101
|
Lemarchand E, Grayston A, Wong R, Rogers M, Ouvrier B, Llewellyn B, Webb F, Lénárt N, Dénes Á, Brough D, Allan SM, Bix GJ, Pinteaux E. Selective deletion of interleukin-1 alpha in microglia does not modify acute outcome but may regulate neurorepair processes after experimental ischemic stroke. J Cereb Blood Flow Metab 2025:271678X251323371. [PMID: 40110693 PMCID: PMC11926816 DOI: 10.1177/0271678x251323371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/22/2025] [Accepted: 02/09/2025] [Indexed: 03/22/2025]
Abstract
Inflammation is a key contributor to stroke pathogenesis and exacerbates brain damage leading to poor outcome. Interleukin-1 (IL-1) is an important regulator of post-stroke inflammation, and blocking its actions is beneficial in pre-clinical stroke models and safe in the clinical setting. However, the distinct roles of the two major IL-1 receptor type 1 agonists, IL-1α and IL-1β, and the specific role of IL-1α in ischemic stroke remain largely unknown. Here we show that IL-1α and IL-1β have different spatio-temporal expression profiles in the brain after experimental stroke, with early microglial IL-1α expression (4 h) and delayed IL-1β expression in infiltrated neutrophils and a small microglial subset (24-72 h). We examined for the first time the specific role of microglial-derived IL-1α in experimental permanent and transient ischemic stroke through microglial-specific tamoxifen-inducible Cre-loxP-mediated recombination. Microglial IL-1α deletion did not influence acute outcome after ischemic stroke. However, microglial IL-1α knock out (KO) mice showed reduced peri-infarct vessel density and reactive astrogliosis at 14 days post-stroke, alongside long-term impaired functional recovery. Our study identifies for the first time a critical role for microglial IL-1α on post-stroke neurorepair and recovery, highlighting the importance of targeting specific IL-1 mechanisms in brain injury to develop effective therapies.
Collapse
Affiliation(s)
- Eloïse Lemarchand
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Alba Grayston
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Raymond Wong
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Miyako Rogers
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Blake Ouvrier
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Orleans, LA, USA
| | - Benjamin Llewellyn
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Freddie Webb
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Nikolett Lénárt
- “Momentum” Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Ádám Dénes
- “Momentum” Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - David Brough
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Stuart M Allan
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Gregory J Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Orleans, LA, USA
| | - Emmanuel Pinteaux
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
102
|
Kour D, Khajuria P, Sharma K, Sharma A, Sharma A, Ali SM, Wazir P, Ramajayan P, Sawant SD, Nandi U, Ahmed Z, Kumar A. Isobavachalcone ameliorates Alzheimer disease pathology by autophagy-mediated clearance of amyloid beta and inhibition of NLRP3 inflammasome in primary astrocytes and 5x-FAD mice. Front Pharmacol 2025; 16:1525364. [PMID: 40183098 PMCID: PMC11965660 DOI: 10.3389/fphar.2025.1525364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Background and Aim Alzheimer's disease (AD) progresses with Aβ plaque deposition and neuroinflammation. Given the complexity of AD pathology, single-target therapies have frequently failed in clinical trials. We hypothesized that a multitarget approach could yield better therapeutic outcomes. To this end, we identified isobavachalcone (IBC), a natural compound with dual pharmacological activity in reducing Aβ plaques and neuroinflammation. Experimental Procedure Primary astrocytes were isolated from 3 to 4 days old C57BL/6J mice pups for in-vitro assays, while in-vivo studies were conducted on 5x-FAD mice. Protein alterations were evaluated using ELISA, western blotting, immunocytochemistry, and immunohistochemistry. Behavioral analyses included the radial arm maze, open field, and rotarod tests. Data from all in vitro and in vivo experiments were analyzed by using one-way ANOVA and post-hoc Bonferroni tests. Results In-vitro analyses in astrocytes demonstrated that IBC at 5 and 10 μM concentrations induce AMPK phosphorylation through CAMKK2, promoting autophagy and inhibiting the NLRP3 inflammasome in primary astrocytes. IBC-treated astrocytes exhibited significant clearance of extracellular amyloid beta. Mechanistic studies highlighted autophagy as a key factor in reducing both NLRP3 inflammasome activity and Aβ levels. Two months of treatment of 5x-FAD mice with IBC at 25 and 50 mg/kg significantly improved cognitive functions, as evidenced by enhanced memory and motor performance in behavioral tests. Subsequent brain tissue analysis revealed that IBC upregulated autophagic proteins to reduce the brain's amyloid beta levels, resulting in decreased expression of neuroinflammation markers. Conclusion IBC effectively ameliorates AD pathology through autophagy-mediated clearance of Aβ and suppressing neuroinflammation in 5x-FAD mice.
Collapse
Affiliation(s)
- Dilpreet Kour
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Parul Khajuria
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kuhu Sharma
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Alpa Sharma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Ankita Sharma
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Syed Mudassir Ali
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Priya Wazir
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - P. Ramajayan
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sanghapal D. Sawant
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Organic Chemsitry Division, CSIR-National Chemical Laboratory, Pune, India
| | - Utpal Nandi
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Zabeer Ahmed
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ajay Kumar
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
103
|
Cognacq G, Attwood JE, DeLuca GC. Traumatic Brain Injury and Alzheimer's Disease: A Shared Neurovascular Hypothesis. Neurosci Insights 2025; 20:26331055251323292. [PMID: 40124421 PMCID: PMC11926848 DOI: 10.1177/26331055251323292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Traumatic brain injury (TBI) is a modifiable risk factor for Alzheimer's disease (AD). TBI and AD share several histopathological hallmarks: namely, beta-amyloid aggregation, tau hyperphosphorylation, and plasma protein infiltration. The relative contributions of these proteinopathies and their interplay in the pathogenesis of both conditions remains unclear although important differences are emerging. This review synthesises emerging evidence for the critical role of the neurovascular unit in mediating protein accumulation and neurotoxicity in both TBI and AD. We propose a shared pathogenic cascade centred on a neurovascular unit, in which increased blood-brain barrier permeability induces a series of noxious mechanisms leading to neuronal loss, synaptic dysfunction and ultimately cognitive dysfunction in both conditions. We explore the application of this hypothesis to outstanding research questions and potential treatments for TBI and AD, as well as other neurodegenerative and neuroinflammatory conditions. Limitations of this hypothesis, including the challenges of establishing a causal relationship between neurovascular damage and proteinopathies, are also discussed.
Collapse
Affiliation(s)
- Gabrielle Cognacq
- John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, Oxfordshire, UK
| | - Jonathan E Attwood
- Nuffield Department of Clinical Neurosciences, Level 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, Oxfordshire, UK
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, Level 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, Oxfordshire, UK
| |
Collapse
|
104
|
Zhou X, Jing XJ, Zhang H. The Potential Role of Neurogranin in Alzheimer's Disease. J Integr Neurosci 2025; 24:25368. [PMID: 40152561 DOI: 10.31083/jin25368] [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: 06/25/2024] [Revised: 08/29/2024] [Accepted: 09/23/2024] [Indexed: 03/29/2025] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is characterized by the excessive deposition of amyloid-β (Aβ) plaques and the formation of neurofibrillary tangles. Numerous new studies also indicate that synaptic damage and loss play crucial roles in AD and form the basis of cognitive impairment. In recent years, synaptic-related proteins have emerged as important biomarkers for the early diagnosis of AD. Among these proteins, neurogranin (Ng), a postsynaptic protein widely present in the dendritic spines of the associative cortex in the brain, plays a significant role in memory, learning, synaptic plasticity, and long-term potentiation (LTP). This review aims to reveal the link between Ng and AD, as well as the potential for the diagnosis of AD, the prediction of the development of the disease, and the identification of a therapeutic target for AD.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 400042 Chongqing, China
| | - Xiao-Jun Jing
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 400042 Chongqing, China
| | - Hua Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 400042 Chongqing, China
| |
Collapse
|
105
|
Komleva Y, Shpiliukova K, Bondar N, Salmina A, Khilazheva E, Illarioshkin S, Piradov M. Decoding brain aging trajectory: predictive discrepancies, genetic susceptibilities, and emerging therapeutic strategies. Front Aging Neurosci 2025; 17:1562453. [PMID: 40177249 PMCID: PMC11962000 DOI: 10.3389/fnagi.2025.1562453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
The global extension of human lifespan has intensified the focus on aging, yet its underlying mechanisms remain inadequately understood. The article highlights aspects of genetic susceptibility to impaired brain bioenergetics, trends in age-related gene expression related to neuroinflammation and brain senescence, and the impact of stem cell exhaustion and quiescence on accelerated brain aging. We also review the accumulation of senescent cells, mitochondrial dysfunction, and metabolic disturbances as central pathological processes in aging, emphasizing how these factors contribute to inflammation and disrupt cellular competition defining the aging trajectory. Furthermore, we discuss emerging therapeutic strategies and the future potential of integrating advanced technologies to refine aging assessments. The combination of several methods including genetic analysis, neuroimaging techniques, cognitive tests and digital twins, offer a novel approach by simulating and monitoring individual health and aging trajectories, thereby providing more accurate and personalized insights. Conclusively, the accurate estimation of brain aging trajectories is crucial for understanding and managing aging processes, potentially transforming preventive and therapeutic strategies to improve health outcomes in aging populations.
Collapse
Affiliation(s)
| | | | - Nikolai Bondar
- Research Center of Neurology, Moscow, Russia
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | - Elena Khilazheva
- Department of Biological Chemistry with Courses in Medical, Research Institute of Molecular Medicine and Pathobiochemistry, Pharmaceutical and Toxicological Chemistry Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University of the Ministry of Healthcare of the Russian Federation, Krasnoyarsk, Russia
| | | | | |
Collapse
|
106
|
Ammothumkandy A, Cayce A, Shariq M, Bonaguidi MA. Astroglia's role in synchronized spontaneous neuronal activity: from physiology to pathology. Front Cell Neurosci 2025; 19:1544460. [PMID: 40177583 PMCID: PMC11961896 DOI: 10.3389/fncel.2025.1544460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
The nervous system relies on a balance of excitatory and inhibitory signals. Aberrant neuronal hyperactivity is a pathological phenotype associated with several neurological disorders, with its most severe effects observed in epilepsy patients. This review explores the literature on spontaneous synchronized neuronal activity, its physiological role, and its aberrant forms in disease. Emphasizing the importance of targeting underlying disease mechanisms beyond traditional neuron-focused therapies, the review delves into the role of astroglia in epilepsy progression. We detail how astroglia transitions from a normal to a pathological state, leading to epileptogenic seizures and cognitive decline. Astroglia activity is correlated with epileptiform activity in both animal models and human tissue, indicating their potential role in seizure induction and modulation. Understanding astroglia's dual beneficial and detrimental roles could lead to novel treatments for epilepsy and other neurological disorders with aberrant neuronal activity as the underlying disease substrate.
Collapse
Affiliation(s)
- Aswathy Ammothumkandy
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States
| | - Alisha Cayce
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States
| | - Mohammad Shariq
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States
| | - Michael A. Bonaguidi
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States
- Keck School of Medicine, Neurorestoration Center, University of Southern California, Los Angeles, CA, United States
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
- Department of Gerontology, University of Southern California, Los Angeles, CA, United States
- Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
107
|
Zou Y, Zhang J, Chen L, Xu Q, Yao S, Chen H. Targeting Neuroinflammation in Central Nervous System Diseases by Oral Delivery of Lipid Nanoparticles. Pharmaceutics 2025; 17:388. [PMID: 40143051 PMCID: PMC11944764 DOI: 10.3390/pharmaceutics17030388] [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: 02/05/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Neuroinflammation within the central nervous system (CNS) is a primary characteristic of CNS diseases, such as Parkinson's disease, Alzheimer's disease (AD), amyotrophic lateral sclerosis, and mental disorders. The excessive activation of immune cells results in the massive release of pro-inflammatory cytokines, which subsequently induce neuronal death and accelerate the progression of neurodegeneration. Therefore, mitigating excessive neuroinflammation has emerged as a promising strategy for the treatment of CNS diseases. Despite advancements in drug discovery and the development of novel therapeutics, the effective delivery of these agents to the CNS remains a serious challenge due to the restrictive nature of the blood-brain barrier (BBB). This underscores the need to develop a novel drug delivery system. Recent studies have identified oral lipid nanoparticles (LNPs) as a promising approach to efficiently deliver drugs across the BBB and treat neurological diseases. This review aims to comprehensively summarize the recent advancements in the development of LNPs designed for the controlled delivery and therapeutic modulation of CNS diseases through oral administration. Furthermore, this review addresses the mechanisms by which these LNPs overcome biological barriers and evaluate their clinical implications and therapeutic efficacy in the context of oral drug delivery systems. Specifically, it focuses on LNP formulations that facilitate oral administration, exploring their potential to enhance bioavailability, improve targeting precision, and alleviate or manage the symptoms associated with a range of CNS diseases.
Collapse
Affiliation(s)
- Yuan Zou
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.Z.); (S.Y.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jing Zhang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan 430074, China; (J.Z.); (Q.X.)
| | - Longmin Chen
- Department of Rheumatology and Immunology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China;
| | - Qianqian Xu
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan 430074, China; (J.Z.); (Q.X.)
| | - Sheng Yao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.Z.); (S.Y.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.Z.); (S.Y.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
108
|
Li TL, Blair JD, Yoo T, Grant GA, Hockemeyer D, Porter BE, Bateup HS. mTORC1 activation drives astrocyte reactivity in cortical tubers and brain organoid models of TSC. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640914. [PMID: 40093155 PMCID: PMC11908165 DOI: 10.1101/2025.02.28.640914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Tuberous Sclerosis Complex (TSC) is a genetic neurodevelopmental disorder associated with early onset epilepsy, intellectual disability and neuropsychiatric disorders. A hallmark of the disorder is cortical tubers, which are focal malformations of brain development containing dysplastic cells with hyperactive mTORC1 signaling. One barrier to developing therapeutic approaches and understanding the origins of tuber cells is the lack of a model system that recapitulates this pathology. To address this, we established a genetically mosaic cortical organoid system that models a somatic "second-hit" mutation, which is thought to drive the formation of tubers in TSC. With this model, we find that loss of TSC2 cell-autonomously promotes the differentiation of astrocytes, which exhibit features of a disease-associated reactive state. TSC2 -/- astrocytes have pronounced changes in morphology and upregulation of proteins that are risk factors for neurodegenerative diseases, such as clusterin and APOE. Using multiplexed immunofluorescence in primary tubers from TSC patients, we show that tuber cells with hyperactive mTORC1 activity also express reactive astrocyte proteins, and we identify a unique population of cells with expression profiles that match those observed in organoids. Together, this work reveals that reactive astrogliosis is a primary feature of TSC that arises early in cortical development. Dysfunctional glia are therefore poised to be drivers of pathophysiology, nominating a potential therapeutic target for treating TSC and related mTORopathies.
Collapse
Affiliation(s)
- Thomas L. Li
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA, USA
| | - John D. Blair
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Taesun Yoo
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA, USA
| | - Gerald A. Grant
- Department of Neurosurgery, Lucile Packard Children’s Hospital and Stanford University Medical Center, Stanford, CA, USA
| | - Dirk Hockemeyer
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Brenda E. Porter
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Helen S. Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
109
|
Thau-Habermann N, Gschwendtberger T, Bodemer C, Petri S. Parthenolide regulates microglial and astrocyte function in primary cultures from ALS mice and has neuroprotective effects on primary motor neurons. PLoS One 2025; 20:e0319866. [PMID: 40100917 PMCID: PMC11918366 DOI: 10.1371/journal.pone.0319866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Over the last twenty years, the role of microgliosis and astrocytosis in the pathophysiology of neurodegenerative diseases has increasingly been recognized. Dysregulation of microglial and astrocyte properties and function has been described also in the fatal degenerative motor neuron disease amyotrophic lateral sclerosis (ALS). Microglia cells, the immune cells of the nervous system, can either have an immunonegative neurotoxic or immunopositive neuroprotective phenotype. The feverfew plant (Tanacetum parthenium) derived compound parthenolide has been found to be capable of interfering with microglial phenotype and properties. Positive treatment effects were shown in animal models of neurodegenerative diseases like Alzheimer's disease and Parkinson's disease. Now we were able to show that PTL has a modulating effect on primary mouse microglia cells, both wild type and SOD1, causing them to adopt a more neuroprotective potential. Furthermore, we were able to show that PTL, through its positive effect on microglia, also has an indirect positive impact on motor neurons, although PTL itself has no direct effect on these primary motor neurons. The results of our study give reason to consider PTL as a drug candidate for ALS.
Collapse
Affiliation(s)
| | | | - Colin Bodemer
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
110
|
Cooper ML, Gildea HK, Selles MC, Katafygiotou E, Liddelow SA, Chao MV. Astrocytes in the mouse brain respond bilaterally to unilateral retinal neurodegeneration. Proc Natl Acad Sci U S A 2025; 122:e2418249122. [PMID: 40063795 PMCID: PMC11929491 DOI: 10.1073/pnas.2418249122] [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: 09/06/2024] [Accepted: 01/16/2025] [Indexed: 03/25/2025] Open
Abstract
Glaucomatous optic neuropathy, or glaucoma, is the world's primary cause of irreversible blindness. Glaucoma is comorbid with other neurodegenerative diseases, but how it might impact the environment of the full central nervous system to increase neurodegenerative vulnerability is unknown. Two neurodegenerative events occur early in the optic nerve, the structural link between the retina and brain: loss of anterograde transport in retinal ganglion cell (RGC) axons and early alterations in astrocyte structure and function. Here, we used whole-mount tissue clearing of full mouse brains to image RGC anterograde transport function and astrocyte responses across retinorecipient regions early in a unilateral microbead occlusion model of glaucoma. Using light sheet imaging, we found that RGC projections terminating specifically in the accessory optic tract are the first to lose transport function. Although degeneration was induced in one retina, astrocytes in both brain hemispheres responded to transport loss in a retinotopic pattern that mirrored the degenerating RGCs. A subpopulation of these astrocytes in contact with large descending blood vessels were immunopositive for LCN2, a marker associated with astrocyte reactivity. Together, these data suggest that even early stages of unilateral glaucoma have broad impacts on the health of astrocytes across both hemispheres of the brain, implying a glial mechanism behind neurodegenerative comorbidity in glaucoma.
Collapse
Affiliation(s)
- Melissa L. Cooper
- Institute for Translational Neuroscience, New York University Grossman School of Medicine, New York, NY10016
| | - Holly K. Gildea
- Institute for Translational Neuroscience, New York University Grossman School of Medicine, New York, NY10016
| | - Maria Clara Selles
- Institute for Translational Neuroscience, New York University Grossman School of Medicine, New York, NY10016
| | - Eleni Katafygiotou
- Institute for Translational Neuroscience, New York University Grossman School of Medicine, New York, NY10016
| | - Shane A. Liddelow
- Institute for Translational Neuroscience, New York University Grossman School of Medicine, New York, NY10016
- Department of Neuroscience, New York University Grossman School of Medicine, New York, NY10016
- Department of Ophthalmology, New York University Langone Health, New York, NY10016
| | - Moses V. Chao
- Institute for Translational Neuroscience, New York University Grossman School of Medicine, New York, NY10016
- Department of Neuroscience, New York University Grossman School of Medicine, New York, NY10016
- Department of Psychiatry, New York University Langone Health, New York, NY10016
| |
Collapse
|
111
|
Schirge PM, Perneczky R, Taoka T, Ruiz-Rizzo AL, Ersoezlue E, Forbrig R, Guersel S, Kurz C, Brendel M, Hellmann-Regen J, Priller J, Schneider A, Jessen F, Düzel E, Buerger K, Teipel S, Laske C, Peters O, Spruth E, Fliessbach K, Rostamzadeh A, Glanz W, Janowitz D, Kilimann I, Sodenkamp S, Ewers M, Rauchmann BS. Perivascular space and white matter hyperintensities in Alzheimer's disease: associations with disease progression and cognitive function. Alzheimers Res Ther 2025; 17:62. [PMID: 40098158 PMCID: PMC11917016 DOI: 10.1186/s13195-025-01707-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/26/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is the leading cause of dementia, characterized by the accumulation of amyloid-beta (Aβ) and neurofibrillary tangles. Recent studies emphasize the role of vascular factors, including the glymphatic system, in AD pathogenesis, particularly in Aβ clearance. The diffusion tensor image analysis along the perivascular space (DTI-ALPS; ALPS-Index) has emerged as a novel, non-invasive method to evaluate the glymphatic system in vivo, showing glymphatic insufficiency in AD. This study aimed to investigate alterations in the function of the glymphatic system in individuals with AD versus healthy controls (HC), and to explore its association with Aβ, cerebrovascular disease (CVD), white matter hyperintensities (WMH), and cognitive function. METHODS DTI MRI data from three independent study cohorts (ActiGliA: AD n = 16, Controls n = 18; DELCODE: AD n = 54, Controls n = 67; ADNI: AD n = 43, Controls n = 49) were used to evaluate the perivascular space (PVS) integrity; a potential biomarker for glymphatic activity. The DTI-Along the Perivascular Space technique was used to measure water diffusion along PVS providing an index to assess the efficiency of the glymphatic system's waste clearance function. WMH load was quantified in FLAIR MRI using the lesion segmentation tool. We quantified WMHs volume within our defined region of interest (ROI) and excluded participants with any WMHs to avoid confounding the ALPS-Index. Associations with cerebrospinal fluid (CSF) AD hallmark biomarkers, cognitive performance (MMSE) and clinical severity (CDR) were assessed. RESULTS AD patients had a significantly lower ALPS-Index vs. healthy controls (ActiGliA: AD: mean = 1.22, SD = 0.12; Controls: mean = 1.36, SD = 0.14, p = 0.004; DELCODE: AD: mean = 1.26, SD = 0.18; Controls: mean = 1.34, SD = 0.2, p = 0.035; ADNI: AD: mean = 1.08, SD = 0.24; Controls: mean = 1.19, SD = 0.13, p = 0.008). The ALPS-Index was associated with CSF Aβ concentration, WMH number and MMSE and CDR. WMH, found in the ROIs correlated negatively with the ALPS-Index. CONCLUSIONS This study highlights the potential of the DTI-ALPS-Index as a biomarker for glymphatic dysfunction in AD. It underscores the importance of considering vascular factors and the glymphatic system in the pathogenesis and diagnosis of AD as WMHs in the ROI could cause disturbances and inaccurate indices.
Collapse
Affiliation(s)
- Philine Marie Schirge
- Department of Psychiatry and Psychotherapy, LMU Hospital, LMU Munich, Munich, Germany
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, LMU Hospital, LMU Munich, Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Ageing Epidemiology (AGE) Research Unit, School of Public HealthImperial College London, London, UK
| | - Toshiaki Taoka
- Department of Innovative Biomedical Visualization (iBMV), Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | | | - Ersin Ersoezlue
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Neurosciences, Charité Universitätsmedizin Berlin, Berlin, Germany
- ECRC Experimental and Clinical Research Center, Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Forbrig
- Institute of Neuroradiology, LMU Hospital, LMU Munich, Munich, Germany
| | - Selim Guersel
- Department of Psychiatry and Psychotherapy, LMU Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Carolin Kurz
- Department of Psychiatry and Psychotherapy, LMU Hospital, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Nuclear Medicine, Ludwig Maximilian University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Julian Hellmann-Regen
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Neurosciences, Charité Universitätsmedizin Berlin, Berlin, Germany
- ECRC Experimental and Clinical Research Center, Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117, Berlin, Germany
- Department of Psychiatry, School of Medicine, Technical University of Munich, Munich, Germany
- University of Edinburgh, UK DRI, Edinburgh, UK
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE)Venusberg-Campus, Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, Bonn, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE)Venusberg-Campus, Bonn, Germany
- Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Institute for Stroke and Dementia Research (ISD), Ludwig Maximilian University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, Rostock, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Psychiatry and Psychotherapy, Section for Dementia Research, Hertie Institute for Clinical Brain Research and University of Tübingen, Tübingen, Germany
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität BerlinHumboldt-Universität zu Berlin-Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Eike Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117, Berlin, Germany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE)Venusberg-Campus, Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, Bonn, Germany
| | - Ayda Rostamzadeh
- Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924, Cologne, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), Ludwig Maximilian University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, Rostock, Germany
| | - Sebastian Sodenkamp
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Michael Ewers
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Institute for Stroke and Dementia Research (ISD), Ludwig Maximilian University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Boris-Stephan Rauchmann
- Institute of Neuroradiology, LMU Hospital, LMU Munich, Munich, Germany.
- Department of Psychiatry and Psychotherapy, LMU Hospital, LMU Munich, Munich, Germany.
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK.
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
- Institute of Neuroradiology, University Hospital, LMU Munich, Marchioninistraße 15, 81377, München, Germany.
| |
Collapse
|
112
|
Gollihue JL, Aung KZ, Rogers CB, Sompol P, Katsumata Y, Weekman EM, Wilcock DM, Morganti JM, Norris CM. Inhibition of astrocyte signaling leads to sex-specific changes in microglia phenotypes in a diet-based model of small cerebral vessel disease. RESEARCH SQUARE 2025:rs.3.rs-6198453. [PMID: 40166012 PMCID: PMC11957200 DOI: 10.21203/rs.3.rs-6198453/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Hyperhomocysteinemia (HHcy)-inducing diets recapitulate small cerebral vessel disease phenotypes in mice including cerebrovascular pathology/dysfunction, neuroinflammation, synaptic deficits, and cognitive decline. We recently showed that astrocyte signaling through calcineurin(CN)/nuclear factor of activated T cells (NFATs) plays a causative role in these phenotypes. Here, we assessed the impact of astrocytic signaling on microglia, which set inflammatory tone in brain. Seven-to-eight-week-old male and female C57BL/6J mice received intrahippocampal injections of AAV2/5-Gfa2-EGFP (control) or adeno-associated virus (AAV) expressing the NFAT inhibitor VIVIT (i.e., AAV2/5-Gfa2-VIVIT-EGFP). Mice were then fed with control chow (CT) or B-vitamin-deficient chow for 12 weeks to induce HHcy. Immunohistochemistry was used to assess the expression of the pan-microglial marker Iba1 and the homeostatic microglial marker P2ry12. Iba1 showed little sensitivity to diet, AAV treatment, or sex. Conversely, P2ry12 expression was reduced with HHcy diet in males, but not females. Treatment of males with AAV-Gfa2-VIVIT prevented the loss of P2ry12. We next conducted single-cell RNA sequencing (scRNAseq) to determine if microglial genes and/or microglial clustering patterns were sensitive to astrocyte signaling in a sex-dependent manner. In males, disease-associated microglial genes and subclusters were overrepresented in HHcy-treated mice, while VIVIT promoted the appearance of homeostatic microglial genes and clusters. In contrast, microglial genes in females were less sensitive to diet and AAV treatments, though disease-like patterns in gene expression were also observed in the HHcy condition. However, very few of the HHcy-sensitive microglial genes in females were affected by VIVIT. The results suggest a sexually dimorphic influence of astrocyte signaling on microglial phenotypes in the context of HHcy and small cerebral vessel disease.
Collapse
|
113
|
Tang W, Wang A, Liu S, Wen G, Qi H, Gu Y, Xu C, Ren S, Zhang S, He Y. Calycosin regulates astrocyte reactivity and astrogliosis after spinal cord injury by targeting STAT3 phosphorylation. J Neuroimmunol 2025; 400:578535. [PMID: 39954615 DOI: 10.1016/j.jneuroim.2025.578535] [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: 09/12/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Astrocytes are the most populous glial cells in the central nervous system (CNS), which can exert detrimental effects through a process of reactive astrogliosis. Our previous study has indicated the potential effect of Calycosin in preventing spinal cord injury (SCI). This study aims to investigate the mechanism by which calycosin regulates the polarization of A1 astrocytes, a neurotoxic subtype of reactive astrocytes, in SCI models. MATERIALS AND METHODS The SCI model was induced by applying mechanical compression to the spinal cord using vascular clamps. A1 astrocyte differentiation was induced by treating astrocytes with microglia supernatant obtained after Lipopolysaccharide (LPS) stimulation. Key protein expression levels were analyzed by Western blotting, and astrocyte markers such as CS56, GFAP, C3, S100A10 were assessed through immunofluorescence staining. RESULTS Calycosin treatment significantly reduced glial scar formation and C3 expression in SCI rats. However, S100A10 expression remained unchanged. Further analysis showed that Calycosin inhibited A1 astrocyte activation, migration, and invasion, which was associated with STAT3 phosphorylation. Calycosin downregulated p-STAT3 levels in both A1 astrocytes and SCI rats. These effects were reversed by Colivelin (a STAT3 activator) in A1 astrocytes. CONCLUSION Calycosin treatment can modulate p-STAT3 expression, thereby altering the functionality of astrocytes during the recovery phase and positively impacting the treatment and rehabilitation of SCI.
Collapse
Affiliation(s)
- Wenhai Tang
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Aitao Wang
- Department of Anesthesiology, Hohhot First Hospital, Hohhot 010030, China
| | - Shengxing Liu
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Guangyu Wen
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Hao Qi
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Yuntao Gu
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Chunzhao Xu
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Shanwu Ren
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Shunli Zhang
- Department of Spine Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China.
| | - Yongxiong He
- Department of Orthopedics, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China.
| |
Collapse
|
114
|
Spetz MR, Kim H, Chavarria D, Conger DJ, Shattuck-Brandt R, Shekharan SR, Shostak A, Ligocki AP, Brien HJ, Embalabala RJ, Mobley BC, Schrag MS, Lippmann ES, Brunger JM. Amyloid-β-regulated gene circuits for programmable Alzheimer's disease therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642808. [PMID: 40161792 PMCID: PMC11952467 DOI: 10.1101/2025.03.12.642808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized in part by the accumulation of the protein amyloid-β (Aβ). Monoclonal antibodies (mAbs) that target Aβ for clearance from the brain have received FDA approval; however, these therapies are accompanied by serious side effects, and their cognitive benefit for patients remains of tremendous debate. Here, we present a potential engineered cell therapy for AD in which we enlist cells of the central nervous system as programmable agents for sculpting the neurodegenerative niche toward one that mitigates glial reactivity and neuronal loss. We constructed a suite of Aβ-sensitive synthetic Notch (synNotch) receptors from clinically tested anti-Aβ mAbs and show that cells expressing these receptors can recognize synthetic Aβ42 and Aβ40 with differential sensitivity. We express these receptors in astrocytes, cells native to the brain that are known to become dysfunctional in AD. These synNotch astrocytes, which upregulate selected transgenes upon exposure to synthetic and human brain-derived amyloid, were engineered to express potential therapeutic transgenes in response to Aβ, including brain-derived neurotrophic factor and antagonists of the cytokines tumor necrosis factor and interleukin-1. SynNotch astrocytes that express such antagonists in response to Aβ partially attenuate a cytokine-induced reactive astrocyte phenotype and promote barrier properties in brain microvascular endothelial cells. Additionally, engineered Aβ-synNotch cells potently upregulate transgene expression in response to Aβ deposited in the 5xFAD mouse brain, demonstrating the capacity to recognize Aβ in situ. Overall, our work supports Aβ-synNotch receptors as promising tools to generate a cell-based therapy for AD with targeted functionalities to positively influence the AD niche.
Collapse
Affiliation(s)
- Madeline R Spetz
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Hyosung Kim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN
| | - Daniel Chavarria
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN
| | - Dylan J Conger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | | | - Swathi R Shekharan
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Alena Shostak
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
| | - Alexander P Ligocki
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN
| | - Hannah J Brien
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Rebecca J Embalabala
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
| | - Bret C Mobley
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN
| | - Matthew S Schrag
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN
| | - Ethan S Lippmann
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| |
Collapse
|
115
|
Ishii T, Wang T, Shibata K, Nishitani S, Yamanashi T, Wahba NE, Seki T, Thompson KJ, Yamanishi K, Nishiguchi T, Shimura A, Aoyama B, Gorantla N, Phuong NJ, Nguyen HD, Santiago TA, Nishizawa Y, Nagao T, Howard MA, Kawasaki H, Hino K, Ikeda A, Snyder MP, Shinozaki G. Glial Contribution to the Pathogenesis of Post-Operative Delirium Revealed by Multi-omic Analysis of Brain Tissue from Neurosurgery Patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.643155. [PMID: 40161597 PMCID: PMC11952519 DOI: 10.1101/2025.03.13.643155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Post-operative delirium (POD) is a common complication after surgery especially in elderly patients, characterized by acute disturbances in consciousness and cognition, which negatively impacts long-term outcomes. Effective treatments remain elusive due to the unclear pathophysiology of POD. To address the knowledge gap, we investigated DNA methylation profiles and gene expression changes in brain cells from POD and non-POD patients who underwent brain resection surgery for medication refractory epilepsy. DNA methylation analysis revealed alteration in epigenetic status of immune and inflammation-related genes. Single-nucleus RNA sequencing (snRNAseq) identified POD-specific glial cell alterations, particularly in microglia, where neuroinflammation was strongly enhanced, consistent with epigenetic findings. Astrocytes exhibited changes in synapse-related functions and migration. Furthermore, downstream analysis indicated similarities between POD-associated glial cell states and pathologies such as encephalitis and dementia. Overall, this study-the first multi-omics analysis of brain tissue from POD patients-provides direct evidence of glial cell contributions to POD pathogenesis, and highlights potential therapeutic targets.
Collapse
Affiliation(s)
- Takaya Ishii
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Osaka, Osaka, Japan
- Current affiliation is RACTHERA Co., Ltd., Kobe, Hyogo, Japan
| | - Tao Wang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kazuki Shibata
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Drug Research Division, Sumitomo Pharma Co., Ltd., Osaka, Osaka, Japan
| | - Shota Nishitani
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
| | - Takehiko Yamanashi
- Faculty of Medicine, Department of Neuropsychiatry, Tottori University, Yonago, Tottori, Japan
| | - Nadia E. Wahba
- Department of Psychiatry, Oregon Health and Science University, School of Medicine, Portland, Oregon, USA
| | - Tomoteru Seki
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Department of Psychiatry, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | | | - Kyosuke Yamanishi
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Department of Neuropsychiatry, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Tsuyoshi Nishiguchi
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Faculty of Medicine, Department of Neuropsychiatry, Tottori University, Yonago, Tottori, Japan
| | - Akiyoshi Shimura
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Department of Psychiatry, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Bun Aoyama
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kochi, Kochi, Japan
| | - Nipun Gorantla
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
| | - Nathan J. Phuong
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
| | - Hieu D. Nguyen
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
| | - Therese A. Santiago
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
| | - Yoshitaka Nishizawa
- Department of Psychiatry, Osaka Medical and Pharmaceutical University School of Medicine, Osaka, Japan
| | - Takaaki Nagao
- Department of Neurosurgery (Sakura), Toho University School of Medicine Faculty of Medicine, Sakura, Chiba, Japan
| | - Mathew A Howard
- Department of Neurosurgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Hiroto Kawasaki
- Department of Neurosurgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Kyosuke Hino
- Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Osaka, Osaka, Japan
- Current affiliation is RACTHERA Co., Ltd., Kobe, Hyogo, Japan
| | - Atsushi Ikeda
- Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Osaka, Osaka, Japan
- Current affiliation is RACTHERA Co., Ltd., Kobe, Hyogo, Japan
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Gen Shinozaki
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
116
|
Zheng J, Wang H, Wu W, Wang L, Qin M, Zhu L, Liu Z, Chen Y, Yu Y. Role of FPR2 antagonism in alleviating social isolation-induced depression and protecting blood-brain barrier integrity. J Neuroinflammation 2025; 22:79. [PMID: 40083006 PMCID: PMC11907847 DOI: 10.1186/s12974-025-03408-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
Social isolation (SI) is a prevalent issue in modern society, particularly exacerbated during the COVID-19 pandemic, and it is a significant contributor to depressive disorders. Inflammation-related markers are upregulated in patients with major depressive disorder (MDD) unresponsive to first-line selective serotonin reuptake inhibitor (SSRI) antidepressants. This study investigates the role of formyl peptide receptor 2 (FPR2), a G-protein coupled receptor expressed in central and peripheral immune cells, in SI-induced depression. We developed a mouse model of SI by housing mice individually for three weeks. SI mice exhibited increased capillary-associated microglia (CAMs) with upregulated FPR2 expression in the prefrontal cortex (PFC) and hippocampus compared to group-housed controls. Notably, subcutaneous administration of the FPR2 antagonist WRW4 alleviated depressive and anxiety-like behaviors in SI mice, reducing microglial activation and neuronal damage. WRW4 treatment decreased CAM numbers and their FPR2 expression. RNA sequencing revealed that SI primarily induced changes in genes associated with blood-brain barrier (BBB) function, followed by alterations in genes related to hormone activity, immune activation, and neuronal function. Transcriptomic changes in brain endothelial cells from SI mice resembled those observed in animal models of several neurological disorders and in MDD patients. WRW4 treatment partially reversed these transcriptomic alterations and restored compromised BBB integrity. Additionally, intracerebroventricular (ICV) injection of WRW4 also alleviated depressive and anxiety-like behaviors in SI mice. Finally, our analysis of public transcriptome databases indicates FPR2 upregulation in the orbital ventral PFC of MDD patients and peripheral blood mononuclear cells of those in severe depressive episodes. These findings suggest that the pharmacological targeting of FPR2 may rescue SI-induced pathology in mice by protecting BBB integrity.
Collapse
Affiliation(s)
- Jiayi Zheng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hanqi Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wanning Wu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Linlin Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Meizhen Qin
- School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, China
| | - Lingfeng Zhu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhen Liu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yijun Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yang Yu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
117
|
Baier MP, Ranjit R, Owen DB, Wilson JL, Stiles MA, Masingale AM, Thomas Z, Bredegaard A, Sherry DM, Logan S. Cellular Senescence Is a Central Driver of Cognitive Disparities in Aging. Aging Cell 2025:e70041. [PMID: 40077862 DOI: 10.1111/acel.70041] [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: 10/15/2024] [Revised: 02/02/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025] Open
Abstract
Cognitive function in aging is heterogeneous: while some older individuals develop significant impairments and dementia, others remain resilient and retain cognitive function throughout their lifespan. The molecular mechanisms that underlie these divergent cognitive trajectories, however, remain largely unresolved. Here, we utilized a high-resolution home-cage-based cognitive testing paradigm to delineate mechanisms that contribute to age-related cognitive heterogeneity. We cognitively stratified aged C57Bl/6N male mice by cognitive performance into intact (resilient) or impaired subgroups based on young performance benchmarks. Cognitively impaired males exhibited marked reactive gliosis in the hippocampus, characterized by microglial activation, increased astrocyte arborization, and elevated transcriptional expression of reactivity markers. These changes were accompanied by increased markers of cellular senescence and the associated senescence-associated secretory phenotype (SASP) in impaired animals, including p16INK4a, SASP factors (e.g., Il-6, Il-1b, Mmp3), and SA-β-gal staining in the hippocampus. Notably, clearance of senescent cells using senolytic agents dasatinib and quercetin ameliorated the heterogeneity in cognitive performance observed with age and attenuated impairment-associated gliosis, senescence markers, and mitochondrial dysfunction. Aged female mice could not be stratified into subgroups yet showed increased neuroinflammation with age that was not resolved with senolytics. Collectively, our findings implicate cellular senescence as a central driver of sex-specific neuroinflammation that drives divergent cognitive trajectories in aging. Thus, we demonstrate that senolytic treatment is an effective therapeutic strategy to mitigate cognitive impairment by reducing neuroinflammation and associated metabolic disturbances.
Collapse
Affiliation(s)
- Matthew P Baier
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
| | - Rojina Ranjit
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
| | - Daniel B Owen
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
| | - Jenna L Wilson
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
| | - Megan A Stiles
- Department of Cell Biology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
| | - Anthony M Masingale
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
| | - Zachary Thomas
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
| | - Anne Bredegaard
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
| | - David M Sherry
- Department of Cell Biology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
- Neuroscience Program, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
| | - Sreemathi Logan
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
| |
Collapse
|
118
|
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] [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
|
119
|
Hołdrowicz A, Żebrowska A. Molecular Link Between Psoriasis and Depression-Update on Pathophysiology. Int J Mol Sci 2025; 26:2467. [PMID: 40141110 PMCID: PMC11942400 DOI: 10.3390/ijms26062467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Psoriasis disease is a chronic, systemic condition. Various epidemiological studies have indicated a connection between psoriasis and psychiatric diseases. It is obvious that easily visible psoriatic skin lesions cause stigmatization of patients and impact noticeably their life quality, increasing the risk of anxiety and depressive disorders. More and more attention is recently being paid to the common pathogenesis of psoriasis and depression. The underlying cause of psoriasis is chronic inflammation, and depression is also increasingly recognized as a result of neuroinflammation. Therefore, the complexity of the processes underlying both disease entities implies the need to observe psoriatic patients in terms of possible comorbidities, such as mental disorders, regardless of the severity of skin lesions and social stigmatization. This study aims to present an update on the common pathophysiology of both diseases.
Collapse
Affiliation(s)
| | - Agnieszka Żebrowska
- Department of Dermatology and Venereology, Medical University of Lodz, 90-647 Lodz, Poland;
| |
Collapse
|
120
|
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] [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
|
121
|
An J, Liu Z, Wang Y, Meng K, Wang Y, Sun H, Li M, Tang Z. Drug delivery strategy of hemostatic drugs for intracerebral hemorrhage. J Control Release 2025; 379:202-220. [PMID: 39793654 DOI: 10.1016/j.jconrel.2025.01.007] [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: 11/06/2024] [Revised: 12/26/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Intracerebral hemorrhage (ICH) is associated with high rates of mortality and disability, underscoring an urgent need for effective therapeutic interventions. The clinical prognosis of ICH remains limited, primarily due to the absence of targeted, precise therapeutic options. Advances in novel drug delivery platforms, including nanotechnology, gel-based systems, and exosome-mediated therapies, have shown potential in enhancing ICH management. This review delves into the pathophysiological mechanisms of ICH and provides a thorough analysis of existing treatment strategies, with an emphasis on innovative drug delivery approaches designed to address critical pathological pathways. We assess the benefits and limitations of these therapies, offering insights into future directions in ICH research and highlighting the transformative potential of next-generation drug delivery systems in improving patient outcomes.
Collapse
Affiliation(s)
- Junyan An
- China-Japan Union Hospital of Jilin University, Department of Neurosurgery, Changchun, Jilin Province 130033, China; Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Zhilin Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yihan Wang
- China-Japan Union Hospital of Jilin University, Department of Neurosurgery, Changchun, Jilin Province 130033, China
| | - Ke Meng
- China-Japan Union Hospital of Jilin University, Department of Neurosurgery, Changchun, Jilin Province 130033, China
| | - Yixuan Wang
- China-Japan Union Hospital of Jilin University, Department of Neurosurgery, Changchun, Jilin Province 130033, China
| | - Hai Sun
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| | - Miao Li
- China-Japan Union Hospital of Jilin University, Department of Neurosurgery, Changchun, Jilin Province 130033, China.
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| |
Collapse
|
122
|
Rowland HA, Miller G, Liu Q, Li S, Sharp NR, Ng B, Wei T, Arunasalam K, Koychev I, Hedegaard A, Ribe EM, Chan D, Chessell T, Kocagoncu E, Lawson J, Malhotra P, Ridha BH, Rowe JB, Thomas AJ, Zamboni G, Zetterberg H, Cader MZ, Wade-Martins R, Lovestone S, Nevado-Holgado A, Kormilitzin A, Buckley NJ. Changes in iPSC-astrocyte morphology reflect Alzheimer's disease patient clinical markers. Stem Cells 2025; 43:sxae085. [PMID: 39704342 PMCID: PMC11907432 DOI: 10.1093/stmcls/sxae085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/03/2024] [Indexed: 12/21/2024]
Abstract
Human induced pluripotent stem cells (iPSCs) provide powerful cellular models of Alzheimer's disease (AD) and offer many advantages over non-human models, including the potential to reflect variation in individual-specific pathophysiology and clinical symptoms. Previous studies have demonstrated that iPSC-neurons from individuals with Alzheimer's disease (AD) reflect clinical markers, including β-amyloid (Aβ) levels and synaptic vulnerability. However, despite neuronal loss being a key hallmark of AD pathology, many risk genes are predominantly expressed in glia, highlighting them as potential therapeutic targets. In this work iPSC-derived astrocytes were generated from a cohort of individuals with high versus low levels of the inflammatory marker YKL-40, in their cerebrospinal fluid (CSF). iPSC-derived astrocytes were treated with exogenous Aβ oligomers and high content imaging demonstrated a correlation between astrocytes that underwent the greatest morphology change from patients with low levels of CSF-YKL-40 and more protective APOE genotypes. This finding was subsequently verified using similarity learning as an unbiased approach. This study shows that iPSC-derived astrocytes from AD patients reflect key aspects of the pathophysiological phenotype of those same patients, thereby offering a novel means of modelling AD, stratifying AD patients and conducting therapeutic screens.
Collapse
Affiliation(s)
- Helen A Rowland
- Department of Psychiatry, University of Oxford, Headington, Oxford OX3 7JX, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Georgina Miller
- Department of Psychiatry, University of Oxford, Headington, Oxford OX3 7JX, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Qiang Liu
- Department of Psychiatry, University of Oxford, Headington, Oxford OX3 7JX, United Kingdom
- Oxford Precision Psychiatry Lab, NIHR Oxford Health Biomedical Research Centre, Oxford OX3 7JX, United Kingdom
- School of Engineering Mathematics and Technology, University of Bristol, Bristol BS8 1TW, United Kingdom
| | - Shuhan Li
- Department of Psychiatry, University of Oxford, Headington, Oxford OX3 7JX, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Nicola R Sharp
- Department of Psychiatry, University of Oxford, Headington, Oxford OX3 7JX, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Bryan Ng
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, United Kingdom
- Department of Physiology Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QU, United Kingdom
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore 117609, Republic of Singapore
| | - Tina Wei
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, United Kingdom
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Kanisa Arunasalam
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Ivan Koychev
- Department of Psychiatry, University of Oxford, Headington, Oxford OX3 7JX, United Kingdom
| | - Anne Hedegaard
- Department of Psychiatry, University of Oxford, Headington, Oxford OX3 7JX, United Kingdom
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Elena M Ribe
- Department of Psychiatry, University of Oxford, Headington, Oxford OX3 7JX, United Kingdom
- Department of Old Age Psychiatry, Maurice Wohl Institute of Clinical Neurosciences, King’s College London, London SE5 8AB, United Kingdom
| | - Dennis Chan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
- Institute of Cognitive Neuroscience, University College, LondonWC1N 3AR, United Kingdom
| | - Tharani Chessell
- Neuroscience, BioPharmaceuticals R&D, AstraZeneca, Granta Park, Cambridge CB21 6GH, United Kingdom
| | - Ece Kocagoncu
- Medical Research Council Cognition and Brain Sciences Unit, Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge CB2 7EF, United Kingdom
| | - Jennifer Lawson
- Department of Psychiatry, University of Oxford, Headington, Oxford OX3 7JX, United Kingdom
| | - Paresh Malhotra
- Department of Brain Sciences, Imperial College London, London W6 8RP, United Kingdom
| | - Basil H Ridha
- Dementia Research Centre, UCL Institute of Neurology, London WC1N 3BG, United Kingdom
| | - James B Rowe
- Medical Research Council Cognition and Brain Sciences Unit, Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge CB2 7EF, United Kingdom
| | - Alan J Thomas
- Translational and Clinical Research Institute, Newcastle University, Newcastle, United Kingdom
| | - Giovanna Zamboni
- Nuffield Department of Clinical Neurosciences, Headington, University of Oxford, Oxford OX3 9DS, United Kingdom
- Department of Biomedical, Metabolic, and Neural Science, University of Modena and Reggio Emilia, Ospedale Civile Baggiovara, 41126 Modena, Italy
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80 Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London WC1N 6BG, United Kingdom
- UK Dementia Research Institute at UCL, London WC1N 6BG, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison WI 53792, United States
| | - M Zameel Cader
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, United Kingdom
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Richard Wade-Martins
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, United Kingdom
- Department of Physiology Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QU, United Kingdom
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Headington, Oxford OX3 7JX, United Kingdom
- Currently at Janssen Medical UK, 50-100 Holmers Farm Way, High Wycombe HP12 4EG, United Kingdom
| | - Alejo Nevado-Holgado
- Department of Psychiatry, University of Oxford, Headington, Oxford OX3 7JX, United Kingdom
| | - Andrey Kormilitzin
- Department of Psychiatry, University of Oxford, Headington, Oxford OX3 7JX, United Kingdom
| | - Noel J Buckley
- Department of Psychiatry, University of Oxford, Headington, Oxford OX3 7JX, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, United Kingdom
| |
Collapse
|
123
|
Zhu M, Peng Q, Li S, Zhang G, Zhang Z. Irisin promotes autophagy and attenuates NLRP3 inflammasome activation in Parkinson's disease. Int Immunopharmacol 2025; 149:114201. [PMID: 39914281 DOI: 10.1016/j.intimp.2025.114201] [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: 12/18/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/22/2025]
Abstract
Parkinson's disease (PD) is characterized by the aggregation and prion-like propagation of α-synuclein (α-syn). Irisin is an exercise-induced myokine that regulates energy metabolism and exerts protective effects in PD by reducing α-syn pathology. However, the molecular mechanisms underlying the role of irisin are not fully understood. Here, we show that irisin inhibits NLRP3 inflammasome activation and promotes autophagy in cultured cells. Additionally, irisin alleviates oxidative stress and reduces cell apoptosis induced by α-syn fibrils. In a PD mouse model induced by intrastriatal injection of α-syn fibrils, irisin mitigated α-syn aggregation, neuroinflammation and neurodegeneration. These observations suggest that irisin functions as a protective mediator against α-syn pathology in PD and that irisin may serve as a potential therapeutic target for the prevention and treatment of PD.
Collapse
Affiliation(s)
- Min Zhu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qinyu Peng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Sheng Li
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Guoxin Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430000, China.
| |
Collapse
|
124
|
Pfeffer LK, Fischbach F, Heesen C, Friese MA. Current state and perspectives of CAR T cell therapy in central nervous system diseases. Brain 2025; 148:723-736. [PMID: 39530593 DOI: 10.1093/brain/awae362] [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: 07/19/2024] [Revised: 10/03/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
B cell-directed CAR T cell therapy has fundamentally changed the treatment of haematological malignancies, and its scope of application is rapidly expanding to include other diseases such as solid tumours or autoimmune disorders. Therapy-refractoriness remains an important challenge in various inflammatory and non-inflammatory disorders of the CNS. The reasons for therapy failure are diverse and include the limited access current therapies have to the CNS, as well as enormous inter- and intra-individual disease heterogeneity. The tissue-penetrating properties of CAR T cells make them a promising option for overcoming this problem and tackling pathologies directly within the CNS. First application of B cell-directed CAR T cells in neuromyelitis optica spectrum disorder and multiple sclerosis patients has recently revealed promising outcomes, expanding the potential of CAR T cell therapy to encompass CNS diseases. Additionally, the optimization of CAR T cells for the therapy of gliomas is a growing field. As a further prospect, preclinical data reveal the potential benefits of CAR T cell therapy in the treatment of primary neurodegenerative diseases such as Alzheimer's disease. Considering the biotechnological optimizations in the field of T cell engineering, such as extension to target different antigens or variation of the modified T cell subtype, new and promising fields of CAR T cell application are rapidly opening up. These innovations offer the potential to address the complex pathophysiological properties of CNS diseases. To use CAR T cell therapy optimally to treat CNS diseases in the future while minimizing therapy risks, further mechanistic research and prospective controlled trials are needed to assess seriously the disease and patient-specific risk-benefit ratio.
Collapse
Affiliation(s)
- Lena Kristina Pfeffer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Felix Fischbach
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christoph Heesen
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
125
|
Takalo M, Jeskanen H, Rolova T, Kervinen I, Hellén M, Heikkinen S, Koivisto H, Jokivarsi K, Müller SA, Koivumäki EM, Mäkinen P, Juopperi SP, Willman RM, Sinisalo R, Hoffmann D, Jäntti H, Peitz M, Fließbach K, Kuulasmaa T, Natunen T, Kemppainen S, Poutiainen P, Leinonen V, Malm T, Martiskainen H, Ramirez A, Haapasalo A, Lichtenthaler SF, Tanila H, Haass C, Rinne J, Koistinaho J, Hiltunen M. The protective PLCγ2-P522R variant mitigates Alzheimer's disease-associated pathologies by enhancing beneficial microglial functions. J Neuroinflammation 2025; 22:64. [PMID: 40038760 DOI: 10.1186/s12974-025-03387-6] [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: 11/21/2024] [Accepted: 02/19/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Phospholipase C gamma 2, proline 522 to arginine (PLCγ2-P522R) is a protective variant that reduces the risk of Alzheimer's disease (AD). Recently, it was shown to mitigate β-amyloid pathology in a 5XFAD mouse model of AD. Here, we investigated the protective functions of the PLCγ2-P522R variant in a less aggressive APP/PS1 mouse model of AD and assessed the underlying cellular mechanisms using mouse and human microglial models. METHODS The effects of the protective PLCγ2-P522R variant on microglial activation, AD-associated β-amyloid and neuronal pathologies, and behavioral changes were investigated in PLCγ2-P522R knock-in variant mice crossbred with APP/PS1 mice. Transcriptomic, proteomic, and functional studies were carried out using microglia isolated from mice carrying the PLCγ2-P522R variant. Finally, microglia-like cell models generated from human blood and skin biopsy samples of PLCγ2-P522R variant carriers were employed. RESULTS The PLCγ2-P522R variant decreased β-amyloid plaque count and coverage in female APP/PS1 mice. Moreover, the PLCγ2-P522R variant promoted anxiety in these mice. The area of the microglia around β-amyloid plaques was also increased in mice carrying the PLCγ2-P522R variant, while β-amyloid plaque-associated neuronal dystrophy and the levels of certain cytokines, including IL-6 and IL-1β, were reduced. These alterations were revealed through [18F]FEPPA PET imaging and behavioral studies, as well as various cytokine immunoassays, transcriptomic and proteomic analyses, and immunohistochemical analyses using mouse brain tissues. In cultured mouse primary microglia, the PLCγ2-P522R variant reduced the size of lipid droplets. Furthermore, transcriptomic and proteomic analyses revealed that the PLCγ2-P522R variant regulated key targets and pathways involved in lipid metabolism, mitochondrial fatty acid oxidation, and inflammatory/interferon signaling in acutely isolated adult mouse microglia and human monocyte-derived microglia-like cells. Finally, the PLCγ2-P522R variant also increased mitochondrial respiration in human iPSC-derived microglia. CONCLUSIONS These findings suggest that the PLCγ2-P522R variant exerts protective effects against β-amyloid and neuronal pathologies by increasing microglial responsiveness to β-amyloid plaques in APP/PS1 mice. The changes observed in lipid/fatty acid and mitochondrial metabolism revealed by the omics and metabolic assessments of mouse and human microglial models suggest that the protective effects of the PLCγ2-P522R variant are potentially associated with increased metabolic capacity of microglia.
Collapse
Affiliation(s)
- Mari Takalo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland.
| | - Heli Jeskanen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Taisia Rolova
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Inka Kervinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Marianna Hellén
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Sami Heikkinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Hennariikka Koivisto
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kimmo Jokivarsi
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE Munich), Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Esa-Mikko Koivumäki
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Petra Mäkinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | | | | | - Rosa Sinisalo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Dorit Hoffmann
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Henna Jäntti
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Michael Peitz
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
- Cell Programming Core Facility, University of Bonn Medical Faculty, Bonn, Germany
| | - Klaus Fließbach
- Department of Old Age Psychiatry and Cognitive Disorders, University of Bonn Medical Center, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE Bonn), Bonn, Germany
| | - Teemu Kuulasmaa
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Teemu Natunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Susanna Kemppainen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Pekka Poutiainen
- Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland
| | - Ville Leinonen
- Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine - Neurosurgery, University of Eastern Finland, Kuopio, Finland
| | - Tarja Malm
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Henna Martiskainen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Alfredo Ramirez
- German Center for Neurodegenerative Diseases (DZNE Bonn), Bonn, Germany
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Disease (CECAD), University of Cologne, Cologne, Germany
| | - Annakaisa Haapasalo
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE Munich), Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Heikki Tanila
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE Munich), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Juha Rinne
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Jari Koistinaho
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
126
|
Li E, Benitez C, Boggess SC, Koontz M, Rose IVL, Martinez D, Dräger N, Teter OM, Samelson AJ, Pierce N, Ullian EM, Kampmann M. CRISPRi-based screens in iAssembloids to elucidate neuron-glia interactions. Neuron 2025; 113:701-718.e8. [PMID: 39814010 PMCID: PMC11886924 DOI: 10.1016/j.neuron.2024.12.016] [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: 04/27/2023] [Revised: 11/25/2024] [Accepted: 12/17/2024] [Indexed: 01/18/2025]
Abstract
The complexity of the human brain makes it challenging to understand the molecular mechanisms underlying brain function. Genome-wide association studies have uncovered variants associated with neurological phenotypes. Single-cell transcriptomics have provided descriptions of changes brain cells undergo during disease. However, these approaches do not establish molecular mechanism. To facilitate the scalable interrogation of causal molecular mechanisms in brain cell types, we developed a 3D co-culture system of induced pluripotent stem cell (iPSC)-derived neurons and glia, termed iAssembloids. Using iAssembloids, we ask how glial and neuronal cells interact to control neuronal death and survival. Our CRISPRi-based screens identified that GSK3β inhibits the protective NRF2-mediated oxidative stress response elicited by high neuronal activity. We then investigate the role of APOE-ε4, a risk variant for Alzheimer's disease, on neuronal survival. We find that APOE-ε4-expressing astrocytes may promote neuronal hyperactivity as compared with APOE-ε3-expressing astrocytes. This platform allows for the unbiased identification of mechanisms of neuron-glia cell interactions.
Collapse
Affiliation(s)
- Emmy Li
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Camila Benitez
- TETRAD Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Steven C Boggess
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Mark Koontz
- Department of Ophthalmology, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Indigo V L Rose
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Delsy Martinez
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Nina Dräger
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Olivia M Teter
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA; UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, San Francisco, San Francisco, CA, USA
| | - Avi J Samelson
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Na'im Pierce
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA; FirstGen Internship, Emerson Collective, Palo Alto, CA, USA; University of California, Berkeley, Berkeley, CA, USA
| | - Erik M Ullian
- Department of Ophthalmology, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
127
|
de Rezende VL, de Aguiar da Costa M, Martins CD, Mathias K, Gonçalves CL, Barichello T, Petronilho F. Systemic Rejuvenating Interventions: Perspectives on Neuroinflammation and Blood-Brain Barrier Integrity. Neurochem Res 2025; 50:112. [PMID: 40035979 DOI: 10.1007/s11064-025-04361-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/06/2025]
Abstract
The aging process results in structural, functional, and immunological changes in the brain, which contribute to cognitive decline and increase vulnerability to neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and stroke-related complications. Aging leads to cognitive changes and also affect executive functions. Additionally, it causes neurogenic and neurochemical alterations, such as a decline in dopamine and acetylcholine levels, which also impact cognitive performance. The chronic inflammation caused by aging contributes to the impairment of the blood-brain barrier (BBB), contributing to the infiltration of immune cells and exacerbating neuronal damage. Therefore, rejuvenating therapies such as heterochronic parabiosis, cerebrospinal fluid (CSF) administration, plasma, platelet-rich plasma (PRP), and stem cell therapy have shown potential to reverse these changes, offering new perspectives in the treatment of age-related neurological diseases. This review focuses on highlighting the effects of rejuvenating interventions on neuroinflammation and the BBB.
Collapse
Affiliation(s)
- Victória Linden de Rezende
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Maiara de Aguiar da Costa
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Carla Damasio Martins
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Khiany Mathias
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
- Laboratory of Immunoparasitology, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Cinara Ludvig Gonçalves
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Tatiana Barichello
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
- Faillace Department of Psychiatry and Behavioral Sciences, Translational Psychiatry Program, Mcgovern Medical School, The University of Texas Health Science Center at Houston (Uthealth), Houston, TX, USA
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil.
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, 1105, Criciúma, SC, 88806-000, Brazil.
| |
Collapse
|
128
|
Liu M, Ma LY, Li QY, Huang LY, Hu HY, Tan L, Hu H. Associations of varicose veins with cerebrospinal fluid biomarkers of Alzheimer's disease pathologies in adults without dementia: the CABLE study. Front Aging Neurosci 2025; 17:1502154. [PMID: 40103929 PMCID: PMC11913850 DOI: 10.3389/fnagi.2025.1502154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/14/2025] [Indexed: 03/20/2025] Open
Abstract
Background Previous studies have found a correlation between varicose veins (VVs) and cognitive decline, and individuals with VVs have a higher prevalence of Alzheimer's disease (AD). However, the associations between VVs and the core pathologies of AD have not yet been investigated. The research was designed to analyze the relationships between VVs and cerebrospinal fluid (CSF) biomarkers of AD pathologies. Methods We included 1,298 participants from the Chinese Alzheimer's Biomarker and LifestylE (CABLE) database without dementia. Multiple linear regression (MLR) model was applied to assess the relationships between the VVs and CSF AD biomarkers. Then, we conducted subgroup analyses according to age, gender, education levels and apolipoprotein E genotype ε4 (APOE-ε4) carrier status. Additionally, mediation effects were assessed using causal mediation analyses with 10,000 bootstrapped iterations. Results In total subjects, VVs had negative correlations with CSF Aβ42 (β = -0.157, p = 0.038) and CSF Aβ42/Aβ40 ratio (β = -0.272, p < 0.001), as well as positive correlations with CSF Aβ40 (β = 0.170, p = 0.024), CSF p-tau (β = 0.192, p = 0.008), CSF t-tau/Aβ42 ratio (β = 0.190, p = 0.011), and CSF p-tau/Aβ42 ratio (β = 0.248, p = 0.001), after adjusting for age, sex, education levels and APOE-ε4 carrier status. Subgroup analyses demonstrated that the relations between VVs and CSF AD biomarkers were more significant in female, mid-life adults (40-65 years), less-educated individuals and APOE-ε4 non-carriers. Moreover, CSF Aβ42/Aβ40 ratio might be a partial mediator of the association between VVs and p-tau pathology. Conclusion Our study found correlations between VVs and CSF AD biomarkers, suggesting that VVs may be a potential risk factor for the development of AD.
Collapse
Affiliation(s)
- Min Liu
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| | - Li-Yun Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Qiong-Yao Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Liang-Yu Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - He-Ying Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
129
|
Kornberg MD, Calabresi PA. Multiple Sclerosis and Other Acquired Demyelinating Diseases of the Central Nervous System. Cold Spring Harb Perspect Biol 2025; 17:a041374. [PMID: 38806240 PMCID: PMC11875095 DOI: 10.1101/cshperspect.a041374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Acquired demyelinating diseases of the central nervous system (CNS) comprise inflammatory conditions, including multiple sclerosis (MS) and related diseases, as well as noninflammatory conditions caused by toxic, metabolic, infectious, traumatic, and neurodegenerative insults. Here, we review the spectrum of diseases producing acquired CNS demyelination before focusing on the prototypical example of MS, exploring the pathologic mechanisms leading to myelin injury in relapsing and progressive MS and summarizing the mechanisms and modulators of remyelination. We highlight the complex interplay between the immune system, oligodendrocytes and oligodendrocyte progenitor cells (OPCs), and other CNS glia cells such as microglia and astrocytes in the pathogenesis and clinical course of MS. Finally, we review emerging therapeutic strategies that exploit our growing understanding of disease mechanisms to limit progression and promote remyelination.
Collapse
Affiliation(s)
- Michael D Kornberg
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21287, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21205, USA
| |
Collapse
|
130
|
Soelter TM, Howton TC, Wilk EJ, Whitlock JH, Clark AD, Birnbaum A, Patterson DC, Cortes CJ, Lasseigne BN. Evaluation of altered cell-cell communication between glia and neurons in the hippocampus of 3xTg-AD mice at two time points. J Cell Commun Signal 2025; 19:e70006. [PMID: 40026671 PMCID: PMC11870853 DOI: 10.1002/ccs3.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 03/05/2025] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is characterized by progressive memory loss and cognitive decline, affecting behavior, speech, and motor abilities. The neuropathology of AD includes the formation of extracellular amyloid-β plaques and intracellular neurofibrillary tangles of phosphorylated tau, along with neuronal loss. Although neuronal loss is an AD hallmark, cell-cell communication between neuronal and non-neuronal cell populations maintains neuronal health and brain homeostasis. To study changes in cell-cell communication during disease progression, we performed snRNA-sequencing of the hippocampus from female 3xTg-AD and wild-type littermates at 6 and 12 months. We inferred differential cell-cell communication between 3xTg-AD and wild-type mice across time points and between senders (astrocytes, microglia, oligodendrocytes, and OPCs) and receivers (excitatory and inhibitory neurons) of interest. We also assessed the downstream effects of altered glia-neuron communication using pseudobulk differential gene expression, functional enrichment, and gene regulatory analyses. We found that glia-neuron communication is increasingly dysregulated in 12-month 3xTg-AD mice. We also identified 23 AD-associated ligand-receptor pairs that are upregulated in the 12-month-old 3xTg-AD hippocampus. Our results suggest increased AD association of interactions originating from microglia. Signaling mediators were not significantly differentially expressed but showed altered gene regulation and transcription factor activity. Our findings indicate that altered glia-neuron communication is increasingly dysregulated and affects the gene regulatory mechanisms in neurons of 12-month-old 3xTg-AD mice.
Collapse
Affiliation(s)
- Tabea M. Soelter
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Timothy C. Howton
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Elizabeth J. Wilk
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Jordan H. Whitlock
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Amanda D. Clark
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Allison Birnbaum
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
- Department of Molecular, Cell and Developmental BiologyUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Dalton C. Patterson
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Constanza J. Cortes
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Brittany N. Lasseigne
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
131
|
Wang J, Zhang B, Li L, Tang X, Zeng J, Song Y, Xu C, Zhao K, Liu G, Lu Y, Li X, Shu K. Repetitive traumatic brain injury-induced complement C1-related inflammation impairs long-term hippocampal neurogenesis. Neural Regen Res 2025; 20:821-835. [PMID: 38886955 PMCID: PMC11433904 DOI: 10.4103/nrr.nrr-d-23-01446] [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: 08/28/2023] [Revised: 11/09/2023] [Accepted: 12/21/2023] [Indexed: 06/20/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202503000-00027/figure1/v/2024-06-17T092413Z/r/image-tiff Repetitive traumatic brain injury impacts adult neurogenesis in the hippocampal dentate gyrus, leading to long-term cognitive impairment. However, the mechanism underlying this neurogenesis impairment remains unknown. In this study, we established a male mouse model of repetitive traumatic brain injury and performed long-term evaluation of neurogenesis of the hippocampal dentate gyrus after repetitive traumatic brain injury. Our results showed that repetitive traumatic brain injury inhibited neural stem cell proliferation and development, delayed neuronal maturation, and reduced the complexity of neuronal dendrites and spines. Mice with repetitive traumatic brain injuryalso showed deficits in spatial memory retrieval. Moreover, following repetitive traumatic brain injury, neuroinflammation was enhanced in the neurogenesis microenvironment where C1q levels were increased, C1q binding protein levels were decreased, and canonical Wnt/β-catenin signaling was downregulated. An inhibitor of C1 reversed the long-term impairment of neurogenesis induced by repetitive traumatic brain injury and improved neurological function. These findings suggest that repetitive traumatic brain injury-induced C1-related inflammation impairs long-term neurogenesis in the dentate gyrus and contributes to spatial memory retrieval dysfunction.
Collapse
Affiliation(s)
- Jing Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Department of Neurosurgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Bing Zhang
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Lanfang Li
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiaomei Tang
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jinyu Zeng
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yige Song
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chao Xu
- Department of Graduate Student, Chongqing Medical University, Chongqing, China
| | - Kai Zhao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Guoqiang Liu
- Department of Basic Medicine, School of Medical Science, Hubei University for Nationalities, Enshi, Hubei Province, China
| | - Youming Lu
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xinyan Li
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Department of Anatomy, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
132
|
Qu Y, Yi L, Tang Y, Yang F, Pan BF, Shi S, Qu C, Li F, Wen S, Pan Y. TSG-6 Protects Against Cerebral Ischemia-Reperfusion Injury via Upregulating Hsp70-1B in Astrocytes. CNS Neurosci Ther 2025; 31:e70354. [PMID: 40130432 PMCID: PMC11933850 DOI: 10.1111/cns.70354] [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: 09/03/2024] [Revised: 02/21/2025] [Accepted: 03/15/2025] [Indexed: 03/26/2025] Open
Abstract
AIMS This study aimed to investigate the relationship between tumor necrosis factor alpha-induced protein (TNFAIP6/TSG-6) and astrocytes in cerebral ischemia/reperfusion (I/R) injury. METHODS Utilizing in vivo and in vitro cerebral I/R models, cerebral infarct volumes, neurobehavioral outcomes, blood-brain barrier (BBB) permeability, as well as indicators of astrocyte apoptosis, reactivity, and A1 phenotype were assessed to evaluate the effects of recombinant rattus TSG-6 (rrTSG-6) on astrocytes in acute cerebral I/R injury. Following mRNA sequencing of all astrocyte groups, astrocyte apoptosis and reactivity were analyzed through a combined intervention of rrTSG-6 and Apoptozole, a heat shock protein 70-1B (Hsp70-1B) inhibitor, in vitro. RESULTS The findings demonstrated that rrTSG-6 significantly reduced cerebral infarct volumes by nearly half, improved neurobehavioral outcomes, mitigated BBB damage, and suppressed the expressions of astrocyte apoptosis markers, reactivity indicators, and A1 phenotype markers. mRNA sequencing revealed that the Hsp70-1B protein level increased to approximately 1.6 times that of the rrTSG-6 non-intervention group. Furthermore, Apoptozole impeded the expressions of astrocyte apoptosis markers, reactivity indicators, and A1 phenotype markers. CONCLUSION TSG-6 inhibited nuclear factor kappa-B (NF-κB) phosphorylation by upregulating Hsp70-1B in oxygen-glucose deprivation/reoxygenation (OGD/R)-induced astrocytes, thereby exerting a protective effect through anti-apoptotic mechanisms and the suppression of astrocyte reactivity and A1 transformation following cerebral I/R injury.
Collapse
Affiliation(s)
- Yewei Qu
- Department of NeurologyFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
- NHC Key Laboratory of Cell TransplantationFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Lian Yi
- Department of NeurologyFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Yushi Tang
- Department of NeurologyFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Fan Yang
- Department of NeurologyFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Byron Fei Pan
- Department of NeurologyFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Shanshan Shi
- Department of NeurologyFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Changda Qu
- Department of NeurologyFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Fangqin Li
- Department of NeurologyFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Shirong Wen
- Department of NeurologyFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Yujun Pan
- Department of NeurologyFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| |
Collapse
|
133
|
Heffernan ÁB, Steinruecke M, Dempsey G, Chandran S, Selvaraj BT, Jiwaji Z, Stavrou M. Role of glia in delirium: proposed mechanisms and translational implications. Mol Psychiatry 2025; 30:1138-1147. [PMID: 39463449 PMCID: PMC11835730 DOI: 10.1038/s41380-024-02801-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 08/23/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024]
Abstract
Delirium is a common acute onset neurological syndrome characterised by transient fluctuations in cognition. It affects over 20% of medical inpatients and 50% of those critically ill. Delirium is associated with morbidity and mortality, causes distress to patients and carers, and has significant socioeconomic costs in ageing populations. Despite its clinical significance, the pathophysiology of delirium is understudied, and many underlying cellular mechanisms remain unknown. There are currently no effective pharmacological treatments which directly target underlying disease processes. Although many studies focus on neuronal dysfunction in delirium, glial cells, primarily astrocytes, microglia, and oligodendrocytes, and their associated systems, are increasingly implicated in delirium pathophysiology. In this review, we discuss current evidence which implicates glial cells in delirium, including biomarker studies, post-mortem tissue analyses and pre-clinical models. In particular, we focus on how astrocyte pathology, including aberrant brain energy metabolism and glymphatic dysfunction, reactive microglia, blood-brain barrier impairment, and white matter changes may contribute to the pathogenesis of delirium. We also outline limitations in this body of work and the unique challenges faced in identifying causative mechanisms in delirium. Finally, we discuss how established neuroimaging and single-cell techniques may provide further mechanistic insight at pre-clinical and clinical levels.
Collapse
Affiliation(s)
- Áine Bríd Heffernan
- UK Dementia Research Institute at The University of Edinburgh, The University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | | | - Georgia Dempsey
- School of Medicine, University of St Andrews, St Andrews, UK
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
| | - Siddharthan Chandran
- UK Dementia Research Institute at The University of Edinburgh, The University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, The University of Edinburgh, Edinburgh, UK
- Anne Rowling Regenerative Neurology Clinic, The University of Edinburgh, Edinburgh, UK
| | - Bhuvaneish T Selvaraj
- UK Dementia Research Institute at The University of Edinburgh, The University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, The University of Edinburgh, Edinburgh, UK
- Anne Rowling Regenerative Neurology Clinic, The University of Edinburgh, Edinburgh, UK
| | - Zoeb Jiwaji
- UK Dementia Research Institute at The University of Edinburgh, The University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Maria Stavrou
- UK Dementia Research Institute at The University of Edinburgh, The University of Edinburgh, Edinburgh, UK.
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK.
- Euan MacDonald Centre for Motor Neuron Disease Research, The University of Edinburgh, Edinburgh, UK.
- Anne Rowling Regenerative Neurology Clinic, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
134
|
Zhang L, Xu Z, Jia Z, Cai S, Wu Q, Liu X, Hu X, Bai T, Chen Y, Li T, Liu Z, Wu B, Zhu J, Zhou H. Modulating mTOR-dependent astrocyte substate transitions to alleviate neurodegeneration. NATURE AGING 2025; 5:468-485. [PMID: 39779911 DOI: 10.1038/s43587-024-00792-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/06/2024] [Indexed: 01/11/2025]
Abstract
Traditional approaches to studying astrocyte heterogeneity have mostly focused on analyzing static properties, failing to identify whether subtypes represent intermediate or final states of reactive astrocytes. Here we show that previously proposed neuroprotective and neurotoxic astrocytes are transitional states rather than distinct subtypes, as revealed through time-series multiomic sequencing. Neuroprotective astrocytes are an intermediate state of the transition from a nonreactive to a neurotoxic state in response to neuroinflammation, a process regulated by the mTOR signaling pathway. In Alzheimer's disease (AD) and aging, we observed an imbalance in neurotoxic and neuroprotective astrocytes in animal models and human patients. Moreover, targeting mTOR in astrocytes with rapamycin or shRNA mitigated astrocyte neurotoxic effects in neurodegenerative mouse models. Overall, our study uncovers a mechanism through which astrocytes exhibit neuroprotective functions before becoming neurotoxic under neuroinflammatory conditions and highlights mTOR modulation specifically in astrocytes as a potential therapeutic strategy for neurodegenerative diseases.
Collapse
Affiliation(s)
- Liansheng Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| | - Zhengzheng Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zhiheng Jia
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shicheng Cai
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qiang Wu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xingyu Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xinde Hu
- Genemagic Biosciences Co., Ltd., Shanghai, China
| | - Tao Bai
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yongyu Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tianwen Li
- Fudan University Huashan Hospital, Department of Neurosurgery, National Center for Neurological Disorders, National Key Library for Medical Neurobiology, Shanghai Key Library of Brain Function and Regeneration, Institutes of Brain Science, MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhen Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Bin Wu
- Department of Neurology & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianhong Zhu
- Fudan University Huashan Hospital, Department of Neurosurgery, National Center for Neurological Disorders, National Key Library for Medical Neurobiology, Shanghai Key Library of Brain Function and Regeneration, Institutes of Brain Science, MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Haibo Zhou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China.
| |
Collapse
|
135
|
Choi HN, Kim SH, Jo MG, Lee B, Kim YJ, Lee SE, Lee JH, Seong HM, Kim SJ, Park SW, Kim HJ, Kang H, Lee CH, Lee MY, Yun SP, Kim M. A2-Astrocyte Activation by Short-Term Hypoxia Rescues α-Synuclein Pre-Formed-Fibril-Induced Neuronal Cell Death. Biomedicines 2025; 13:604. [PMID: 40149582 PMCID: PMC11940376 DOI: 10.3390/biomedicines13030604] [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: 11/06/2024] [Revised: 02/15/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Parkinson's disease (PD) is a neuro-degenerative disease for which a radical cure is not available, only symptomatic control. Studies have shown that hypoxia may have disease-modifying effects on PD. Methods: Herein, we investigated whether short-term hypoxia activates astrocytes and whether it has a protective effect on pre-formed fibril (PFF)-treated primary cortical neurons. Results: Long-term hypoxia suppresses astrocyte activation and induces cell death, whereas short-term hypoxia activates astrocytes without affecting cellular apoptosis or viability. Short-term hypoxia restored the cellular apoptosis and viability of PFF-treated neurons and reduced toxic phospho-α-synuclein (p-α-syn) aggregation. Similarly, the short-term hypoxia-exposed astrocyte-conditioned medium rescued cellular apoptosis and the viability of PFF-treated neurons and p-α-syn expression. Quantitative polymerase chain reaction revealed that short-term hypoxia promotes protective A2 astrocytes and suppresses toxic A1 astrocytes. Conclusions: Our findings suggest that short-term hypoxia has a neuro-protective effect against PD by activating protective A2 astrocytes, which rescue PFF-induced neuronal cell death. This provides insights into the clinical implications of short-term hypoxia as a disease-modifying PD strategy.
Collapse
Affiliation(s)
- Ha Nyeoung Choi
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (H.N.C.); (S.-H.K.); (B.L.); (Y.J.K.); (S.E.L.); (J.H.L.); (S.W.P.); (H.J.K.)
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Seon-Hee Kim
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (H.N.C.); (S.-H.K.); (B.L.); (Y.J.K.); (S.E.L.); (J.H.L.); (S.W.P.); (H.J.K.)
| | - Min Gi Jo
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Bina Lee
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (H.N.C.); (S.-H.K.); (B.L.); (Y.J.K.); (S.E.L.); (J.H.L.); (S.W.P.); (H.J.K.)
| | - Young Jin Kim
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (H.N.C.); (S.-H.K.); (B.L.); (Y.J.K.); (S.E.L.); (J.H.L.); (S.W.P.); (H.J.K.)
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - So Eun Lee
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (H.N.C.); (S.-H.K.); (B.L.); (Y.J.K.); (S.E.L.); (J.H.L.); (S.W.P.); (H.J.K.)
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Jeong Hyun Lee
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (H.N.C.); (S.-H.K.); (B.L.); (Y.J.K.); (S.E.L.); (J.H.L.); (S.W.P.); (H.J.K.)
| | - Hye Min Seong
- Department of Ophthalmology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (H.M.S.); (S.J.K.)
| | - Seong Jae Kim
- Department of Ophthalmology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (H.M.S.); (S.J.K.)
| | - Sang Won Park
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (H.N.C.); (S.-H.K.); (B.L.); (Y.J.K.); (S.E.L.); (J.H.L.); (S.W.P.); (H.J.K.)
| | - Hye Jung Kim
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (H.N.C.); (S.-H.K.); (B.L.); (Y.J.K.); (S.E.L.); (J.H.L.); (S.W.P.); (H.J.K.)
| | - Heeyoung Kang
- Department of Neurology, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea; (H.K.); (C.H.L.)
- Department of Neurology, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Chan Hyun Lee
- Department of Neurology, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea; (H.K.); (C.H.L.)
| | - Min Young Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Seung Pil Yun
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (H.N.C.); (S.-H.K.); (B.L.); (Y.J.K.); (S.E.L.); (J.H.L.); (S.W.P.); (H.J.K.)
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Minkyeong Kim
- Department of Neurology, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea; (H.K.); (C.H.L.)
| |
Collapse
|
136
|
Ji J, Wang Y, Chen B, Xu X, Lv M. Is there an association between cognitive impairment and urinary adrenaline, norepinephrine, gamma-aminobutyric acid, and taurine levels in children with obstructive sleep apnea?: A case control study. BMC Pediatr 2025; 25:156. [PMID: 40025451 PMCID: PMC11871638 DOI: 10.1186/s12887-025-05500-3] [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: 11/11/2024] [Accepted: 02/10/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Children with obstructive sleep apnea (OSA) can develop cognitive impairments. Urinary adrenaline (EPI), norepinephrine (NE) and gamma-aminobutyric acid (GABA) are elevated, and taurine is decreased in children with OSA. The purpose of this study was to investigate the above-mentioned neurotransmitter levels in children with and without OSA, and explore their association with OSA-related cognitive impairments. METHODS Children underwent overnight polysomnography (PSG) for habitual snoring or mouth breathing in the pediatric sleep center from February 2023 to February 2024, as well as a group of healthy controls were enrolled in this study. Pediatric Quality of Life Inventory (PedsQL) and Child Behavior Checklist (CBCL) were used to evaluate the cognitive function of these children. Morning urine samples were collected to measure the urinary neurotransmitter levels. RESULTS This study recruited 74 children with OSA, 30 children with primary snoring (PS) and 16 healthy controls. In the comparison of PedsQL scores, social function (85(75, 100)), school function (65.88 ± 18.52), and total scores (74.15 ± 12.74) of OSA group were significantly lower than that of non-OSA group (P < 0.05); OSA group also exhibited increased withdrawn (1 (0, 2)) and attention problems (2 (1, 5)) scores in CBCL than non-OSA group (P < 0.05). A total of 39 cases in the OSA group (PedsQL total score below 77.42) were considered to have mild cognitive impairment (MCI), who had higher urinary EPI (190.68 ± 38.77 ng/ml) and lower taurine (432.20 ± 53.52 ng/ml) levels than both PS and OSA without MCI groups (P < 0.001). Logistic regression analysis showed that high levels of urinary NE (OR = 1.027, 95%CI: 1.002 ~ 1.052) and low levels of taurine (OR = 0.982, 95%CI: 0.969 ~ 0.995) are significantly associated with cognitive impairment in children with OSA, and their combination has a larger area under the curve (0.695) for prediction, with a sensitivity of 64.1% and specificity of 68.6% (P = 0.004). CONCLUSIONS Children with OSA presented impaired cognitive functions such as school, social function deficits and attention problems. Measuring urinary EPI and taurine levels may contribute to the prediction of OSA-related cognitive impairments.
Collapse
Affiliation(s)
- Jiapeng Ji
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, 215003, Jiangsu, China
| | - Yuqing Wang
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, 215003, Jiangsu, China.
| | - Bolin Chen
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, 215003, Jiangsu, China
| | - Xueyun Xu
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, 215003, Jiangsu, China
| | - Meng Lv
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, 215003, Jiangsu, China
| |
Collapse
|
137
|
Cardaci V, Di Pietro L, Zupan MC, Sibbitts J, Privitera A, Lunte SM, Caraci F, Hartley MD, Caruso G. Characterizing oxidative stress induced by Aβ oligomers and the protective role of carnosine in primary mixed glia cultures. Free Radic Biol Med 2025; 229:213-224. [PMID: 39824445 PMCID: PMC11895860 DOI: 10.1016/j.freeradbiomed.2025.01.030] [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: 09/07/2024] [Revised: 12/18/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline and memory loss. A critical aspect of AD pathology is represented by oxidative stress, which significantly contributes to neuronal damage and death. Microglia and astrocytes, the primary glial cells in the brain, are crucial for managing oxidative stress and supporting neuronal function. Carnosine is an endogenous dipeptide possessing a multimodal mechanism of action that includes antioxidant, anti-inflammatory, and anti-aggregant activities. The present study investigated the effects of Aβ1-42 oligomers (oAβ), small aggregates associated with the neurodegeneration observed in AD, on primary rat mixed glia cultures composed of both microglia and astrocytes, focusing on the ability of these detrimental species to induce oxidative stress. We assessed intracellular reactive oxygen species (ROS) and nitric oxide (NO) levels as markers of oxidative stress. Exposure to oAβ significantly elevated both ROS and NO intracellular levels compared to control cells. However, this effect was completely inhibited by the pre-treatment of mixed cultures with carnosine, resulting in ROS and NO levels similar to those observed in untreated (control) cells. Single-cell analysis of cellular responses to oAβ revealed heterogeneous ROS production, resulting in two distinct clusters of cells, one of which was very responsive to the treatment. The presence of carnosine counteracted the overproduction of ROS, also leading to a single, homogeneous cluster, similar to that observed in the case of control cells. Interestingly, unlike ROS response, single-cell analysis of NO production did not show any distinct clusters. Overall, our findings demonstrated the ability of carnosine to mitigate Aβ-induced oxidative stress in mixed glia cells, by rescuing ROS and NO intracellular levels, as well as to normalize the heterogeneous response to the treatment measured in terms of clusters' formation. The present study suggests a therapeutic potential of carnosine in pathologies characterized by oxidative stress including AD.
Collapse
Affiliation(s)
| | - Lucia Di Pietro
- Department of Drug and Health Sciences, University of Catania, Catania, Italy; Scuola Superiore di Catania, University of Catania, Catania, Italy
| | - Matthew C Zupan
- Department of Chemistry, University of Kansas, Lawrence, KS, USA
| | - Jay Sibbitts
- Department of Chemistry, University of Kansas, Lawrence, KS, USA; Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Anna Privitera
- Department of Drug and Health Sciences, University of Catania, Catania, Italy; Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Susan M Lunte
- Department of Chemistry, University of Kansas, Lawrence, KS, USA; Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, KS, USA; Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, Catania, Italy; Unit of Neuropharmacology and Translational Neurosciences, Oasi Research Institute-IRCCS, Troina, Italy
| | | | - Giuseppe Caruso
- Department of Drug and Health Sciences, University of Catania, Catania, Italy; Unit of Neuropharmacology and Translational Neurosciences, Oasi Research Institute-IRCCS, Troina, Italy.
| |
Collapse
|
138
|
Chen W, Mao T, Ma R, Xiong Y, Han R, Wang L. The role of astrocyte metabolic reprogramming in ischemic stroke (Review). Int J Mol Med 2025; 55:49. [PMID: 39930815 PMCID: PMC11781528 DOI: 10.3892/ijmm.2025.5490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/08/2025] [Indexed: 02/13/2025] Open
Abstract
Ischemic stroke, a leading cause of disability and mortality worldwide, is characterized by the sudden loss of blood flow in specific area of the brain. Intravenous thrombolysis with recombinant tissue plasminogen activator is the only approved pharmacological treatment for acute ischemic stroke; however, the aforementioned treatment has significant clinical limitations, thus there is an urgent need for the development of novel mechanisms and therapeutic strategies for ischemic stroke. Astrocytes, abundant and versatile cells in the central nervous system, offer crucial support to neurons nutritionally, structurally and physically. They also contribute to blood‑brain barrier formation and regulate neuronal extracellular ion concentrations. Accumulated evidence has revealed the involvement of astrocytes in the regulation of host neurotransmitter metabolism, immune response and tissue repair, and different metabolic characteristics of astrocytes can contribute to the process and development of ischemic stroke, suggesting that targeted regulation of astrocyte metabolic reprogramming may contribute to the treatment and prognosis of ischemic stroke. In the present review, the current understanding of the multifaceted mechanisms of astrocyte metabolic reprogramming in ischemic stroke, along with its regulatory factors and pathways, as well as the strategies to promote its polarization balance, which hold promise for astrocyte immunometabolism‑targeted therapies in the treatment of ischemic stroke, were summarized.
Collapse
Affiliation(s)
- Weixin Chen
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100105, P.R. China
| | - Tangyou Mao
- Gastroenterology Department, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, P.R. China
| | - Rui Ma
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100105, P.R. China
| | - Yuxuan Xiong
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100105, P.R. China
| | - Ran Han
- Clinical Laboratory Department, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, P.R. China
| | - Le Wang
- Cerebrovascular Disease Department, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, P.R. China
| |
Collapse
|
139
|
Xu W, Guo Y, Zhao L, Fu R, Qin X, Zhang Y, Cheng X, Xu S. The Aging Immune System: A Critical Attack on Ischemic Stroke. Mol Neurobiol 2025; 62:3322-3342. [PMID: 39271626 DOI: 10.1007/s12035-024-04464-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 08/29/2024] [Indexed: 09/15/2024]
Abstract
Ischemic stroke caused by cerebrovascular embolism is an age-related disease with high rates of disability and mortality. Although the mechanisms of immune and inflammatory development after stroke have been of great interest, most studies have neglected the critical and unavoidable factor of age. As the global aging trend intensifies, the number of stroke patients is constantly increasing, emphasizing the urgency of finding effective measures to address the needs of elderly stroke patients. The concept of "immunosenescence" appears to explain the worse stroke outcomes in older individuals. Immune remodeling due to aging involves dynamic changes at all levels of the immune system, and the overall consequences of central (brain-resident) and peripheral (non-brain-resident) immune cells in stroke vary according to the age of the individual. Lastly, the review outlines recent strategies aimed at immunosenescence to improve stroke prognosis.
Collapse
Affiliation(s)
- Wenzhe Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuying Guo
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Linna Zhao
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Rong Fu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoli Qin
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunsha Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xueqi Cheng
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.
| |
Collapse
|
140
|
Xie ZF, Wang SY, Gao Y, Zhang YD, Han YN, Huang J, Gao MN, Wang CG. Vagus nerve stimulation (VNS) preventing postoperative cognitive dysfunction (POCD): two potential mechanisms in cognitive function. Mol Cell Biochem 2025; 480:1343-1357. [PMID: 39138750 DOI: 10.1007/s11010-024-05091-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Postoperative cognitive dysfunction (POCD) impacts a significant number of patients annually, frequently impairing their cognitive abilities and resulting in unfavorable clinical outcomes. Aimed at addressing cognitive impairment, vagus nerve stimulation (VNS) is a therapeutic approach, which was used in many mental disordered diseases, through the modulation of vagus nerve activity. In POCD model, the enhancement of cognition function provided by VNS was shown, demonstrating VNS effect on cognition in POCD. In the present study, we primarily concentrates on elucidating the role of the VNS improving the cognitive function in POCD, via two potential mechanisms: the inflammatory microenvironment and epigenetics. This study provided a theoretical support for the feasibility that VNS can be a potential method to enhance cognition function in POCD.
Collapse
Affiliation(s)
- Zi-Feng Xie
- Department of Anesthesiology, The First Central Hospital of Baoding, Northern Great Wall Street 320#, Baoding, 071000, Hebei, China
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
- The First Clinical Medical College, Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
| | - Sheng-Yu Wang
- Department of Anesthesiology, The First Central Hospital of Baoding, Northern Great Wall Street 320#, Baoding, 071000, Hebei, China
- Graduate College, Chengde Medical College, Chengde, 067000, Hebei, China
| | - Yuan Gao
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
- The First Clinical Medical College, Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
| | - Yi-Dan Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
- The First Clinical Medical College, Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
| | - Ya-Nan Han
- Department of Anesthesiology, The First Central Hospital of Baoding, Northern Great Wall Street 320#, Baoding, 071000, Hebei, China
- Graduate College, Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Jin Huang
- Department of Anesthesiology, The First Central Hospital of Baoding, Northern Great Wall Street 320#, Baoding, 071000, Hebei, China
- Graduate College, Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Mei-Na Gao
- Department of Anesthesiology, The First Central Hospital of Baoding, Northern Great Wall Street 320#, Baoding, 071000, Hebei, China
| | - Chun-Guang Wang
- Department of Anesthesiology, The First Central Hospital of Baoding, Northern Great Wall Street 320#, Baoding, 071000, Hebei, China.
| |
Collapse
|
141
|
Zhang Q, Wang Y, Wu D, Chen Z. Stimuli-responsive nanoscale drug delivery system for epilepsy theranostics. Acta Biomater 2025; 194:58-79. [PMID: 39880180 DOI: 10.1016/j.actbio.2025.01.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/12/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Epilepsy is a common neurological disease characterized by distinct pathological changes in the epileptogenic zone. Antiseizure drugs (ASDs) are widely used as the primary treatment for epilepsy. To improve the efficiency of ASDs medication, stimuli-responsive nanoscale drug delivery systems (nanoDDSs), triggered by either endogenous or exogenous factors, have been developed and been considered as a noninvasive and spatial-temporal approach to epilepsy theranostics. In this review, we introduce the pathological variations observed in epileptic lesions such as dysregulated neurotransmitter systems, disrupted ion homeostasis, and dynamic inflammatory cytokine networks. Furthermore, we summarize the recent advances in functional nano-assemblies that could be activated by endogenous stimuli of pathological alterations or exogenous stimuli such as electricity, light, and other interventions. Finally, we discuss the remaining challenges and prospect the insight into perspective of future development in this field. In summary, this review aims to highlight the potential of stimuli-responsive nanoDDSs as precise, controllable and efficient strategies for addressing unresolved issues in epilepsy theranostics. STATEMENT OF SIGNIFICANCE: This review summarizes recent progress in pathological changes such as dysregulated neurotransmitter system, disrupted ion homeostasis and dynamic inflammatory cytokine network, and emphasizes endogenous/exogenous stimuli-responsive nanoscale platforms including neurotransmitter-, ion-, and other stimuli-responsive nanoDDSs, providing the prospects of smart nanoDDSs applications and discussing the challenges to offer generalized guideline for further development of epilepsy theranostics.
Collapse
Affiliation(s)
- Qi Zhang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences and School of Basic Medical Sciences, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences and School of Basic Medical Sciences, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences and School of Basic Medical Sciences, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, PR China.
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences and School of Basic Medical Sciences, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, PR China.
| |
Collapse
|
142
|
Hong X, Chen T, Liu Y, Li J, Huang D, Ye K, Liao W, Wang Y, Liu M, Luan P. Design, current states, and challenges of nanomaterials in anti-neuroinflammation: A perspective on Alzheimer's disease. Ageing Res Rev 2025; 105:102669. [PMID: 39864562 DOI: 10.1016/j.arr.2025.102669] [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: 12/04/2024] [Revised: 01/08/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
Alzheimer's disease (AD), an age-related neurodegenerative disease, brings huge damage to the society, to the whole family and even to the patient himself. However, until now, the etiological factor of AD is still unknown and there is no effective treatment for it. Massive deposition of amyloid-beta peptide(Aβ) and hyperphosphorylation of Tau proteins are acknowledged pathological features of AD. Recent studies have revealed that neuroinflammation plays a pivotal role in the pathology of AD. With the rise of nanomaterials in the biomedical field, researchers are exploring how the unique properties of these materials can be leveraged to develop effective treatments for AD. This article has summarized the influence of neuroinflammation in AD, the design of nanoplatforms, and the current research status and inadequacy of nanomaterials in improving neuroinflammation in AD.
Collapse
Affiliation(s)
- Xinyang Hong
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Yunyun Liu
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Neurology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Jun Li
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Dongqing Huang
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Kaiyu Ye
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Wanchen Liao
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Yulin Wang
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Mengling Liu
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Ping Luan
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China; School of Basic Medical Sciences, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
143
|
Zhang B, Chen J, Wang J, Pan X. Arsenic exposure induces neural cells senescence and abnormal lipid droplet accumulation leading to social memory impairment in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125779. [PMID: 39894154 DOI: 10.1016/j.envpol.2025.125779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/08/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
The long-term harmful effects of arsenic exposure remain one of the important public health issues. The effects of arsenic exposure on the central nervous system, particularly concerning brain structure and function, have been garnering increasing attention. Hence, the aim of this study was to investigate the impact of chronic low-dose arsenic exposure on murine social memory and to elucidate the underlying molecular mechanisms. Male C57BL/6 mice at six months of age were randomly assigned to a control group and three treatment groups with different arsenic concentrations (50, 100, and 200 μg/L), with exposure durations of 30, 90, 180, and 360 days. The five-social memory test and three-chamber social memory test results indicated that chronic low-dose arsenic exposure disrupted social memory in mice. Further analysis revealed that arsenic exposure led to degeneration of neurons within the dorsal CA2 of the hippocampus (dCA2) and the lateral entorhinal cortex (LEC), which are pivotal for the modulation of social memory, and dCA2 neurons demonstrated structural disruptions and cytoplasmic fragmentation. In addition, arsenic exposure induced neurons and glial cells senescence in both dCA2 and LEC, with a particularly pronounced effect in microglia, and worse with dosage increasing of arsenic exposure, correlating with elevated expression levels of p16INK4A, ferritin light chain and the senescence-associated secretory factors TNF-α and IL-1β, and reduced expression of Lamin B1. Moreover, arsenic exposure triggered substantial cytoplasmic lipid droplets accumulation in neurons, astrocytes and microglia, with an upregulation of PLIN2 expression, a protein associated with lipid droplet formation in astrocytes. At the same time, the aberrant accumulation of lipid droplets further aggravated the astrocytes and microglia aging, especially microglia. Additionally, correlation analysis revealed that social memory impairment was negatively correlated with nerve cell senescence and lipid accumulation. Our findings suggest that arsenic exposure induced cellular functional abnormalities by triggering cellular senescence and the accumulation of lipid droplets, thereby exacerbated neuronal degeneration and result in impaired social memory in mice.
Collapse
Affiliation(s)
- Bo Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 561113, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed By the Province and Ministry, Guizhou Medical University, Guiyang, 561113, China.
| | - Junhong Chen
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 561113, China
| | - Jiaojiao Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 561113, China
| | - Xueli Pan
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 561113, China.
| |
Collapse
|
144
|
Monnot C, Kalomoiri M, MacNicol E, Kim E, Mesquita M, Damberg P, Van Kampen JM, Kay DG, Turkheimer F, Robertson HA, Cash D, Svenningsson P. Early alterations of functional connectivity, regional brain volumes and astrocyte markers in the beta-sitosterol beta-d-glucoside (BSSG) rat model of parkinsonism. Exp Neurol 2025; 385:115118. [PMID: 39716587 DOI: 10.1016/j.expneurol.2024.115118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/01/2024] [Accepted: 12/15/2024] [Indexed: 12/25/2024]
Abstract
The β-sitosterol-β-ᴅ-glucoside (BSSG) rat model of experimental parkinsonism develops pathological behaviour and motor changes that progress over time. The purpose of this study was to identify early changes in structure and function of the brain of rats treated with BSSG using both structural and resting-state functional MRI. BSSG and non-BSSG rats were fed five days a week for sixteen weeks, then underwent in vivo MRI scans and an assessment of motor performance 2 and 8 weeks later (18 and week 24 from BSSG). Groups of rats were killed at weeks 19 and 25, then imaged again with MR ex vivo, and immunostained for tyrosine hydroxylase (TH). Since BSSG may interfere with cholesterol metabolism in astrocytes, we also studied potential target engagement and measured levels of astrocyte markers GFAP and S100b. At both studied timepoints, functional connectivity (FC) between brain areas with intermediate connectivity was decreased, but brain volumes increased in the BSSG-treated rats. At week 18/19, fine movements were normal, whereas TH and GFAP were decreased in the striatum, but not in the substantia nigra. At week 24/25, fine movements were impaired, and TH was decreased both in the striatum and the substantia nigra and S100b was increased in the substantia nigra. Astrogliosis may contribute to the increased brain volume found after BSSG exposure. Using the BSSG model of parkinsonism, the results demonstrate early functional and structural alterations in MRI imaging that occur before the appearance of detectable motor symptoms.
Collapse
Affiliation(s)
- C Monnot
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - M Kalomoiri
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - E MacNicol
- Department of Neuroimaging, Institute of Psychology, Psychiatry and Neuroscience, King's College London, London, UK
| | - E Kim
- Department of Neuroimaging, Institute of Psychology, Psychiatry and Neuroscience, King's College London, London, UK
| | - M Mesquita
- Department of Neuroimaging, Institute of Psychology, Psychiatry and Neuroscience, King's College London, London, UK
| | - P Damberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - J M Van Kampen
- Neurodyn Life Sciences Inc., Charlottetown, Prince Edward Island, Canada
| | - D G Kay
- Neurodyn Life Sciences Inc., Charlottetown, Prince Edward Island, Canada
| | - F Turkheimer
- Department of Neuroimaging, Institute of Psychology, Psychiatry and Neuroscience, King's College London, London, UK
| | - H A Robertson
- Neurodyn Life Sciences Inc., Charlottetown, Prince Edward Island, Canada
| | - D Cash
- Department of Neuroimaging, Institute of Psychology, Psychiatry and Neuroscience, King's College London, London, UK.
| | - P Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Basic and Clinical Neuroscience, King's College London, London, UK.
| |
Collapse
|
145
|
McInnis JJ, LeComte MD, Reed LF, Torsney EE, Del Rio-Guerra R, Poynter ME, Spees JL. Microglial cell proliferation is regulated, in part, by reactive astrocyte ETB R signaling after ischemic stroke. Exp Neurol 2025; 385:115125. [PMID: 39716588 PMCID: PMC11781953 DOI: 10.1016/j.expneurol.2024.115125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/01/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
Reciprocal communication between reactive astrocytes and microglial cells provides local, coordinated control over critical processes such as neuroinflammation, neuroprotection, and scar formation after CNS injury, but is poorly understood. The vasoactive peptide hormone endothelin (ET) is released and/or secreted by endothelial cells, microglial cells and astrocytes early after ischemic stroke and other forms of brain injury. To better understand glial cell communication after stroke, we sought to identify paracrine effectors produced and secreted downstream of astroglial endothelin receptor B (ETBR) signaling. Using a genetic loss-of-function screen, we identified angiopoietin-2 (Ang-2) as a factor produced by reactive astrocytes in response to ET. In experiments with primary adult astrocytes stimulated by IRL1620, a specific ETBR agonist, we found that ERK1/2 and NFkB mediated the effects of ET on Ang-2 production. To determine astroglial Ang-2 levels in vivo, reactive astrocytes expressing the high affinity glutamate transporter (GLAST, EAAT1) were isolated by magnetic-activated cell sorting 3 days after stroke. Astrocytes obtained from the ipsilateral hemisphere expressed significantly more Ang-2 compared with astrocytes isolated from the contralateral hemisphere, or from cortices of sham-operated (control) mice. Notably, analysis of microglia sorted from CX3CR1-eGFP mice demonstrated increased cell surface expression of Tie-2, the Ang-2 receptor, on cells obtained from ipsilateral versus contralateral tissue. Addition of recombinant Ang-2 to astrocyte-conditioned medium significantly increased the number of SIM-A9 murine microglial cells cultured under hypoxic conditions (1 % oxygen for 48 h). In transgenic GFAP-CreER™-EDNRB-fl/fl mice with stroke, conditional knockout of astroglial ETBR significantly decreased the number of proliferating cells in the peri-infarct area with a microglial phenotype (Ki67+/CD11b+). Our results indicate that Ang-2, and possibly other paracrine effectors functioning downstream of astroglial ETBR signaling, are important mediators of microglial cell dynamics after stroke.
Collapse
Affiliation(s)
- John J McInnis
- Department of Medicine, Cardiovascular Research Institute, University of Vermont, Colchester, VT 05446, USA; Department of Neurological Sciences and Neuroscience Graduate Program, University of Vermont, Burlington, VT 05401, USA
| | - Matthew D LeComte
- Department of Medicine, Cardiovascular Research Institute, University of Vermont, Colchester, VT 05446, USA; Department of Neurological Sciences and Neuroscience Graduate Program, University of Vermont, Burlington, VT 05401, USA
| | - Leah F Reed
- Department of Medicine, Pulmonary Disease and Critical Care, University of Vermont, Burlington, VT 05405, USA
| | - Emily E Torsney
- Department of Neurological Sciences and Neuroscience Graduate Program, University of Vermont, Burlington, VT 05401, USA
| | - Roxana Del Rio-Guerra
- Harry Hood Bassett Flow Cytometry and Cell Sorting Facility, University of Vermont, Burlington, VT 05401, USA
| | - Matthew E Poynter
- Department of Medicine, Pulmonary Disease and Critical Care, University of Vermont, Burlington, VT 05405, USA
| | - Jeffrey L Spees
- Department of Medicine, Cardiovascular Research Institute, University of Vermont, Colchester, VT 05446, USA; Department of Neurological Sciences and Neuroscience Graduate Program, University of Vermont, Burlington, VT 05401, USA.
| |
Collapse
|
146
|
Chen L, Zhao X, Sheng R, Lazarovici P, Zheng W. Artemisinin alleviates astrocyte overactivation and neuroinflammation by modulating the IRE1/NF-κB signaling pathway in in vitro and in vivo Alzheimer's disease models. Free Radic Biol Med 2025; 229:96-110. [PMID: 39826816 DOI: 10.1016/j.freeradbiomed.2025.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/25/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Recent studies have shown that neuroinflammation and heightened glial activity, particularly astrocyte overactivation, are associated with Alzheimer's disease (AD). Abnormal accumulation of amyloid-beta (Aβ) induces endoplasmic reticulum (ER) stress and activates astrocytes. Artemisinin (ART), a frontline anti-malarial drug, has been found to have neuroprotective properties. However, its impact on astrocytes remains unclear. In this study, we used Aβ1-42 induced astrocyte cultures and 3 × Tg-AD mice as in vitro and in vivo models, respectively, to investigate the effects of ART on AD related astrocyte overactivation and its underlying mechanisms. ART attenuated Aβ1-42-induced astrocyte activation, ER stress, and inflammatory responses in astrocyte cultures by inhibiting IRE1 phosphorylation and the NF-κB pathway, as evidenced by the overexpression of IRE1 WT and IRE1-K599A (kinase activity invalidated), along with application of activators and inhibitors related to ER stress. Furthermore, ART alleviated the detrimental effects and restored neurotrophic function of astrocytes on co-cultured neurons, preventing neuronal apoptosis during Aβ1-42 treatment. In 3 × Tg-AD mice, ART treatment improved cognitive function and reduced astrocyte overactivation, neuroinflammation, ER stress, and neuronal apoptosis. Moreover, ART attenuated the upregulation of IRE1/NF-κB pathway activity in AD mice. Astrocyte-specific overexpression of IRE1 via adeno-associated virus in AD mice reversed the ameliorating effects of ART. Our findings suggest that ART inhibits astrocyte overactivation and neuroinflammation in both in vitro and in vivo AD models by modulating the IRE1/NF-κB signaling pathway, thereby enhancing neuronal functions. This study underscores the therapeutic potential of ART in AD and highlights the significance of modulating the ER stress-inflammatory cycle and normalizing astrocyte-neuron communication.
Collapse
Affiliation(s)
- Lei Chen
- Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Macau, Taipa, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, China
| | - Xia Zhao
- Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Macau, Taipa, China
| | - Rui Sheng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, China.
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 9112002, Israel
| | - Wenhua Zheng
- Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Macau, Taipa, China.
| |
Collapse
|
147
|
Xian Y, Liu J, Dai M, Zhang W, He M, Wei Z, Jiang Y, Le S, Lin Z, Tang S, Zhou Y, Dong L, Liang J, Zhang J, Wang L. Microglia Promote Lymphangiogenesis Around the Spinal Cord Through VEGF-C/VEGFR3-Dependent Autophagy and Polarization After Acute Spinal Cord Injury. Mol Neurobiol 2025; 62:2740-2755. [PMID: 39158788 DOI: 10.1007/s12035-024-04437-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/09/2024] [Indexed: 08/20/2024]
Abstract
Reducing secondary injury is a key focus in the field of spinal cord injury (SCI). Recent studies have revealed the role of lymphangiogenesis in reducing secondary damage to central nerve. However, the mechanism of lymphangiogenesis is not yet clear. Macrophages have been shown to play an important role in peripheral tissue lymphangiogenesis. Microglia is believed to play a role similar to macrophages in the central nervous system (CNS); we hypothesized that there was a close relationship between microglia and central nerve system lymphangiogenesis. Herein, we used an in vivo model of SCI to explored the relationship between microglia and spinal cord lymphangiogenesis and further investigated the polarization of microglia and its role in promoting spinal cord lymphangiogenesis by a series of in vitro experiments. The current study elucidated for the first time the relationship between microglia and lymphangiogenesis around the spinal cord after SCI. Classical activated (M1) microglia can promote lymphangiogenesis by secreting VEGF-C which further increases polarization and secretion of lymphatic growth factor by activating VEGFR3. The VEGF-C/VEGFR3 pathway activation downregulates microglia autophagy, thereby regulating the microglia phenotype. These results indicate that M1 microglia promote lymphangiogenesis after SCI, and activated VEGF-C/VEGFR3 signaling promotes M1 microglia polarization by inhibiting autophagy, thereby facilitates lymphangiogenesis.
Collapse
Affiliation(s)
- Yeyang Xian
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Jie Liu
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Mengxuan Dai
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Wensheng Zhang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Minye He
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Zhengnong Wei
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Yutao Jiang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Shiyong Le
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Zhuoang Lin
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Shuai Tang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Yunfei Zhou
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Liming Dong
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Jinzheng Liang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Jie Zhang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China.
| | - Liang Wang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China.
| |
Collapse
|
148
|
Majewski S, Klein P, Boillée S, Clarke BE, Patani R. Towards an integrated approach for understanding glia in Amyotrophic Lateral Sclerosis. Glia 2025; 73:591-607. [PMID: 39318236 PMCID: PMC11784848 DOI: 10.1002/glia.24622] [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: 04/17/2024] [Revised: 09/03/2024] [Accepted: 09/15/2024] [Indexed: 09/26/2024]
Abstract
Substantial advances in technology are permitting a high resolution understanding of the salience of glia, and have helped us to transcend decades of predominantly neuron-centric research. In particular, recent advances in 'omic' technologies have enabled unique insights into glial biology, shedding light on the cellular and molecular aspects of neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Here, we review studies using omic techniques to attempt to understand the role of glia in ALS across different model systems and post mortem tissue. We also address caveats that should be considered when interpreting such studies, and how some of these may be mitigated through either using a multi-omic approach and/or careful low throughput, high fidelity orthogonal validation with particular emphasis on functional validation. Finally, we consider emerging technologies and their potential relevance in deepening our understanding of glia in ALS.
Collapse
Affiliation(s)
- Stanislaw Majewski
- Department of Neuromuscular Diseases, Queen Square Institute of NeurologyUniversity College LondonLondonUK
- The Francis Crick InstituteLondonUK
| | - Pierre Klein
- Department of Neuromuscular Diseases, Queen Square Institute of NeurologyUniversity College LondonLondonUK
- The Francis Crick InstituteLondonUK
| | - Séverine Boillée
- Sorbonne Université, Institut du Cerveau—Paris Brain Institute—ICM, Inserm, CNRS, APHPHôpital de la Pitié‐SalpêtrièreParisFrance
| | - Benjamin E. Clarke
- Department of Neuromuscular Diseases, Queen Square Institute of NeurologyUniversity College LondonLondonUK
- The Francis Crick InstituteLondonUK
| | - Rickie Patani
- Department of Neuromuscular Diseases, Queen Square Institute of NeurologyUniversity College LondonLondonUK
- The Francis Crick InstituteLondonUK
| |
Collapse
|
149
|
İş Ö, Min Y, Wang X, Oatman SR, Abraham Daniel A, Ertekin‐Taner N. Multi Layered Omics Approaches Reveal Glia Specific Alterations in Alzheimer's Disease: A Systematic Review and Future Prospects. Glia 2025; 73:539-573. [PMID: 39652363 PMCID: PMC11784841 DOI: 10.1002/glia.24652] [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: 08/12/2024] [Revised: 11/11/2024] [Accepted: 11/16/2024] [Indexed: 02/01/2025]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative dementia with multi-layered complexity in its molecular etiology. Multiple omics-based approaches, such as genomics, epigenomics, transcriptomics, proteomics, metabolomics, and lipidomics are enabling researchers to dissect this molecular complexity, and to uncover a plethora of alterations yielding insights into the pathophysiology of this disease. These approaches reveal multi-omics alterations essentially in all cell types of the brain, including glia. In this systematic review, we screen the literature for human studies implementing any omics approach within the last 10 years, to discover AD-associated molecular perturbations in brain glial cells. The findings from over 200 AD-related studies are reviewed under four different glial cell categories: microglia, oligodendrocytes, astrocytes and brain vascular cells. Under each category, we summarize the shared and unique molecular alterations identified in glial cells through complementary omics approaches. We discuss the implications of these findings for the development, progression and ultimately treatment of this complex disease as well as directions for future omics studies in glia cells.
Collapse
Affiliation(s)
- Özkan İş
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Yuhao Min
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Xue Wang
- Department of Quantitative Health SciencesMayo ClinicJacksonvilleFloridaUSA
| | | | | | - Nilüfer Ertekin‐Taner
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Department of NeurologyMayo ClinicJacksonvilleFloridaUSA
| |
Collapse
|
150
|
Zhai S, Chen Y, Jiang T, Wu F, Cheng X, Wang Q, Wang M. Traditional Chinese medicine provides candidates for mutiple seclorsis: A review based on the progress of MS and potent treatment medicine. Mult Scler Relat Disord 2025; 95:106319. [PMID: 39951915 DOI: 10.1016/j.msard.2025.106319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/18/2024] [Accepted: 02/02/2025] [Indexed: 02/17/2025]
Abstract
Multiple Sclerosis(MS) is a chronic inflammatory and degenerative autoimmune neurological disease, characterized by immune cells infiltration, demyelination, axonal loss and neuron degeneration. At present, the precise mechanism of the disease is still not very clear. Latest studies clarified that immune imbalance, microglia polarization, oxidative stress, the destruction of blood-brain barrier(BBB) and blood-spinal cord barrier(BSCB), and intestinal flora imbalance may participate in the pathogenesis and promote the progress of the disease. Traditional Chinese medicine(TCM) and their bioeffective components were found to have capacity to regulate these mechanisms, and have the advantages of multi-target activity, low toxicity and side effects, making TCM promising therapy candidates. In this review, we summarized the progress of TCM in treating MS or its animal model in recent five years, in order to further demonstrate the mechanism of MS and provide more potential effective drug choice.
Collapse
Affiliation(s)
- Shaopeng Zhai
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Yan Chen
- Department of Rehabilitation, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, Henan, China
| | - Taotao Jiang
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Fengjuan Wu
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Xiaorong Cheng
- Department of Rehabilitation, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Qi Wang
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Manxia Wang
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China.
| |
Collapse
|