, excitotoxicity, synaptic dysfunction, and myelin degeneration, which contribute to neurodegeneration and cognitive decline. Here, we review the current understanding of astrocyte and oligodendrocyte pathology in Alzheimer's disease and highlight research that supports the therapeutic potential of modulating astrocyte and oligodendrocyte functions to treat Alzheimer's disease.
Collapse
Affiliation(s)
- Rania Ziar
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Paul J Tesar
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Benjamin L L Clayton
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
22
|
Fu Y, Adler GL, Youssef P, Phan K, Halliday GM, Dzamko N, Kim WS. Human Endogenous Retrovirus K in Astrocytes Is Altered in Parkinson's Disease. Mov Disord 2025; 40:683-692. [PMID: 39840837 PMCID: PMC12006878 DOI: 10.1002/mds.30128] [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: 10/09/2024] [Revised: 01/01/2025] [Accepted: 01/06/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the most common neurodegenerative movement disease. Human endogenous retroviruses (HERVs) are proviral remnants of ancient retroviral infection of germ cells that now constitute about 8% of the human genome. Under certain disease conditions, HERV genes are activated and partake in the disease process. However, virtually nothing is known about the pathological relationship, if any, between HERV and PD. OBJECTIVE The objectives of this study were to unravel the pathological relationship between human endogenous retrovirus K (HERV-K) and PD, determine the localization of HERV-K in the brain, determine whether HERV-K levels are altered in PD brain and blood, and examine whether HERV-K could serve as a biomarker for PD. METHODS In situ HERV-K and glial fibrillary acidic protein (GFAP) expression in the superior frontal and fusiform cortices of PD and control brain were analyzed using immunofluorescence and confocal microscopy. HERV-K load and copy number in PD and control blood were measured by digital droplet polymerase chain reaction and GFAP by single-molecule array. HERV-K load was analyzed in relation to the Hoehn and Yahr Scale and Movement Disorder Society Unified Parkinson's Disease Rating Scale Part III. RESULTS HERV-K is predominantly expressed in astrocytes and colocalized with astrocytic GFAP, with decreased expression of both HERV-K and GFAP in PD brain compared with controls. Consistent with this, HERV-K levels were decreased in PD blood compared with controls and were correlated to blood GFAP levels. HERV-K levels were inversely correlated to PD severity and duration. CONCLUSIONS These findings suggest that HERV-K is related to astrocyte function and to PD progression, and that HERV-K could be neuroprotective. © 2025 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- YuHong Fu
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Gabrielle L. Adler
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Priscilla Youssef
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Katherine Phan
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Glenda M. Halliday
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Nicolas Dzamko
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Woojin Scott Kim
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| |
Collapse
|
23
|
Manavi MA, Nourhashemi M, Emami S, Fathian Nasab MH, Dehnavi F, Küçükkılınç TT, Foroumadi A, Sharifzadeh M, Khoobi M. Lipoic acid scaffold applications in the design of multitarget-directed ligands against Alzheimer's disease. Bioorg Chem 2025; 157:108241. [PMID: 39922042 DOI: 10.1016/j.bioorg.2025.108241] [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: 11/03/2024] [Revised: 01/20/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
Alzheimer's disease (AD) is becoming a fast-growing public health problem which can result in psychological problems as well as loss of speech, language, short-term memory, and motor coordination. Many medications were developed and produced to treat AD, however due to the complexity of the pathology involved in the illness, many of these medications often failed in clinical or preclinical studies. The main issue with the current anti-AD medications is their low efficacy since they use a single target. Multi-target-directed ligands (MTDLs) based on "one molecule; multiple targets" have been introduced to address these two fundamental issues. MTDLs have demonstrated improved efficacy and safety since they regulate many biological targets simultaneously. Alpha-lipoic acid (LA), a natural molecule with distinct properties, is a viable scaffold for developing new MTDLs in treating many neurodegenerative diseases, particularly AD. It is a key mitochondrial enzymes' cofactor and an organic molecule with disulfide functionality. It also has potent antioxidant characteristics that enhance mitochondrial activity. Considering the neuroprotective and anti-inflammatory effects of LA, various hybrids of LA with tacrine, rivastigmine, coumarin and chromone, ibuprofen, melatonin, niacin have been synthesized and biologically evaluated as the MTDLs. In this article, we review the design of LA-based hybrids or conjugates, their biological activities, and structure-activity relationship studies, to develop new MTDLs in the field of AD pharmacotherapy.
Collapse
Affiliation(s)
- Mohammad Amin Manavi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Nourhashemi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran; Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeed Emami
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Hosein Fathian Nasab
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Fateme Dehnavi
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Alireza Foroumadi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran; Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14176-14411, Iran
| | - Mehdi Khoobi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran; Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Zhang X, Zhang Y, Peng X, Yang L, Miao J, Yue Y, Wang Y, Wang X, Zhu C, Song J. Targeting Neuroinflammation in Preterm White Matter Injury: Therapeutic Potential of Mesenchymal Stem Cell-Derived Exosomes. Cell Mol Neurobiol 2025; 45:23. [PMID: 40072734 PMCID: PMC11903990 DOI: 10.1007/s10571-025-01540-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 03/02/2025] [Indexed: 03/14/2025]
Abstract
Neuroinflammation is a key factor in the development of preterm white matter injury (PWMI), leading to glial cell dysfunction, arrest of oligodendrocyte maturation, and long-term neurological damage. As a potential therapeutic strategy, mesenchymal stem cells (MSCs) exhibit significant immunomodulatory and regenerative potential. Recent studies suggest that the primary mechanism of MSC action is their paracrine effects, particularly mediated by extracellular vesicles, with MSC-derived exosomes (MSC-Exos) being the key mediators. MSC-Exos, enriched with lipids, proteins, and nucleic acids, regulate neuroinflammation by modulating glial cell activity and influencing signaling pathways associated with inflammation and repair. Preclinical evidence has indicated that MSC-Exos can suppress the activation of microglia and astrocytes, promote oligodendrocyte maturation, and enhance myelination, highlighting their potential as a cell-free treatment for PWMI. However, there are a paucity of comprehensive reviews on how MSC-Exos regulate neuroinflammation in PWMI through specific signaling pathways. This review aims to summarize the key signaling pathways through which MSC-Exos modulate neuroinflammation in PWMI and discuss the challenges associated with the clinical application of MSC-Exos-based therapies.
Collapse
Affiliation(s)
- Xinling Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Kangfu Qian Street 7, Zhengzhou, 450052, China
| | - Yuhang Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Kangfu Qian Street 7, Zhengzhou, 450052, China
| | - Xirui Peng
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Kangfu Qian Street 7, Zhengzhou, 450052, China
| | - Luxiang Yang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Kangfu Qian Street 7, Zhengzhou, 450052, China
| | - Jingwen Miao
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Kangfu Qian Street 7, Zhengzhou, 450052, China
| | - Yuyang Yue
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Kangfu Qian Street 7, Zhengzhou, 450052, China
| | - Yong Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Kangfu Qian Street 7, Zhengzhou, 450052, China
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Kangfu Qian Street 7, Zhengzhou, 450052, China
- Center for Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Kangfu Qian Street 7, Zhengzhou, 450052, China.
- Department of Women's and Children's Health, Karolinska Institutet, 17176, Stockholm, Sweden.
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 11, Box 436, 405 30, Gothenburg, Sweden.
| | - Juan Song
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Kangfu Qian Street 7, Zhengzhou, 450052, China.
| |
Collapse
|
25
|
Drozhdev AI, Gorbatenko VO, Goriainov SV, Chistyakov DV, Sergeeva MG. ATP Alters the Oxylipin Profiles in Astrocytes: Modulation by High Glucose and Metformin. Brain Sci 2025; 15:293. [PMID: 40149814 PMCID: PMC11940397 DOI: 10.3390/brainsci15030293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Astrocytes play a key role in the inflammatory process accompanying various neurological diseases. Extracellular ATP accompanies inflammatory processes in the brain, but its effect on lipid mediators (oxylipins) in astrocytes remains elusive. Metformin is a hypoglycemic drug with an anti-inflammatory effect that has been actively investigated in the context of therapy for neuroinflammation, but its mechanisms of action are not fully elucidated. Therefore, we aimed to characterize the effects of ATP on inflammatory markers and oxylipin profiles; determine the dependence of these effects on the adaptation of astrocytes to high glucose levels; and evaluate the possibility of modulating ATP effects using metformin. Methods: We estimated the ATP-mediated response of primary rat astrocytes cultured at normal (NG, 5 mM) and high (HG, 22.5 mM) glucose concentrations for 48 h before stimulation. Cell responses were assessed by monitoring changes in the expression of inflammatory markers (TNFα, IL-6, IL-10, IL-1β, iNOS, and COX-2) and the synthesis of oxylipins (41 compounds), assayed with ultra-high-performance liquid chromatography and tandem mass spectrometry (UPLC-MS/MS). Intracellular pathways were assessed by analyzing the phosphorylation of p38; ERK MAPK; transcription factors STAT3 and NF-κB; and the enzymes mediating oxylipin synthesis, COX-1 and cPLA2. Results: The stimulation of cells with ATP does not affect the expression of pro-inflammatory markers, increases the activities of p38 and ERK MAPKs, and activates oxylipin synthesis, shifting the profiles toward an increase in anti-inflammatory compounds (PGD2, PGA2, 12-HHT, and 18-HEPE). The ATP effects are reduced in HG astrocytes. Metformin potentiated ATP-induced oxylipin synthesis (11-HETE, PGD2, 12-HHT, 15-HETE, 13-HDoHE, and 15-HETrE), which was predominantly evident in NG cells. Conclusions: Our data provide new evidence showing that ATP induces the release of anti-inflammatory oxylipins, and metformin enhances these effects. These results should be considered in the development of anti-inflammatory therapeutic approaches aimed at modulating astrocyte function in various pathologies.
Collapse
Affiliation(s)
- Alexey I. Drozhdev
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.I.D.); (V.O.G.)
| | - Vladislav O. Gorbatenko
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.I.D.); (V.O.G.)
| | - Sergey V. Goriainov
- Institute of Pharmacy and Biotechnology, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia;
| | - Dmitry V. Chistyakov
- Institute of Pharmacy and Biotechnology, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia;
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia;
| | - Marina G. Sergeeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia;
| |
Collapse
|
26
|
Chen Y, Xiao D, Li X. Lactylation and Central Nervous System Diseases. Brain Sci 2025; 15:294. [PMID: 40149815 PMCID: PMC11940311 DOI: 10.3390/brainsci15030294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/01/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
As the final product of glycolysis, lactate serves as an energy substrate, metabolite, and signaling molecule in various diseases and mediates lactylation, an epigenetic modification that occurs under both physiological and pathological conditions. Lactylation is a crucial mechanism by which lactate exerts its functions, participating in vital biological activities such as glycolysis-related cellular functions, macrophage polarization, and nervous system regulation. Lactylation links metabolic regulation to central nervous system (CNS) diseases, such as traumatic brain injury, Alzheimer's disease, acute ischemic stroke, and schizophrenia, revealing the diverse functions of lactylation in the CNS. In the future, further exploration of lactylation-associated enzymes and proteins is needed to develop specific lactylation inhibitors or activators, which could provide new tools and strategies for the treatment of CNS diseases.
Collapse
Affiliation(s)
- Ye Chen
- Department of Emergency Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (D.X.)
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu 610041, China
| | - Dongqiong Xiao
- Department of Emergency Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (D.X.)
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu 610041, China
| | - Xihong Li
- Department of Emergency Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (D.X.)
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu 610041, China
| |
Collapse
|
27
|
Zeng J, Indajang J, Pitt D, Lo CH. Lysosomal acidification impairment in astrocyte-mediated neuroinflammation. J Neuroinflammation 2025; 22:72. [PMID: 40065324 PMCID: PMC11892208 DOI: 10.1186/s12974-025-03410-w] [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: 05/02/2024] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Astrocytes are a major cell type in the central nervous system (CNS) that play a key role in regulating homeostatic functions, responding to injuries, and maintaining the blood-brain barrier. Astrocytes also regulate neuronal functions and survival by modulating myelination and degradation of pathological toxic protein aggregates. Astrocytes have recently been proposed to possess both autophagic activity and active phagocytic capability which largely depend on sufficiently acidified lysosomes for complete degradation of cellular cargos. Defective lysosomal acidification in astrocytes impairs their autophagic and phagocytic functions, resulting in the accumulation of cellular debris, excessive myelin and lipids, and toxic protein aggregates, which ultimately contributes to the propagation of neuroinflammation and neurodegenerative pathology. Restoration of lysosomal acidification in impaired astrocytes represent new neuroprotective strategy and therapeutic direction. In this review, we summarize pathogenic factors, including neuroinflammatory signaling, metabolic stressors, myelin and lipid mediated toxicity, and toxic protein aggregates, that contribute to lysosomal acidification impairment and associated autophagic and phagocytic dysfunction in astrocytes. We discuss the role of lysosomal acidification dysfunction in astrocyte-mediated neuroinflammation primarily in the context of neurodegenerative diseases along with other brain injuries. We then highlight re-acidification of impaired lysosomes as a therapeutic strategy to restore autophagic and phagocytic functions as well as lysosomal degradative capacity in astrocytes. We conclude by providing future perspectives on the role of astrocytes as phagocytes and their crosstalk with other CNS cells to impart neurodegenerative or neuroprotective effects.
Collapse
Affiliation(s)
- Jialiu Zeng
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA.
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, 13244, USA.
| | - Jonathan Indajang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - David Pitt
- Department of Neurology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Chih Hung Lo
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, 13244, USA.
- Department of Biology, Syracuse University, Syracuse, NY, 13244, USA.
| |
Collapse
|
28
|
Zhou H, Dai J, Li D, Wang L, Ye M, Hu X, LoTurco J, Hu J, Sun W. Efficient gene delivery admitted by small metabolites specifically targeting astrocytes in the mouse brain. Mol Ther 2025; 33:1166-1179. [PMID: 39799395 PMCID: PMC11897751 DOI: 10.1016/j.ymthe.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/25/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025] Open
Abstract
The development of efficient and targeted methods for delivering DNA in vivo has long been a major focus of research. In this study, we introduce a gene delivery approach admitted by small metabolites (gDAM) for the efficient and targeted delivery of naked DNA into astrocytes in the adult brains of mice. gDAM uses a straightforward combination of DNA and small metabolites, including glycine, L-proline, L-serine, L-histidine, D-alanine, Gly-Gly, and Gly-Gly-Gly, to achieve astrocyte-specific delivery of naked DNA, resulting in transient and robust gene expression in these cells. Using gDAM, we successfully co-deliver the PiggyBac transposon and the CRISPR-Cas9 system to induce long-term overexpression of the oncogene EGFRvIII and knockout of tumor suppressor genes Nf1, Pten, and Trp53 in astrocytes, leading to the development of astrocyte-derived gliomas in immunocompetent mice. Furthermore, gDAM facilitates the delivery of naked DNA to peripheral glioma astrocytes. The overexpression of interferon-β and granulocyte-macrophage colony-stimulating factor in these peripheral glioma astrocytes significantly prolongs the overall survival of mice bearing 73C glioma cells. This approach offers a new perspective on developing gene delivery systems that specifically target astrocytes to meet the varied needs of both research and gene therapy. The innovative strategy behind gDAM is expected to provide fresh inspiration in the quest for DNA delivery to other tissues, such as skeletal muscle and skin.
Collapse
Affiliation(s)
- Haibin Zhou
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Jiajing Dai
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Dong Li
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Luyao Wang
- Chinese Institute for Brain Research, Beijing 102206, China; College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Meng Ye
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Xiaoling Hu
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Joseph LoTurco
- Department of Physiology and Neurobiology, Institute for Systems Genomics, Institute for Brain and Cognitive Science, University of Connecticut, Storrs, CT 06268, USA
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai 200030, China.
| | - Wenzhi Sun
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Chinese Institute for Brain Research, Beijing 102206, China.
| |
Collapse
|
29
|
Andersen JV. The Glutamate/GABA-Glutamine Cycle: Insights, Updates, and Advances. J Neurochem 2025; 169:e70029. [PMID: 40066661 PMCID: PMC11894596 DOI: 10.1111/jnc.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025]
Abstract
Synaptic homeostasis of the principal neurotransmitters glutamate and GABA is tightly regulated by an intricate metabolic coupling between neurons and astrocytes known as the glutamate/GABA-glutamine cycle. In this cycle, astrocytes take up glutamate and GABA from the synapse and convert these neurotransmitters into glutamine. Astrocytic glutamine is subsequently transferred to neurons, serving as the principal precursor for neuronal glutamate and GABA synthesis. The glutamate/GABA-glutamine cycle integrates multiple cellular processes, including neurotransmitter release, uptake, synthesis, and metabolism. All of these processes are deeply interdependent and closely coupled to cellular energy metabolism. Astrocytes display highly active mitochondrial oxidative metabolism and several unique metabolic features, including glycogen storage and pyruvate carboxylation, which are essential to sustain continuous glutamine release. However, new roles of oligodendrocytes and microglia in neurotransmitter recycling are emerging. Malfunction of the glutamate/GABA-glutamine cycle can lead to severe synaptic disruptions and may be implicated in several brain diseases. Here, I review central aspects and recent advances of the glutamate/GABA-glutamine cycle to highlight how the cycle is functionally connected to critical brain functions and metabolism. First, an overview of glutamate, GABA, and glutamine transport is provided in relation to neurotransmitter recycling. Then, central metabolic aspects of the glutamate/GABA-glutamine cycle are reviewed, with a special emphasis on the critical metabolic roles of glial cells. Finally, I discuss how aberrant neurotransmitter recycling is linked to neurodegeneration and disease, focusing on astrocyte metabolic dysfunction and brain lipid homeostasis as emerging pathological mechanisms. Instead of viewing the glutamate/GABA-glutamine cycle as individual biochemical processes, a more holistic and integrative approach is needed to advance our understanding of how neurotransmitter recycling modulates brain function in both health and disease.
Collapse
Affiliation(s)
- Jens V. Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
30
|
Arndt H, Bachurski M, Yuanxiang P, Franke K, Wessjohann LA, Kreutz MR, Grochowska KM. A Screen of Plant-Based Natural Products Revealed That Quercetin Prevents Pyroglutamylated Amyloid-β (Aβ3(pE)-42) Uptake in Astrocytes As Well As Resulting Astrogliosis and Synaptic Dysfunction. Mol Neurobiol 2025; 62:3730-3745. [PMID: 39317890 PMCID: PMC11790700 DOI: 10.1007/s12035-024-04509-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
Two connected histopathological hallmarks of Alzheimer's disease (AD) are chronic neuroinflammation and synaptic dysfunction. The accumulation of the most prevalent posttranslationally modified form of Aβ1-42, pyroglutamylated amyloid-β (Aβ3(pE)-42) in astrocytes is directly linked to glial activation and the release of proinflammatory cytokines that in turn contribute to early synaptic dysfunction in AD. At present, the mechanisms of Aβ3(pE)-42 uptake to astrocytes are unknown and pharmacological interventions that interfere with this process are not available. Here we developed a simple screening assay to identify substances from a plant extract library that prevent astroglial Aβ3(pE)-42 uptake. We first show that this approach yields valid and reproducible results. Second, we show endocytosis of Aβ3(pE)-42 oligomers by astrocytes and that quercetin, a plant flavonol, is effective to specifically block astrocytic buildup of oligomeric Aβ3(pE)-42. Importantly, quercetin does not induce a general impairment of endocytosis. However, it efficiently protects against early synaptic dysfunction following exogenous Aβ3(pE)-42 application.
Collapse
Affiliation(s)
- Helene Arndt
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Mark Bachurski
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - PingAn Yuanxiang
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Katrin Franke
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, 06108, Halle, Germany
- Institute of Biology/Geobotany and Botanical Garden, Martin Luther University Halle-Wittenberg, 06108, Halle, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, 04103, Leipzig, Germany
| | - Ludger A Wessjohann
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, 06108, Halle, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, 04103, Leipzig, Germany
- Institut Für Chemie, Chair of Natural Products Chemistry, Martin-Luther-University Halle-Wittenberg, 06120, Halle (Saale), Germany
| | - Michael R Kreutz
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany.
- Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
- German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany.
- Center for Behavioral Brain Sciences, Otto Von Guericke University, 39120, Magdeburg, Germany.
| | - Katarzyna M Grochowska
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany.
- Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| |
Collapse
|
31
|
Villa-Cedillo SA, Acosta-Espinoza EJ, Soto-Domínguez A, Rodríguez-Rocha H, Montes-de-Oca-Saucedo CR, García-García A, Loera-Arias MDJ, Ríos-Vazquez CS, Sánchez-Torres G, Valdés J, Saucedo-Cárdenas O. Antioxidant PRDX3 gene therapy protects brain cells and prevents neurodegeneration in an animal model of Parkinson's disease. Neuropeptides 2025; 110:102494. [PMID: 39736192 DOI: 10.1016/j.npep.2024.102494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/06/2024] [Accepted: 12/22/2024] [Indexed: 01/01/2025]
Abstract
Neurodegenerative diseases, including Parkinson's Disease (PD), are a significant global health challenge with no effective therapies to counteract neurodegeneration. Genetic and environmental factors lead to mitochondrial dysfunction and increased reactive oxygen species (ROS), resulting in oxidative stress. This stress reduces levels of Peroxiredoxin 3 (PRDX3), a key protein for maintaining ROS balance at the mitochondrial level, increasing the substantia nigra's susceptibility to damage. To investigate the protective role of antioxidant gene therapy in a PD model, we overexpressed the PRDX3 enzyme using a cell-penetrating peptide-based delivery system (mRVG9R-PRDX3 complex). The mRVG9R peptide was combined with a green fluorescent protein (GFP) reporter plasmid expressing PRDX3 to create the complex. Overexpression of the PRDX3 gene in neuronal phenotype cells was confirmed in vitro using dopaminergic SH-SY5Y cells. Following successful in vitro expression, the mRVG9R-PRDX3 complex was stereotaxically injected into the striatum of male C57BL/6 mice. The PD model was induced by administering paraquat (PQ) twice a week for 6 weeks. After the final PQ injection, motor and cognitive functions were evaluated, followed by histological analysis. Animals treated with the mRVG9R-PRDX3 complex showed a clear reduction in PQ-induced PD symptomatology and prevented cellular senescence in the substantia nigra's neuronal population. The mRVG9R-PRDX3 gene therapy improved motor and cognitive functions in the PD animal model and demonstrated potential in protecting substantia nigra dopaminergic neurons from PQ-induced death.
Collapse
Affiliation(s)
- Sheila Adela Villa-Cedillo
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Departamento de Histología, Monterrey, Nuevo León, Mexico
| | - Esrom Jared Acosta-Espinoza
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Departamento de Histología, Monterrey, Nuevo León, Mexico
| | - Adolfo Soto-Domínguez
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Departamento de Histología, Monterrey, Nuevo León, Mexico
| | - Humberto Rodríguez-Rocha
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Departamento de Histología, Monterrey, Nuevo León, Mexico
| | | | - Aracely García-García
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Departamento de Histología, Monterrey, Nuevo León, Mexico
| | - María de Jesús Loera-Arias
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Departamento de Histología, Monterrey, Nuevo León, Mexico
| | - Cristina Sarahi Ríos-Vazquez
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Departamento de Histología, Monterrey, Nuevo León, Mexico
| | - Guillermo Sánchez-Torres
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Departamento de Histología, Monterrey, Nuevo León, Mexico
| | - Jesús Valdés
- Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Departamento de Bioquímica, México City, Mexico
| | - Odila Saucedo-Cárdenas
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Departamento de Histología, Monterrey, Nuevo León, Mexico.
| |
Collapse
|
32
|
Wen Y, Guo F, Gu T, Zeng Y, Cao X. Transcriptomic Regulation by Astrocytic m6A Methylation in the mPFC. Genes Cells 2025; 30:e70003. [PMID: 39904743 PMCID: PMC11794193 DOI: 10.1111/gtc.70003] [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: 07/03/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 02/06/2025]
Abstract
Astrocytes, the most prevalent type of glial cells, have been found to play a crucial part in numerous physiological functions. By offering metabolic and structural support, astrocytes are vital for the proper functioning of the brain and regulating information processing and synaptic transmission. Astrocytes located in the medial prefrontal cortex (mPFC) are highly responsive to environmental changes and have been associated with the development of brain disorders. One of the primary mechanisms through which the brain responds to environmental factors is epitranscriptome modification. M6-methyladenosine methylation is the most prevalent internal modification of eukaryotic messenger RNA (mRNA), and it significantly impacts transcript processing and protein synthesis. However, the effects of m6A on astrocyte transcription and function are still not well understood. Our research demonstrates that ALKBH5, an RNA demethylase of m6A found in astrocytes, affects gene expression in the mPFC. These findings suggest that further investigation into the potential role of astrocyte-mediated m6A methylation in the mPFC is warranted.
Collapse
Affiliation(s)
- You‐Lu Wen
- Department of Psychology and Behavior, Guangdong 999 Brain Hospital, Institute for Brain Research and RehabilitationSouth China Normal UniversityGuangzhouChina
| | - Fang Guo
- Department of Neurobiology, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Ting‐ting Gu
- Department of Psychology and Behavior, Guangdong 999 Brain Hospital, Institute for Brain Research and RehabilitationSouth China Normal UniversityGuangzhouChina
| | - Yan‐ping Zeng
- Department of Psychology and Behavior, Guangdong 999 Brain Hospital, Institute for Brain Research and RehabilitationSouth China Normal UniversityGuangzhouChina
| | - Xiong Cao
- Department of Neurobiology, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
33
|
Naveed M, Smedlund K, Zhou QG, Cai W, Hill JW. Astrocyte involvement in metabolic regulation and disease. Trends Endocrinol Metab 2025; 36:219-234. [PMID: 39214743 PMCID: PMC11868460 DOI: 10.1016/j.tem.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Astrocytes, the predominant glial cell type in the mammalian brain, influence a wide variety of brain parameters including neuronal energy metabolism. Exciting recent studies have shown that obesity and diabetes can impact on astrocyte function. We review evidence that dysregulation of astrocytic lipid metabolism and glucose sensing contributes to dysregulation of whole-body energy balance, thermoregulation, and insulin sensitivity. In addition, we consider the overlooked topic of the sex-specific roles of astrocytes and their response to hormonal fluctuations that provide insights into sex differences in metabolic regulation. Finally, we provide an update on potential ways to manipulate astrocyte function, including genetic targeting, optogenetic and chemogenetic techniques, transplantation, and tailored exosome-based therapies, which may lead to improved treatments for metabolic disease.
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Physiology and Pharmacology, School of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Kathryn Smedlund
- Department of Physiology and Pharmacology, School of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Qi-Gang Zhou
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Weikang Cai
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY, USA
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, School of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA; Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
34
|
黄 鹏, 陈 洁, 邹 金, 高 雪, 曹 虹. [Quercetin mitigates HIV-1 gp120-induced rat astrocyte neurotoxicity via promoting G3BP1 disassembly in stress granules]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:304-312. [PMID: 40031974 PMCID: PMC11875855 DOI: 10.12122/j.issn.1673-4254.2025.02.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Indexed: 03/05/2025]
Abstract
OBJECTIVES To explore the effect of quercetin for mitigating HIV-1 gp120-induced astrocyte neurotoxicity and its underlying mechanism. METHODS Primary rat astrocytes were isolated and treated with quercetin, HIV-1 gp120, or gradient concentrations of quercetin combined with HIV-1 gp120. The formation of stress granules (SGs) in the treated cells was observed with immunofluorescence assay, and the levels of oxidative stress markers and protein expressions were measured using specific assay kits and Western blotting. HIV-1 gp120 transgenic mice were treated with quercetin (50 mg/kg) by gavage for 4 weeks, and the changes in cognitive functions and oxidative stress levels were examined by behavioral assessments, oxidative stress index analysis in serum, and immunohistochemical and Western blotting of the brain tissue. RESULTS In primary rat astrocytes, treatment with quercetin significantly reduced HIV-1 gp120-induced SG formation, increased the levels of antioxidant indexes, decreased the levels of oxidative substances, and up-regulated protein level associated with SG depolymerization. In the transgenic mouse models, quercetin obviously improved the cognitive function of the rats, reduced oxidative stress levels, and promoted the expression of proteins associate with SG depolymerization in the brain tissues. CONCLUSIONS Quercetin mitigates HIV-1 gp120-induced astrocyte neurotoxicity and cognitive function impairment by inhibiting oxidative stress, enhancing expressions of SG depolymerization-related proteins, and promoting SG disassembly, suggesting the value of quercetin as a potential therapeutic agent for neuroprotection in HIV-associated neurocognitive disorders.
Collapse
|
35
|
Tian Y, Li W, Zhang Y. 3-N-Butylphthalide alleviate Aβ-induced cellular senescence through the CDK2-pRB1-Caspase3 axis. Brain Res 2025; 1849:149435. [PMID: 39736372 DOI: 10.1016/j.brainres.2024.149435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/17/2024] [Accepted: 12/27/2024] [Indexed: 01/01/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the accumulation of amyloid-beta (Aβ) and leading to cellular senescence and cognitive deficits. Cellular senescence contributes significantly to the pathogenesis of AD through the senescence-associated secretory phenotype (SASP), exacerbating Aβ deposition. This study investigates the protective effects of 3-N-Butylphthalide (NBP), a compound derived from Apium graveolens Linn (Chinese celery), on Aβ-induced cellular senescence in U87 cells. Using RNA-sequencing and biochemical assays, we demonstrate that NBP ameliorate Aβ oligomer-induced cellular senescence and apoptosis, and regulated the expression of cyclin-dependent kinase inhibitor 2A (CDKN2A) and components of the cyclin-dependent kinase 2 (CDK2)- phosphorylated retinoblastoma 1 (pRB1)-Caspase3 pathway. Moreover, NBP was shown to suppress the expression of SASP-related genes. These findings suggest that NBP rescues U87 cells from Aβ oligomer-induced senescence and apoptosis through modulating the CDK2-pRB1-Caspase3 axis and SASP expression. Our results underscore the potential of NBP as a senostatic agent for AD which have not been reported in previous studies, offering insights into its mechanisms of action and paving the way for future studies on its efficacy in vivo and in clinical settings. Thus, we contribute to growing evidence supporting the use of senolytic and senostatic agents in the treatment of AD.
Collapse
Affiliation(s)
- Yuanruhua Tian
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Wenke Li
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, the Chinese Academy of Medical Sciences, Beijing 100037, China
| | - Yongbo Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
36
|
Gao X, Xiong Y, Ma H, Zhou H, Liu W, Sun Q. Visualizing bulk autophagy in vivo by tagging endogenous LC3B. Autophagy 2025:1-17. [PMID: 39952286 DOI: 10.1080/15548627.2025.2457910] [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: 08/14/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/17/2025] Open
Abstract
Macroautophagy/autophagy plays a crucial role in maintaining cellular and organismal health, making the measurement of autophagy flux in vivo essential for its study. Current tools often depend on the overexpression of autophagy probes. In this study, we developed a knock-in mouse model, termed tfLC3-KI, by inserting a tandem fluorescent tag coding sequence into the native Map1lc3b gene locus. We found that tfLC3-KI mice exhibit optimal expression of mRFP-eGFP-LC3B, allowing for convenient measurement of autophagic structures and flux at single-cell resolution, both in vivo and in primary cell cultures. Additionally, we compared autophagy in neurons and glial cells across various brain regions between tfLC3-KI mice and CAG-tfLC3 mice, the latter overexpressing the probe under the strong CMV promoter. Finally, we used tfLC3-KI mice to map the spatial and temporal dynamics of basal autophagy activity in the reproductive system. Our findings highlight the value of the tfLC3-KI mouse model for investigating autophagy flux in vivo and demonstrate the feasibility of tagging endogenous proteins to visualize autophagic structures and flux in both bulk and selective autophagy research in vivo.Abbreviation: BafA1: bafilomycin A1; CQ: chloroquine; EBSS: Earle's balanced salt solution; Es: elongating spermatids; HPF: hippocampalformation; HY: hypothalamus; LCs: leydig cells; OLF: olfactory areas; PepA: pepstatin A; Rs: round spermatids; SCs: sertoli cells; Spc: spermatocytes; Spg: spermatogonia; tfLC3: tandem fluorescently tagged mRFP-eGFP-LC3; TH: thalamus.
Collapse
Affiliation(s)
- Xiukui Gao
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Yue Xiong
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Hangbin Ma
- Department of Urology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Hao Zhou
- Department of Urology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Wei Liu
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Qiming Sun
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
- Department of Biochemistry, and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
37
|
Osterman C, Hamlin D, Suter CM, Affleck AJ, Gloss BS, Turner CP, Faull RLM, Stein TD, McKee A, Buckland ME, Curtis MA, Murray HC. Perivascular glial reactivity is a feature of phosphorylated tau lesions in chronic traumatic encephalopathy. Acta Neuropathol 2025; 149:16. [PMID: 39921702 PMCID: PMC11807024 DOI: 10.1007/s00401-025-02854-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025]
Abstract
Chronic traumatic encephalopathy (CTE), a neurodegenerative disease associated with repetitive head injuries, is characterised by perivascular hyperphosphorylated tau (p-tau) accumulations within the depths of cortical sulci. Although the majority of CTE literature focuses on p-tau pathology, other pathological features such as glial reactivity, vascular damage, and axonal damage are relatively unexplored. In this study, we aimed to characterise these other pathological features, specifically in CTE p-tau lesion areas, to better understand the microenvironment surrounding the lesion. We utilised multiplex immunohistochemistry to investigate the distribution of 32 different markers of cytoarchitecture and pathology that are relevant to both traumatic brain injury and neurodegeneration. We qualitatively assessed the multiplex images and measured the percentage area of labelling for each marker in the lesion and non-lesion areas of CTE cases. We identified perivascular glial reactivity as a prominent feature of CTE p-tau lesions, largely driven by increases in astrocyte reactivity compared to non-lesion areas. Furthermore, we identified astrocytes labelled for both NAD(P)H quinone dehydrogenase 1 (NQO1) and L-ferritin, indicating that lesion-associated glial reactivity may be a compensatory response to iron-induced oxidative stress. Our findings demonstrate that perivascular inflammation is a consistent feature of the CTE pathognomonic lesion and may contribute to the pathogenesis of brain injury-related neurodegeneration.
Collapse
Affiliation(s)
- Chelsie Osterman
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Danica Hamlin
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Catherine M Suter
- Department of Neuropathology, Royal Prince Alfred Hospital, 94 Mallet St, Camperdown, NSW, 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Andrew J Affleck
- Department of Neuropathology, Royal Prince Alfred Hospital, 94 Mallet St, Camperdown, NSW, 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Brian S Gloss
- Westmead Research Hub, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Clinton P Turner
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
- Department of Anatomical Pathology, Pathology and Laboratory Medicine, Auckland City Hospital, 2 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Thor D Stein
- Department of Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston, MA, USA
- Department of Pathology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Alzheimer's Disease and CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Bedford Veterans Affairs Medical Center, Bedford, MA, USA
| | - Ann McKee
- Department of Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston, MA, USA
- Department of Pathology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Alzheimer's Disease and CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Bedford Veterans Affairs Medical Center, Bedford, MA, USA
| | - Michael E Buckland
- Department of Neuropathology, Royal Prince Alfred Hospital, 94 Mallet St, Camperdown, NSW, 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Maurice A Curtis
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Helen C Murray
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand.
| |
Collapse
|
38
|
Huang Y, Zhang G, Li S, Feng J, Zhang Z. Innate and adaptive immunity in neurodegenerative disease. Cell Mol Life Sci 2025; 82:68. [PMID: 39894884 PMCID: PMC11788272 DOI: 10.1007/s00018-024-05533-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 02/04/2025]
Abstract
Neurodegenerative diseases (NDs) are a group of neurological disorders characterized by the progressive loss of selected neurons. Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common NDs. Pathologically, NDs are characterized by progressive failure of neural interactions and aberrant protein fibril aggregation and deposition, which lead to neuron loss and cognitive and behavioral impairments. Great efforts have been made to delineate the underlying mechanism of NDs. However, the very first trigger of these disorders and the state of the illness are still vague. Existing therapeutic strategies can relieve symptoms but cannot cure these diseases. The human immune system is a complex and intricate network comprising various components that work together to protect the body against pathogens and maintain overall health. They can be broadly divided into two main types: innate immunity, the first line of defense against pathogens, which acts nonspecifically, and adaptive immunity, which follows a defense process that acts more specifically and is targeted. The significance of brain immunity in maintaining the homeostatic environment of the brain, and its direct implications in NDs, has increasingly come into focus. Some components of the immune system have beneficial regulatory effects, whereas others may have detrimental effects on neurons. The intricate interplay and underlying mechanisms remain an area of active research. This review focuses on the effects of both innate and adaptive immunity on AD and PD, offering a comprehensive understanding of the initiation and regulation of brain immunity, as well as the interplay between innate and adaptive immunity in influencing the progression of NDs.
Collapse
Affiliation(s)
- Yeyu Huang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guoxin Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Sheng Li
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jin Feng
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
39
|
Derevyanko A, Tao T, Allen NJ. Common alterations to astrocytes across neurodegenerative disorders. Curr Opin Neurobiol 2025; 90:102970. [PMID: 39879721 DOI: 10.1016/j.conb.2025.102970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 12/16/2024] [Accepted: 01/02/2025] [Indexed: 01/31/2025]
Abstract
Astrocytes perform multiple functions in the nervous system, many of which are altered in neurodegenerative disorders. In this review, we explore shared astrocytic alterations across neurodegenerative disorders, including Alzheimer's disease, Huntington's disease, Parkinson's disease, amyotrophic lateral sclerosis, and frontotemporal lobe degeneration. Assessing recent datasets of single-nucleus RNA-sequencing of human brains, a theme emerges of common alterations in astrocyte state across disorders including in neuroinflammation, synaptic organization, metabolic support, and the cellular stress response. Immune pathways are upregulated by astrocytes across disorders and may exacerbate neurodegeneration. Dysregulated expression of synaptogenic factors could contribute to synaptic loss, while compromised metabolic support affects neuronal homeostasis. On the other hand, upregulated responses to cellular stress may represent a protective response of astrocytes and thus mitigate pathology. Understanding these shared responses offers insights into disease progression and provides potential therapeutic targets for various neurodegenerative disorders.
Collapse
Affiliation(s)
- Aksinya Derevyanko
- Salk Institute for Biological Studies, Molecular Neurobiology Laboratory, 10010 North Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Tao Tao
- Salk Institute for Biological Studies, Molecular Neurobiology Laboratory, 10010 North Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Nicola J Allen
- Salk Institute for Biological Studies, Molecular Neurobiology Laboratory, 10010 North Torrey Pines Rd, La Jolla, CA, 92037, USA.
| |
Collapse
|
40
|
Bobkova NV, Chuvakova LN, Kovalev VI, Zhdanova DY, Chaplygina AV, Rezvykh AP, Evgen'ev MB. A Mouse Model of Sporadic Alzheimer's Disease with Elements of Major Depression. Mol Neurobiol 2025; 62:1337-1358. [PMID: 38980563 DOI: 10.1007/s12035-024-04346-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
After olfactory bulbectomy, animals are often used as a model of major depression or sporadic Alzheimer's disease and, hence, the status of this model is still disputable. To elucidate the nature of alterations in the expression of the genome after the operation, we analyzed transcriptomes of the cortex, hippocampus, and cerebellum of the olfactory bulbectomized (OBX) mice. Analysis of the functional significance of genes in the brain of OBX mice indicates that the balance of the GABA/glutamatergic systems is disturbed with hyperactivation of the latter in the hippocampus, leading to the development of excitotoxicity and induction of apoptosis in the background of severe mitochondrial dysfunction and astrogliosis. On top of this, the synthesis of neurotrophic factors decreases leading to the disruption of the cytoskeleton of neurons, an increase in the level of intracellular calcium, and the activation of tau protein hyperphosphorylation. Moreover, the acetylcholinergic system is deficient in the background of the hyperactivation of acetylcholinesterase. Importantly, the activity of the dopaminergic, endorphin, and opiate systems in OBX mice decreases, leading to hormonal dysfunction. On the other hand, genes responsible for the regulation of circadian rhythms, cell migration, and innate immunity are activated in OBX animals. All this takes place in the background of a drastic downregulation of ribosomal protein genes in the brain. The obtained results indicate that OBX mice represent a model of Alzheimer's disease with elements of major depression.
Collapse
Affiliation(s)
- N V Bobkova
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - L N Chuvakova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - V I Kovalev
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - D Y Zhdanova
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - A V Chaplygina
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - A P Rezvykh
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - M B Evgen'ev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia.
| |
Collapse
|
41
|
Chen L, Jiao J, Lei F, Zhou B, Li H, Liao P, Li X, Kang Y, Liu J, Jiang R. Ezrin-mediated astrocyte-synapse signaling regulates cognitive function via astrocyte morphological changes in fine processes in male mice. Brain Behav Immun 2025; 124:177-191. [PMID: 39580057 DOI: 10.1016/j.bbi.2024.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024] Open
Abstract
Astrocytes, which actively participate in cognitive processes, have a complex spongiform morphology, highlighted by extensive ramified fine processes that closely enwrap the pre- and post-synaptic compartments, forming tripartite synapses. However, the role of astrocyte morphology in cognitive processes remains incompletely understood and even controversial. The actin-binding protein Ezrin is highly expressed in astrocytes and is a key structural determinant of astrocyte morphology. Here, we found that Ezrin expression and astrocyte fine process volume in the hippocampus of male mice increased after learning but decreased after lipopolysaccharide injection and in a mouse model of postoperative cognitive dysfunction, both of which involved models with impaired cognitive function. Additionally, astrocytic Ezrin knock-out led to significantly decreased astrocytic fine process volumes, decreased astrocyte-neuron proximity, and induced anxiety-like behaviors and cognitive dysfunction. Astrocytic Ezrin deficiency in the hippocampus was achieved by using a microRNA silencing technique delivered by adeno-associated viruses. Down-regulation of Ezrin in hippocampal astrocytes led to disrupted astrocyte-synapse interactions and impaired synaptic functions, including synaptic transmission and synaptic plasticity, which could be rescued by exogenous administration of D-serine. Remarkably, decreased Ezrin expression and reduced astrocyte fine processes volumes were also observed in aged mice with decreased cognitive function. Moreover, overexpression of astrocytic Ezrin increased astrocyte fine process volumes and improved cognitive function in aged mice. Overall, our results indicate Ezrin-mediated astrocyte fine processes integrity shapes astrocyte-synapse signaling contributing to cognitive function.
Collapse
Affiliation(s)
- Lingmin Chen
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiao Jiao
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Fan Lei
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Bin Zhou
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hong Li
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ping Liao
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xin Li
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yi Kang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Ruotian Jiang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
42
|
Luo S, Wang Y, Hisatsune T. P2Y1 receptor in Alzheimer's disease. Neural Regen Res 2025; 20:440-453. [PMID: 38819047 PMCID: PMC11317937 DOI: 10.4103/nrr.nrr-d-23-02103] [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: 12/29/2023] [Revised: 03/15/2024] [Accepted: 03/28/2024] [Indexed: 06/01/2024] Open
Abstract
Alzheimer's disease is the most frequent form of dementia characterized by the deposition of amyloid-beta plaques and neurofibrillary tangles consisting of hyperphosphorylated tau. Targeting amyloid-beta plaques has been a primary direction for developing Alzheimer's disease treatments in the last decades. However, existing drugs targeting amyloid-beta plaques have not fully yielded the expected results in the clinic, necessitating the exploration of alternative therapeutic strategies. Increasing evidence unravels that astrocyte morphology and function alter in the brain of Alzheimer's disease patients, with dysregulated astrocytic purinergic receptors, particularly the P2Y1 receptor, all of which constitute the pathophysiology of Alzheimer's disease. These receptors are not only crucial for maintaining normal astrocyte function but are also highly implicated in neuroinflammation in Alzheimer's disease. This review delves into recent insights into the association between P2Y1 receptor and Alzheimer's disease to underscore the potential neuroprotective role of P2Y1 receptor in Alzheimer's disease by mitigating neuroinflammation, thus offering promising avenues for developing drugs for Alzheimer's disease and potentially contributing to the development of more effective treatments.
Collapse
Affiliation(s)
- Shan Luo
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| | - Yifei Wang
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| | - Tatsuhiro Hisatsune
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| |
Collapse
|
43
|
Zheng Q, Wang X. Alzheimer's disease: insights into pathology, molecular mechanisms, and therapy. Protein Cell 2025; 16:83-120. [PMID: 38733347 PMCID: PMC11786724 DOI: 10.1093/procel/pwae026] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
Alzheimer's disease (AD), the leading cause of dementia, is characterized by the accumulation of amyloid plaques and neurofibrillary tangles in the brain. This condition casts a significant shadow on global health due to its complex and multifactorial nature. In addition to genetic predispositions, the development of AD is influenced by a myriad of risk factors, including aging, systemic inflammation, chronic health conditions, lifestyle, and environmental exposures. Recent advancements in understanding the complex pathophysiology of AD are paving the way for enhanced diagnostic techniques, improved risk assessment, and potentially effective prevention strategies. These discoveries are crucial in the quest to unravel the complexities of AD, offering a beacon of hope for improved management and treatment options for the millions affected by this debilitating disease.
Collapse
Affiliation(s)
- Qiuyang Zheng
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Xin Wang
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| |
Collapse
|
44
|
Sha R, Wu M, Wang P, Chen Z, Lei W, Wang S, Gong S, Liang G, Zhao R, Tao Y. Adolescent mice exposed to TBI developed PD-like pathology in middle age. Transl Psychiatry 2025; 15:27. [PMID: 39863574 PMCID: PMC11763066 DOI: 10.1038/s41398-025-03232-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 12/08/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Traumatic brain injury (TBI) is identified as a risk factor for Parkinson's disease (PD), which is a neurodegenerative disease characterized by the loss of dopaminergic neurons in the substantia nigra (SN). However, the precise mechanism by which chronic TBI initiates PD pathogenesis is not yet fully understood. In our present study, we assessed the chronic progression and pathogenesis of PD-like behavior at different intervals in TBI mice. More than half of the mice exhibited PD-like behavior at 6 months post injury. PD-like behavioral dysfunction and pathological changes were aggravated with the injured time extension in chronic phase of TBI. The loss of tyrosine hydroxylase positive (TH+) neurons in the SN were partly associated with the accumulation of misfolded a-Synuclein and the cytoplasmic translocation of TDP-43 from nuclear. Moreover, the present of chronic inflammation was observed in SN of TBI mice, as evidenced by the enhancement of proinflammatory cytokines and reactive astrocytes and microgliosis post lesion. The enhanced phagocytosis of reactive microglia accounted for the reduction of dendrite spines. Our results revealed that chronic inflammation associated with the damage of TH+ neurons and the development of progressive PD-like pathology after chronic TBI in mice. Our study shed new light on the TBI-triggered molecular events on PD-like pathology. Additional research is required to have a deeper understanding of the molecular factors underlying the impairment of dopaminergic neurons following TBI.
Collapse
Affiliation(s)
- Rong Sha
- Department of Neurosurgery, General Hospital of Northern Theater Command, Postgraduate Training Base of General Hospital of Northern Theater Command of Jinzhou Medical University, Shenyang, Liaoning, China
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, China
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mingzhe Wu
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, China
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, China
| | - Pengfei Wang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, China
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, Liaoning, China
| | - Ziyuan Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, China
| | - Wei Lei
- Department of Neurosurgery, General Hospital of Northern Theater Command, Postgraduate Training Base of General Hospital of Northern Theater Command of Jinzhou Medical University, Shenyang, Liaoning, China
| | - Shimiao Wang
- Department of Neurosurgery, General Hospital of Northern Theater Command, Postgraduate Training Base of General Hospital of Northern Theater Command of Jinzhou Medical University, Shenyang, Liaoning, China
| | - Shun Gong
- Department of Neurosurgery, General Hospital of Northern Theater Command, Postgraduate Training Base of General Hospital of Northern Theater Command of Jinzhou Medical University, Shenyang, Liaoning, China.
| | - Guobiao Liang
- Department of Neurosurgery, General Hospital of Northern Theater Command, Postgraduate Training Base of General Hospital of Northern Theater Command of Jinzhou Medical University, Shenyang, Liaoning, China.
- China Medical University, Shenyang, Liaoning, China.
| | - Rui Zhao
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, China.
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, China.
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, Liaoning, China.
| | - Yingqun Tao
- Department of Neurosurgery, General Hospital of Northern Theater Command, Postgraduate Training Base of General Hospital of Northern Theater Command of Jinzhou Medical University, Shenyang, Liaoning, China.
- China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
45
|
Lee E, Chang Y. Modulating Neuroinflammation as a Prospective Therapeutic Target in Alzheimer's Disease. Cells 2025; 14:168. [PMID: 39936960 PMCID: PMC11817173 DOI: 10.3390/cells14030168] [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: 11/26/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
The recent approval of lecanemab highlights that the amyloid beta (Aβ) protein is an important pathological target in Alzheimer's disease (AD) and further emphasizes the significance of neuroinflammatory pathways in regulating Aβ accumulation. Indeed, Aβ accumulation triggers microglia activation, which are key mediators in neuroinflammation. The inflammatory responses in this process can lead to neuronal damage and functional decline. Microglia secrete proinflammatory cytokines that accelerate neuronal death and release anti-inflammatory cytokines and growth factors contributing to neuronal recovery and protection. Thus, microglia play a dual role in neurodegeneration and neuroprotection, complicating their function in AD. Therefore, elucidating the complex interactions between Aβ protein, microglia, and neuroinflammation is essential for developing new strategies for treating AD. This review investigates the receptors and pathways involved in activating microglia and aims to enhance understanding of how these processes impact neuroinflammation in AD, as well as how they can be regulated. This review also analyzed studies reported in the existing literature and ongoing clinical trials. Overall, these studies will contribute to understanding the regulatory mechanisms of neuroinflammation and developing new therapies that can slow the pathological progression of AD.
Collapse
Affiliation(s)
- Eunshil Lee
- Institute of Biomedical Engineering Research, Kyungpook National University, Daegu 41944, Republic of Korea;
| | - Yongmin Chang
- Institute of Biomedical Engineering Research, Kyungpook National University, Daegu 41944, Republic of Korea;
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Radiology, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| |
Collapse
|
46
|
Jiao W, Lin J, Deng Y, Ji Y, Liang C, Wei S, Jing X, Yan F. The immunological perspective of major depressive disorder: unveiling the interactions between central and peripheral immune mechanisms. J Neuroinflammation 2025; 22:10. [PMID: 39828676 PMCID: PMC11743025 DOI: 10.1186/s12974-024-03312-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 11/26/2024] [Indexed: 01/22/2025] Open
Abstract
Major depressive disorder is a prevalent mental disorder, yet its pathogenesis remains poorly understood. Accumulating evidence implicates dysregulated immune mechanisms as key contributors to depressive disorders. This review elucidates the complex interplay between peripheral and central immune components underlying depressive disorder pathology. Peripherally, systemic inflammation, gut immune dysregulation, and immune dysfunction in organs including gut, liver, spleen and adipose tissue influence brain function through neural and molecular pathways. Within the central nervous system, aberrant microglial and astrocytes activation, cytokine imbalances, and compromised blood-brain barrier integrity propagate neuroinflammation, disrupting neurotransmission, impairing neuroplasticity, and promoting neuronal injury. The crosstalk between peripheral and central immunity creates a vicious cycle exacerbating depressive neuropathology. Unraveling these multifaceted immune-mediated mechanisms provides insights into major depressive disorder's pathogenic basis and potential biomarkers and targets. Modulating both peripheral and central immune responses represent a promising multidimensional therapeutic strategy.
Collapse
Affiliation(s)
- Wenli Jiao
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
| | - Jiayi Lin
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
| | - Yanfang Deng
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yelin Ji
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
| | - Chuoyi Liang
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
| | - Sijia Wei
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
| | - Xi Jing
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geoscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, Guangdong, China.
| | - Fengxia Yan
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
47
|
Wen H, Deng H, Li B, Chen J, Zhu J, Zhang X, Yoshida S, Zhou Y. Mitochondrial diseases: from molecular mechanisms to therapeutic advances. Signal Transduct Target Ther 2025; 10:9. [PMID: 39788934 PMCID: PMC11724432 DOI: 10.1038/s41392-024-02044-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/28/2024] [Accepted: 10/31/2024] [Indexed: 01/12/2025] Open
Abstract
Mitochondria are essential for cellular function and viability, serving as central hubs of metabolism and signaling. They possess various metabolic and quality control mechanisms crucial for maintaining normal cellular activities. Mitochondrial genetic disorders can arise from a wide range of mutations in either mitochondrial or nuclear DNA, which encode mitochondrial proteins or other contents. These genetic defects can lead to a breakdown of mitochondrial function and metabolism, such as the collapse of oxidative phosphorylation, one of the mitochondria's most critical functions. Mitochondrial diseases, a common group of genetic disorders, are characterized by significant phenotypic and genetic heterogeneity. Clinical symptoms can manifest in various systems and organs throughout the body, with differing degrees and forms of severity. The complexity of the relationship between mitochondria and mitochondrial diseases results in an inadequate understanding of the genotype-phenotype correlation of these diseases, historically making diagnosis and treatment challenging and often leading to unsatisfactory clinical outcomes. However, recent advancements in research and technology have significantly improved our understanding and management of these conditions. Clinical translations of mitochondria-related therapies are actively progressing. This review focuses on the physiological mechanisms of mitochondria, the pathogenesis of mitochondrial diseases, and potential diagnostic and therapeutic applications. Additionally, this review discusses future perspectives on mitochondrial genetic diseases.
Collapse
Affiliation(s)
- Haipeng Wen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Hui Deng
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Bingyan Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Junyu Chen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Junye Zhu
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Xian Zhang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Shigeo Yoshida
- Department of Ophthalmology, Kurume University School of Medicine, Kurume, Fukuoka, 830-0011, Japan
| | - Yedi Zhou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| |
Collapse
|
48
|
Wang M, Zhang H, Liang J, Huang J, Wu T, Chen N. Calcium signaling hypothesis: A non-negligible pathogenesis in Alzheimer's disease. J Adv Res 2025:S2090-1232(25)00026-8. [PMID: 39793962 DOI: 10.1016/j.jare.2025.01.007] [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: 08/25/2024] [Revised: 11/23/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) presents a significant challenge to global healthcare systems, with an exacerbation by an aging population. Although the plethora of hypotheses are proposed to elucidate the underlying mechanisms of AD, from amyloid-beta (Aβ) accumulation and Tau protein aggregation to neuroinflammation, a comprehensive understanding of its pathogenesis remains elusive. Recent research has highlighted the critical role of calcium (Ca2+) signaling pathway in the progression of AD, indicating a complex interplay between Ca2+ dysregulation and various pathological processes. AIM OF REVIEW This review aims to consolidate the current understanding of the role of Ca2+ signaling dysregulation in AD, thus emphasizing its central role amidst various pathological hypotheses. We aim to evaluate the potential of the Ca2+ signaling hypothesis to unify existing theories of AD pathogenesis and explore its implications for developing innovative therapeutic strategies through targeting Ca2+ dysregulation. KEY SCIENTIFIC CONCEPTS OF REVIEW The review focuses on three principal concepts. First, the indispensable role of Ca2+ homeostasis in neuronal function and its disruption in AD. Second, the interaction between Ca2+ signaling dysfunction and established AD hypotheses posited that Ca2+ dysregulation is a unifying pathway. Third, the dual role of Ca2+ in neurodegeneration and neuroprotection, highlighting the nuanced effects of Ca2+ levels on AD pathology.
Collapse
Affiliation(s)
- Minghui Wang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Hu Zhang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Jiling Liang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Jielun Huang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Tong Wu
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China.
| | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China.
| |
Collapse
|
49
|
Labarta-Bajo L, Allen NJ. Astrocytes in aging. Neuron 2025; 113:109-126. [PMID: 39788083 PMCID: PMC11735045 DOI: 10.1016/j.neuron.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/05/2024] [Accepted: 12/11/2024] [Indexed: 01/12/2025]
Abstract
The mammalian nervous system is impacted by aging. Aging alters brain architecture, is associated with molecular damage, and can manifest with cognitive and motor deficits that diminish the quality of life. Astrocytes are glial cells of the CNS that regulate the development, function, and repair of neural circuits during development and adulthood; however, their functions in aging are less understood. Astrocytes change their transcriptome during aging, with astrocytes in areas such as the cerebellum, the hypothalamus, and white matter-rich regions being the most affected. While numerous studies describe astrocyte transcriptional changes in aging, many questions still remain. For example, how is astrocyte function altered by transcriptional changes that occur during aging? What are the mechanisms promoting astrocyte aged states? How do aged astrocytes impact brain function? This review discusses features of aged astrocytes and their potential triggers and proposes ways in which they may impact brain function and health span.
Collapse
Affiliation(s)
- Lara Labarta-Bajo
- Salk Institute for Biological Studies, Molecular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Nicola J Allen
- Salk Institute for Biological Studies, Molecular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
50
|
Kavakli E, Gul N, Begentas OC, Kiris E. Astrocytes in Primary Familial Brain Calcification (PFBC): Emphasis on the Importance of Induced Pluripotent Stem Cell-Derived Human Astrocyte Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1479:19-38. [PMID: 39841380 DOI: 10.1007/5584_2024_840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Primary familial brain calcification (PFBC) is a rare, progressive central nervous system (CNS) disorder without a cure, and the current treatment methodologies primarily aim to relieve neurological and psychiatric symptoms of the patients. The disease is characterized by abnormal bilateral calcifications in the brain, however, our mechanistic understanding of the biology of the disease is still limited. Determining the roles of the specific cell types and molecular mechanisms involved in the pathophysiological processes of the disease is of great importance for the development of novel and effective treatment methodologies. There is a growing interest in the involvement of astrocytes in PFBC, as recent studies have suggested that astrocytes play a central role in the disease and that functional defects in these cells are critical for the development and progression of the disease. This review aims to discuss recent findings on the roles of astrocytes in PFBC pathophysiology, with a focus on known expression and roles of PFBC genes in astrocytes. Additionally, we discuss the importance of human astrocytes for PFBC disease modeling, and astrocytes as a potential therapeutic target in PFBC. Utilization of species-specific and physiologically relevant PFBC model systems can open new avenues for basic research, drug development, and regenerative medicine.
Collapse
Affiliation(s)
- Ebru Kavakli
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| | - Nazli Gul
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| | - Onur Can Begentas
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| | - Erkan Kiris
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye.
| |
Collapse
|