251
|
Timofeeva AV, Akhmetzyanova ER, Rizvanov AA, Mukhamedshina YO. Interaction of microglia with the microenvironment in spinal cord injury. Neuroscience 2025; 565:594-603. [PMID: 39622381 DOI: 10.1016/j.neuroscience.2024.11.074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024]
Abstract
This article discusses the peculiarities of microglia behaviour and their interaction with other cells of the central nervous system (CNS) during neural tissue injury with a focus on spinal cord injury (SCI). Taking into account the plasticity of microglia, the influence of the microenvironment should be taken into account to establish the mechanisms determining the polarization pathways of these cells. Determination of the system of microglia interactions with other CNS cells during injury will reveal the patterns of post-traumatic microglia responses, in particular, determining both pro-inflammatory and anti-inflammatory responses. This review compiles information on changes in microglia activation, migration and phagocytosis, as well as their reciprocal effects on other CNS cells, such as neurons, astrocytes and oligodendrocytes, in the background of SCI. The information contained in this article may be of interest not only to scientists studying traumatic injuries of the central nervous system, but also to specialists in the field of studying and treating neurodegenerative diseases, since the mechanisms occurring in the injured spinal cord may also be characteristic of pathological events in degenerative processes.
Collapse
Affiliation(s)
- A V Timofeeva
- Kazan (Volga Region) Federal University, Kazan, Russia
| | | | - A A Rizvanov
- Kazan (Volga Region) Federal University, Kazan, Russia; Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, Russia
| | - Y O Mukhamedshina
- Kazan (Volga Region) Federal University, Kazan, Russia; Kazan State Medical University, Kazan, Russia
| |
Collapse
|
252
|
Absalyamova M, Traktirov D, Burdinskaya V, Artemova V, Muruzheva Z, Karpenko M. Molecular basis of the development of Parkinson's disease. Neuroscience 2025; 565:292-300. [PMID: 39653246 DOI: 10.1016/j.neuroscience.2024.12.009] [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: 08/08/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
Parkinson's disease is one of the most prevalent neurodegenerative motor disorders worldwide with postural instability, bradykinesia, resting tremor and rigidity being the most common symptoms of the disease. Despite the fact that the molecular mechanisms of Parkinson's disease pathogenesis have already been well described, there is still no coherent picture of the etiopathogenesis of this disease. According to modern concepts, neurodegeneration is induced mainly by oxidative stress, neuroinflammation, dysregulation of cerebral proteostasis, apoptotic dysregulation, and impaired autophagy. This review describes how various factors contribute to neurodegeneration in Parkinson's disease. Understanding the factors affecting fundamental cellular processes and responsible for disease progression may help develop therapeutic strategies to improve the quality of life of patients suffering from the disease. The review also discusses the role of calpains in the development of Parkinson's disease. It is known that α-synuclein is a substrate of calcium-dependent proteases of the calpain family. Truncated forms of α-synuclein are not only involved in the process of formation of the aggregates, but also increase their toxicity.
Collapse
Affiliation(s)
| | | | | | | | | | - Marina Karpenko
- Peter the Great St Petersburg Polytechnic University, Russia
| |
Collapse
|
253
|
Liu Q, Tan Y, Zhang ZW, Tang W, Han L, Peng KP, Liu MH, Tian GX. The role of NLRP3 inflammasome-mediated pyroptosis in astrocytes during hyperoxia-induced brain injury. Inflamm Res 2025; 74:25. [PMID: 39862240 DOI: 10.1007/s00011-024-01984-4] [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: 08/07/2024] [Revised: 10/08/2024] [Accepted: 10/18/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Hyperoxia-induced brain injury is a severe neurological complication that is often accompanied by adverse long-term prognosis. The pathogenesis of hyperoxia-induced brain injury is highly complex, with neuroinflammation playing a crucial role. The activation of the nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome, which plays a pivotal role in regulating and amplifying the inflammatory response, is the pathological core of hyperoxia-induced brain injury. Additionally, astrocytes actively participate in neuroinflammatory responses. However, there is currently no comprehensive overview summarizing the role of astrocytes in hyperoxia-induced brain injury and the NLRP3 signaling pathways in astrocytes. OBJECTIVE This article aims to provide an overview of studies reported in the literature investigating the pathological role of astrocyte involvement during the inflammatory response in hyperoxia-induced brain injury, the mechanisms of hyperoxia activateing the NLRP3 inflammasome to mediate pyroptosis in astrocytes, and the potential therapeutic effects of drugs targeting the NLRP3 inflammasome to alleviate hyperoxia-induced brain injury. METHOD We searched major databases (including PubMed, Web of Science, and Google Scholar, etc.) for literature encompassing astrocytes, NLRP3 inflammasome, and pyroptosis during hyperoxia-induced brain injury up to Oct 2024. We combined with studies found in the reference lists of the included studies. CONCLUSION In this study, we elucidated the transition of function in astrocytes and activation mechanisms under hyperoxic conditions, and we summarized the potential upstream of the trigger involved in NLRP3 inflammasome activation during hyperoxia-induced brain injury, such as ROS and potassium efflux. Furthermore, we described the signaling pathways of the NLRP3 inflammasome and pyroptosis executed by GSDMD and GSDME in astrocytes under hyperoxic conditions. Finally, we summarized the inhibitors targeting the NLRP3 inflammasome in astrocytes to provide new insights for treating hyperoxia-induced brain injury.
Collapse
Affiliation(s)
- Qiao Liu
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Yan Tan
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Zhan-Wei Zhang
- Department of Neurosurgery, The First Hospital of Hunan University of Chinese Medicine, 40007, Changsha, China
| | - Wang Tang
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Lei Han
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Ke-Ping Peng
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Hospital of Hunan University of Chinese Medicine, 40007, Changsha, China
| | - Ming-Hui Liu
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Gui-Xiang Tian
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| |
Collapse
|
254
|
Jiang D, Ding Y, Hu S, Wei G, Trujillo C, Yang Z, Wei Z, Li W, Liu D, Li C, Gan W, Santos HA, Yin G, Fan J. Broad-spectrum downregulation of inflammatory cytokines by polydopamine nanoparticles to protect the injured spinal cord. Acta Biomater 2025; 193:559-570. [PMID: 39674244 DOI: 10.1016/j.actbio.2024.12.028] [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: 08/30/2024] [Revised: 11/14/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Acute neuroinflammation, which is notably characterized by a significant elevation in pro-inflammatory cytokines and chemokines, often rapidly develops following a traumatic spinal cord injury and exacerbates damage in the lesion area. This study addresses the limitations inherent in strategies that regulate only a single or a few cytokines, which are often insufficient to counteract the progression of secondary injuries. We explore the use of polydopamine nanoparticles as a broad-spectrum immunomodulator, capable of capturing by adsorption a wide range of cytokines and thereby effectively suppressing neuroinflammation. Leveraging their adhesive properties, these nanoparticles promptly reduce levels of various excessive cytokines, including IL-1α, IL-1β, IL-6, IL-10, IL-17A, IL-18, TNF-α, MCP-1, GRO/KC, M-CSF, MIP-3α, and IFN-γ, primarily through physical adsorption. This reduction in cytokine levels contributes to the subsequent inhibition of pro-inflammatory M1 microglia and A1 astrocyte activation, aiding in the recovery of motor functions in vivo. In summary, polydopamine nanoparticles represent a versatile and effective approach for modulating acute neuroinflammation in spinal cord injuries. By broadly down-regulating cytokines, polydopamine nanoparticles propose an innovative approach for treating spinal cord injuries. STATEMENT OF SIGNIFICANCE: The current study demonstrated the immunomodulatory potential of polydopamine nanoparticles in mitigating neuroinflammation following spinal cord injury. Both in vitro and in vivo analyses revealed significant downregulation of several key cytokines among a panel of 23 cytokines and chemokines. The potential underlying mechanisms governing these interactions were elucidated through comprehensive molecular dynamics simulations for the first time. Consequently, the downregulation of these cytokines and chemokines led to the inhibition of pro-inflammatory M1 microglia and A1 astrocyte activation in both in vitro and in vivo models. This inhibition protected neurons within the microenvironment, resulting in improved locomotor functions. Overall, this study underscores the prominent therapeutic efficacy of polydopamine nanoparticles in alleviating neuroinflammation, highlighting their potential as broad-spectrum regulators in intricate microenvironments.
Collapse
Affiliation(s)
- Dongdong Jiang
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Yaping Ding
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074 China; Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Shuai Hu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing 210009, China
| | - Guangfei Wei
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing 210009, China
| | - Claudia Trujillo
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Zhiyuan Yang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing 210009, China
| | - Zhenyang Wei
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Wei Li
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Dongfei Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing 210009, China
| | - Cong Li
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Wenwu Gan
- Department of Orthopedics, Xuyi People's Hospital Xuyi 211700, Jiangsu, China
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland; Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, AV 9713, the Netherlands.
| | - Guoyong Yin
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Jin Fan
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
255
|
Ma B, Ren J, Qian X. Study on the Polarization of Astrocytes in the Optic Nerve Head of Rats Under High Intraocular Pressure: In Vitro. Bioengineering (Basel) 2025; 12:104. [PMID: 40001624 PMCID: PMC11852053 DOI: 10.3390/bioengineering12020104] [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: 12/08/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Astrocytes, the most common glial cells in the optic nerve head (ONH), provide support and nutrition to retinal ganglion cells. This study aims to investigate the polarization types of astrocytes in the ONH of rats under high intraocular pressure (IOP) and explore signaling pathways potentially associated with different types of polarized astrocytes. The rat models with chronic high IOP were established. High IOP lasted for 2, 4, 6, and 8 weeks. Astrocytes were extracted from the ONH of rats using the tissue block cultivation method. Western blot was used to detect the expression of proteins associated with astrocyte polarization. Proteomics was employed to identify differential proteins associated with astrocyte polarization. Astrocytes polarized into A2 astrocytes after 2, 4, 6, and 8 weeks of high IOP, while polarization into A1 astrocytes began only after 8 weeks of high IOP. The differential proteins associated with A1 astrocyte polarization are primarily enriched in pathways of neurodegeneration with respect to multiple diseases, while the differential proteins associated with A2 astrocyte polarization are primarily enriched in pathways of spliceosome in amyotrophic lateral sclerosis. Our findings could provide a better understanding of the role of ONH astrocytes in the pathogenesis of glaucoma and offer new perspectives for glaucoma treatment.
Collapse
Affiliation(s)
| | | | - Xiuqing Qian
- School of Biomedical Engineering, Capital Medical University, Beijing 100069, China; (B.M.)
| |
Collapse
|
256
|
Tran KM, Kwang NE, Butler CA, Gomez-Arboledas A, Kawauchi S, Mar C, Chao D, Barahona RA, Da Cunha C, Tsourmas KI, Shi Z, Wang S, Collins S, Walker A, Shi KX, Alcantara JA, Neumann J, Duong DM, Seyfried NT, Tenner AJ, LaFerla FM, Hohsfield LA, Swarup V, MacGregor GR, Green KN. APOE Christchurch enhances a disease-associated microglial response to plaque but suppresses response to tau pathology. Mol Neurodegener 2025; 20:9. [PMID: 39844286 PMCID: PMC11752804 DOI: 10.1186/s13024-024-00793-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 12/22/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Apolipoprotein E ε4 (APOE4) is the strongest genetic risk factor for late-onset Alzheimer's disease (LOAD). A recent case report identified a rare variant in APOE, APOE3-R136S (Christchurch), proposed to confer resistance to autosomal dominant Alzheimer's Disease (AD). However, it remains unclear whether and how this variant exerts its protective effects. METHODS We introduced the R136S variant into mouse Apoe (ApoeCh) and investigated its effect on the development of AD-related pathology using the 5xFAD model of amyloidosis and the PS19 model of tauopathy. We used immunohistochemical and biochemical analysis along with single-cell spatial omics and bulk proteomics to explore the impact of the ApoeCh variant on AD pathological development and the brain's response to plaques and tau. RESULTS In 5xFAD mice, ApoeCh enhances a Disease-Associated Microglia (DAM) phenotype in microglia surrounding plaques, and reduces plaque load, dystrophic neurites, and plasma neurofilament light chain. By contrast, in PS19 mice, ApoeCh suppresses the microglial and astrocytic responses to tau-laden neurons and does not reduce tau accumulation or phosphorylation, but partially rescues tau-induced synaptic and myelin loss. We compared how microglia responses differ between the two mouse models to elucidate the distinct DAM signatures induced by ApoeCh. We identified upregulation of antigen presentation-related genes in the DAM response in a PS19 compared to a 5xFAD background, suggesting a differential response to amyloid versus tau pathology that is modulated by the presence of ApoeCh. Bulk proteomics show upregulated mitochondrial protein abundance with ApoeCh in 5xFAD mice, but reductions in mitochondrial and translation associated proteins in PS19 mice. CONCLUSIONS These findings highlight the ability of the ApoeCh variant to modulate microglial responses based on the type of pathology, enhancing DAM reactivity in amyloid models and dampening neuroinflammation to promote protection in tau models. This suggests that the Christchurch variant's protective effects likely involve multiple mechanisms, including changes in receptor binding and microglial programming.
Collapse
Affiliation(s)
- Kristine M Tran
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Nellie E Kwang
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Claire A Butler
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Angela Gomez-Arboledas
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Shimako Kawauchi
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA
| | - Cassandra Mar
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Donna Chao
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Rocio A Barahona
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Celia Da Cunha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Kate I Tsourmas
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Zechuan Shi
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Shuling Wang
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA
| | - Sherilyn Collins
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA
| | - Amber Walker
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA
| | - Kai-Xuan Shi
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA
| | - Joshua A Alcantara
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA
| | - Jonathan Neumann
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA
| | | | - Nicholas T Seyfried
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Andrea J Tenner
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
- Department of Molecular Biology & Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697, USA
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, 92697, USA
| | - Frank M LaFerla
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Lindsay A Hohsfield
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, 92697, USA
| | - Grant R MacGregor
- Transgenic Mouse Facility, ULAR, Office of Research, University of California, Irvine, CA, 92697-2300, USA.
- Department of Developmental and Cell Biology, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697, USA.
| | - Kim N Green
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697-4545, USA.
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
257
|
Lee E, Chang Y. Modulating Neuroinflammation as a Prospective Therapeutic Target in Alzheimer's Disease. Cells 2025; 14:168. [PMID: 39936960 PMCID: PMC11817173 DOI: 10.3390/cells14030168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
The recent approval of lecanemab highlights that the amyloid beta (Aβ) protein is an important pathological target in Alzheimer's disease (AD) and further emphasizes the significance of neuroinflammatory pathways in regulating Aβ accumulation. Indeed, Aβ accumulation triggers microglia activation, which are key mediators in neuroinflammation. The inflammatory responses in this process can lead to neuronal damage and functional decline. Microglia secrete proinflammatory cytokines that accelerate neuronal death and release anti-inflammatory cytokines and growth factors contributing to neuronal recovery and protection. Thus, microglia play a dual role in neurodegeneration and neuroprotection, complicating their function in AD. Therefore, elucidating the complex interactions between Aβ protein, microglia, and neuroinflammation is essential for developing new strategies for treating AD. This review investigates the receptors and pathways involved in activating microglia and aims to enhance understanding of how these processes impact neuroinflammation in AD, as well as how they can be regulated. This review also analyzed studies reported in the existing literature and ongoing clinical trials. Overall, these studies will contribute to understanding the regulatory mechanisms of neuroinflammation and developing new therapies that can slow the pathological progression of AD.
Collapse
Affiliation(s)
- Eunshil Lee
- Institute of Biomedical Engineering Research, Kyungpook National University, Daegu 41944, Republic of Korea;
| | - Yongmin Chang
- Institute of Biomedical Engineering Research, Kyungpook National University, Daegu 41944, Republic of Korea;
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Radiology, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| |
Collapse
|
258
|
Gongwer MW, Etienne F, Moca EN, Chappell MS, Blagburn-Blanco SV, Riley JP, Enos AS, Haratian M, Qi A, Rojo R, Wilke SA, Pridans C, DeNardo LA, De Biase LM. Microglia regulate nucleus accumbens synaptic development and circuit function underlying threat avoidance behaviors. RESEARCH SQUARE 2025:rs.3.rs-5837701. [PMID: 39975894 PMCID: PMC11838711 DOI: 10.21203/rs.3.rs-5837701/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
While CNS microglia have well-established roles in synapse pruning during neurodevelopment, only a few studies have identified roles for microglia in synapse formation. These studies focused on the cortex and primary sensory circuits during restricted developmental time periods, leaving substantial gaps in our understanding of the early developmental functions of microglia. Here we investigated how the absence of microglia impacts synaptic development in the nucleus accumbens (NAc), a region critical for emotional regulation and motivated behaviors and where dysfunction is implicated in psychiatric disorders that arise early in life. Using a genetically modified mouse that lacks microglia (Csf1r ΔFIRE/ΔFIRE), we found blunted excitatory synapse formation in the NAc. This effect was most prominent during the second and third postnatal weeks, when we previously found microglia to be overproduced, and was accompanied by an increase in presynaptic release probability and alterations in postsynaptic kinetics. Tissue-level NAc proteomics confirmed that microglial absence impacted numerous proteins involved in synapse structure, trans-synaptic signaling, and pre-synaptic function. However, microglial absence did not perturb levels of astrocyte-derived cues and adhesive proteins that promote synaptogenesis, suggesting that reduced synapse number may be caused by absence of a microglial-derived synaptogenic cue. Although observed electrophysiological synaptic changes were largely normalized by adulthood, we identified lasting effects of microglial absence on threat avoidance behavior, and these behavioral effects were directly associated with alterations of NAc neuronal activity. Together, these results indicate a critical role for microglia in regulating the synaptic landscape of the developing NAc and in establishing functional circuits underlying adult behavioral repertoires.
Collapse
Affiliation(s)
- Michael W Gongwer
- Department of Physiology, University of California Los Angeles, CA, USA
- Neuroscience Interdepartmental Program, University of California Los Angeles, CA, USA
- UCLA Medical Scientist Training Program, University of California Los Angeles, CA, USA
| | - Fanny Etienne
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Eric N Moca
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Megan S Chappell
- Department of Physiology, University of California Los Angeles, CA, USA
- Neuroscience Interdepartmental Program, University of California Los Angeles, CA, USA
| | - Sara V Blagburn-Blanco
- Department of Physiology, University of California Los Angeles, CA, USA
- Neuroscience Interdepartmental Program, University of California Los Angeles, CA, USA
- UCLA Medical Scientist Training Program, University of California Los Angeles, CA, USA
| | - Jack P Riley
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Alexander S Enos
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Melody Haratian
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Alex Qi
- Department of Psychiatry, University of California Los Angeles, CA, USA
| | - Rocio Rojo
- Institution for Regeneration and Repair, University of Edinburgh, Scotland
| | - Scott A Wilke
- Department of Psychiatry, University of California Los Angeles, CA, USA
| | - Clare Pridans
- Institution for Regeneration and Repair, University of Edinburgh, Scotland
| | - Laura A DeNardo
- Department of Physiology, University of California Los Angeles, CA, USA
| | | |
Collapse
|
259
|
Lapin D, Sharma A, Wang P. Extracellular cold-inducible RNA-binding protein in CNS injury: molecular insights and therapeutic approaches. J Neuroinflammation 2025; 22:12. [PMID: 39838468 PMCID: PMC11752631 DOI: 10.1186/s12974-025-03340-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025] Open
Abstract
Central nervous system (CNS) injuries, such as ischemic stroke (IS), intracerebral hemorrhage (ICH) and traumatic brain injury (TBI), are a significant global burden. The complex pathophysiology of CNS injury is comprised of primary and secondary injury. Inflammatory secondary injury is incited by damage-associated molecular patterns (DAMPs) which signal a variety of resident CNS cells and infiltrating immune cells. Extracellular cold-inducible RNA-binding protein (eCIRP) is a DAMP which acts through multiple immune and non-immune cells to promote inflammation. Despite the well-established role of eCIRP in systemic and sterile inflammation, its role in CNS injury is less elucidated. Recent literature suggests that eCIRP is a pleiotropic inflammatory mediator in CNS injury. eCIRP is also being evaluated as a clinical biomarker to indicate prognosis in CNS injuries. This review provides a broad overview of CNS injury, with a focus on immune-mediated secondary injury and neuroinflammation. We then review what is known about eCIRP in CNS injury, and its known mechanisms in both CNS and non-CNS cells, identifying opportunities for further study. We also explore eCIRP's potential as a prognostic marker of CNS injury severity and outcome. Next, we provide an overview of eCIRP-targeting therapeutics and suggest strategies to develop these agents to ameliorate CNS injury. Finally, we emphasize exploring novel molecular mechanisms, aside from neuroinflammation, by which eCIRP acts as a critical mediator with significant potential as a therapeutic target and prognostic biomarker in CNS injury.
Collapse
Affiliation(s)
- Dmitriy Lapin
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
- Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
| | - Archna Sharma
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA.
- Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA.
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA.
- Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA.
| |
Collapse
|
260
|
Li LJ, Liang SY, Sun XY, Zhu J, Niu XY, Du XY, Huang YR, Liu RT. Microglial double stranded DNA accumulation induced by DNase II deficiency drives neuroinflammation and neurodegeneration. J Neuroinflammation 2025; 22:11. [PMID: 39833906 PMCID: PMC11745000 DOI: 10.1186/s12974-025-03333-6] [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: 11/07/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Deoxyribonuclease 2 (DNase II) is pivotal in the clearance of cytoplasmic double stranded DNA (dsDNA). Its deficiency incurs DNA accumulation in cytoplasm, which is a hallmark of multiple neurodegenerative diseases. Our previous study showed that neuronal DNase II deficiency drove tau hyperphosphorylation and neurodegeneration (Li et al., Transl Neurodegener 13:39, 2024). Although it has been verified that DNase II participates in type I interferons (IFN-I) mediated autoinflammation and senescence in peripheral systems, the role of microglial DNase II in neuroinflammation and neurodegenerative diseases such as Alzheimer's disease (AD) is still unknown. METHODS The levels of microglial DNase II in triple transgenic AD mice (3xTg-AD) were measured by immunohistochemistry. The cognitive performance of microglial DNase II deficient WT and AD mice was determined using the Morris water maze test, Y-maze test, novel object recognition test and open filed test. To investigate the impact of microglial DNase II deficiency on microglial morphology, cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway and IFN-I pathway, neuroinflammation, synapses loss, amyloid pathology and tauopathy, the levels of cGAS-STING and IFN-I pathway related protein, gliosis and proinflammatory cytokines, synaptic protein, complement protein, Aβ levels, phosphorylated tau in the brains of the microglial DNase II deficient WT and AD mice were evaluated by immunolabeling, immunoblotting, q-PCR or ELISA. RESULTS We found that the levels of DNase II were significantly decreased in the microglia of 3xTg-AD mice. Microglial DNase II deficiency altered microglial morphology and transcriptional signatures, activated the cGAS-STING and IFN-I pathway, initiated neuroinflammation, led to synapse loss via complement-dependent pathway, increased Aβ levels and tauopathy, and induced cognitive decline. CONCLUSIONS Our study shows the effect of microglial DNase II deficiency and cytoplasmic accumulated dsDNA on neuroinflammation, and reveals the initiatory mechanism of AD pathology, suggesting that DNase II is a potential target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Ling-Jie Li
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shi-Yu Liang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao-Ying Sun
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
| | - Jie Zhu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao-Yun Niu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
- College of Life Science, Ningxia University, Yinchuan, 750021, China
| | - Xiao-Yu Du
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ya-Ru Huang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China.
| | - Rui-Tian Liu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Haidian District, Beijing, 100190, China.
| |
Collapse
|
261
|
Milligan C, Cowley DO, Stewart W, Curry AM, Forbes E, Rector B, Hastie A, Liu L, Hawkins GA. Enhanced Interleukin 6 Trans-Signaling Modulates Disease Process in Amyotrophic Lateral Sclerosis Mouse Models. Brain Sci 2025; 15:84. [PMID: 39851451 PMCID: PMC11764401 DOI: 10.3390/brainsci15010084] [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: 12/21/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/26/2025] Open
Abstract
Background/Objectives: Charcot first described ALS in 1869, but the specific mechanisms that mediate the disease pathology are still not clear. Intense research efforts have provided insight into unique neuroanatomical regions, specific neuronal populations and genetic associations for ALS and other neurodegenerative diseases; however, the experimental results also suggest a convergence of these events to common toxic pathways. We propose that common toxic pathways can be therapeutically targeted, and this intervention will be effective in slowing progression and improving patient quality of life. Here, we focus on understanding the role of IL6 trans-signaling in ALS disease processes. Methods: We leveraged unique mouse models of IL6 trans-signaling that we developed that recapitulate the production of active sIL6R in a genotypic and quantitative fashion observed in humans. Given that the SOD1 transgenic mouse is one of the most highly studied and characterized models of ALS, we bred SOD1G93A mice with IL6R trans-signaling mice to determine how enhanced trans-signaling influenced symptom onset and pathological processes, including neuromuscular junction (NMJ) denervation, glial activation and motoneuron (MN) survival. Results: The results indicate that in animals with enhanced trans-signaling, symptom onset and pathological processes were accelerated, suggesting a role in disease modification. Administration of an IL6R functional blocking antibody failed to alter accelerated symptom onset and disease progression. Conclusions: Future work to investigate the site-specific influence of enhanced IL6 trans-signaling and the tissue-specific bioavailability of potential therapeutics will be necessary to identify targets for precise therapeutic interventions that may limit disease progression in the 60% of ALS patients who inherit the common Il6R Asp358Ala variant.
Collapse
Affiliation(s)
- Carol Milligan
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Dale O. Cowley
- Department of Genetics and Animal Models Core Facility, University of North Carolina, Chapel Hill, NC 27599, USA;
| | - William Stewart
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Alyson M. Curry
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Elizabeth Forbes
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Brian Rector
- Department of Internal Medicine Section on Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Annette Hastie
- Department of Internal Medicine Section on Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Liang Liu
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Gregory A. Hawkins
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
262
|
Peters JJ, Teng C, Peng K, Li X. Deciphering the Blood-Brain Barrier Paradox in Brain Metastasis Development and Therapy. Cancers (Basel) 2025; 17:298. [PMID: 39858080 PMCID: PMC11764143 DOI: 10.3390/cancers17020298] [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: 12/07/2024] [Revised: 01/01/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Gatekeeper or accomplice? That is the paradoxical role of the blood-brain barrier (BBB) in developing brain metastasis (BM). BM occurs when cancerous cells from primary cancer elsewhere in the body gain the ability to metastasize and invade the brain parenchyma despite the formidable defense of the BBB. These metastatic cells manipulate the BBB's components, changing them from gatekeepers of the brain to accomplices that aid in their progression into the brain tissue. This dual role of the BBB-as both a protective system and a potential facilitator of metastatic cells-highlights its complexity. Even with metastasis therapy such as chemotherapy, BM usually recurs due to the BBB limiting the crossing of drugs via the efflux transporters; therefore, treatment efficacy is limited. The pathophysiology is also complex, and our understanding of the paradoxical interplay between the BBB components and metastatic cells still needs to be improved. However, advancements in clinical research are helping to bridge the knowledge gap, which is essential for developing effective metastasis therapy. By targeting the BBB neurovascular unit components such as the polarization of microglia, astrocytes, and pericytes, or by utilizing technological tools like focused ultrasound to transiently disrupt the BBB and therapeutic nanoparticles to improve drug delivery efficiency to BM tissue, we can better address this pathology. This narrative review delves into the latest literature to analyze the paradoxical role of the BBB components in the manifestation of BM and explores potential therapeutic avenues targeting the BBB-tumor cell interaction.
Collapse
Affiliation(s)
- Jens Jeshu Peters
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.P.); (C.T.); (K.P.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chubei Teng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.P.); (C.T.); (K.P.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Kang Peng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.P.); (C.T.); (K.P.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.P.); (C.T.); (K.P.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha 410008, China
- Xiangya School of Medicine, Central South University, Changsha 410008, China
| |
Collapse
|
263
|
Gongwer MW, Etienne F, Moca EN, Chappell MS, Blagburn-Blanco SV, Riley JP, Enos AS, Haratian M, Qi A, Rojo R, Wilke SA, Pridans C, DeNardo LA, De Biase LM. Microglia regulate nucleus accumbens synaptic development and circuit function underlying threat avoidance behaviors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633068. [PMID: 39868334 PMCID: PMC11761117 DOI: 10.1101/2025.01.15.633068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
While CNS microglia have well-established roles in synapse pruning during neurodevelopment, only a few studies have identified roles for microglia in synapse formation. These studies focused on the cortex and primary sensory circuits during restricted developmental time periods, leaving substantial gaps in our understanding of the early developmental functions of microglia. Here we investigated how the absence of microglia impacts synaptic development in the nucleus accumbens (NAc), a region critical for emotional regulation and motivated behaviors and where dysfunction is implicated in psychiatric disorders that arise early in life. Using a genetically modified mouse that lacks microglia (Csf1r ΔFIRE/ΔFIRE), we found blunted excitatory synapse formation in the NAc. This effect was most prominent during the second and third postnatal weeks, when we previously found microglia to be overproduced, and was accompanied by an increase in presynaptic release probability and alterations in postsynaptic kinetics. Tissue-level NAc proteomics confirmed that microglial absence impacted numerous proteins involved in synapse structure, trans-synaptic signaling, and pre-synaptic function. However, microglial absence did not perturb levels of astrocyte-derived cues and adhesive proteins that promote synaptogenesis, suggesting that reduced synapse number may be caused by absence of a microglial-derived synaptogenic cue. Although observed electrophysiological synaptic changes were largely normalized by adulthood, we identified lasting effects of microglial absence on threat avoidance behavior, and these behavioral effects were directly associated with alterations of NAc neuronal activity. Together, these results indicate a critical role for microglia in regulating the synaptic landscape of the developing NAc and in establishing functional circuits underlying adult behavioral repertoires.
Collapse
Affiliation(s)
- Michael W Gongwer
- Department of Physiology, University of California Los Angeles, CA, USA
- Neuroscience Interdepartmental Program, University of California Los Angeles, CA, USA
- UCLA Medical Scientist Training Program, University of California Los Angeles, CA, USA
| | - Fanny Etienne
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Eric N Moca
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Megan S Chappell
- Department of Physiology, University of California Los Angeles, CA, USA
- Neuroscience Interdepartmental Program, University of California Los Angeles, CA, USA
| | - Sara V Blagburn-Blanco
- Department of Physiology, University of California Los Angeles, CA, USA
- Neuroscience Interdepartmental Program, University of California Los Angeles, CA, USA
- UCLA Medical Scientist Training Program, University of California Los Angeles, CA, USA
| | - Jack P Riley
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Alexander S Enos
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Melody Haratian
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Alex Qi
- Department of Psychiatry, University of California Los Angeles, CA, USA
| | - Rocio Rojo
- Institution for Regeneration and Repair, University of Edinburgh, Scotland
| | - Scott A Wilke
- Department of Psychiatry, University of California Los Angeles, CA, USA
| | - Clare Pridans
- Institution for Regeneration and Repair, University of Edinburgh, Scotland
| | - Laura A DeNardo
- Department of Physiology, University of California Los Angeles, CA, USA
| | | |
Collapse
|
264
|
Poongodi R, Hsu YW, Yang TH, Huang YH, Yang KD, Lin HC, Cheng JK. Stem Cell-Derived Extracellular Vesicle-Mediated Therapeutic Signaling in Spinal Cord Injury. Int J Mol Sci 2025; 26:723. [PMID: 39859437 PMCID: PMC11765593 DOI: 10.3390/ijms26020723] [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: 12/23/2024] [Revised: 01/14/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have emerged as a promising therapeutic strategy for spinal cord injury (SCI). These nanosized vesicles possess unique properties such as low immunogenicity and the ability to cross biological barriers, making them ideal carriers for delivering bioactive molecules to injured tissues. MSC-EVs have been demonstrated to exert multiple beneficial effects in SCI, including reducing inflammation, promoting neuroprotection, and enhancing axonal regeneration. Recent studies have delved into the molecular mechanisms underlying MSC-EV-mediated therapeutic effects. Exosomal microRNAs (miRNAs) have been identified as key regulators of various cellular processes involved in SCI pathogenesis and repair. These miRNAs can influence inflammation, oxidative stress, and apoptosis by modulating gene expression. This review summarized the current state of MSC-EV-based therapies for SCI, highlighting the underlying mechanisms and potential clinical applications. We discussed the challenges and limitations of translating these therapies into clinical practice, such as inconsistent EV production, complex cargo composition, and the need for targeted delivery strategies. Future research should focus on optimizing EV production and characterization, identifying key therapeutic miRNAs, and developing innovative delivery systems to maximize the therapeutic potential of MSC-EVs in SCI.
Collapse
Affiliation(s)
- Raju Poongodi
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan; (R.P.); (T.-H.Y.)
| | - Yung-Wei Hsu
- Department of Anesthesiology, MacKay Memorial Hospital, Taipei 10449, Taiwan; (Y.-W.H.); (Y.-H.H.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
| | - Tao-Hsiang Yang
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan; (R.P.); (T.-H.Y.)
| | - Ya-Hsien Huang
- Department of Anesthesiology, MacKay Memorial Hospital, Taipei 10449, Taiwan; (Y.-W.H.); (Y.-H.H.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
| | - Kuender D. Yang
- Institute of Long-Term Care, MacKay Medical College, New Taipei City 25245, Taiwan;
- MacKay Children’s Hospital, Taipei 10449, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Hsin-Chieh Lin
- Department of Materials Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan;
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Jen-Kun Cheng
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan; (R.P.); (T.-H.Y.)
- Department of Anesthesiology, MacKay Memorial Hospital, Taipei 10449, Taiwan; (Y.-W.H.); (Y.-H.H.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
| |
Collapse
|
265
|
Ju JJ, Hang LH. Neuroinflammation and iron metabolism after intracerebral hemorrhage: a glial cell perspective. Front Neurol 2025; 15:1510039. [PMID: 39882361 PMCID: PMC11774705 DOI: 10.3389/fneur.2024.1510039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Intracerebral hemorrhage (ICH) is the most common subtype of hemorrhagic stroke causing significant morbidity and mortality. Previously clinical treatments for ICH have largely been based on a single pathophysiological perspective, and there remains a lack of curative interventions. Following the rupture of cerebral blood vessels, blood metabolites activate resident immune cells such as microglia and astrocytes, and infiltrate peripheral immune cells, leading to the release of a series of inflammatory mediators. Degradation of hemoglobin produces large amounts of iron ions, leading to an imbalance of iron homeostasis and the production of large quantities of harmful hydroxyl radicals. Neuroinflammation and dysregulation of brain iron metabolism are both important pathophysiological changes in ICH, and both can exacerbate secondary brain injury. There is an inseparable relationship between brain iron metabolism disorder and activated glial cells after ICH. Glial cells participate in brain iron metabolism through various mechanisms; meanwhile, iron accumulation exacerbates neuroinflammation by activating inflammatory signaling pathways modulating the functions of inflammatory cells, and so on. This review aims to explore neuroinflammation from the perspective of iron metabolism, linking the complex pathophysiological changes, delving into the exploration of treatment approaches for ICH, and offering insights that could enhance clinical management strategies.
Collapse
Affiliation(s)
- Jia-Jun Ju
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, China
| | - Li-Hua Hang
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, China
- Kunshan Cancer Pain Prevention and Treatment Key Laboratory, Kunshan, China
| |
Collapse
|
266
|
Lin L, Guo Z, Ren Z, Feng Y, Fang P, Wang T, Chen M. Bibliometric insights into astrocytic roles in depression and treatment. Front Cell Neurosci 2025; 18:1521398. [PMID: 39882216 PMCID: PMC11775634 DOI: 10.3389/fncel.2024.1521398] [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: 11/01/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025] Open
Abstract
Objective Depression is a mental disorder that significantly impairs both physical and mental health. Recent studies have shown that reactive astrogliosis have gained significant attention for their involvement in the pathophysiology of depression. However, there is no bibliometric analysis in this research field. This study aims to provide a comprehensive overview of the knowledge structure and research hotspots regarding the role of astrocytes in the mechanisms and treatment of depression through bibliometric analysis. The scope of the literature review encompasses both basic and clinical research. Methods Publications related to astrocytes in depression and treatment from 2014 to 2023 were searched in the Web of Science Core Collection (WoSCC) database. VOSviewer, CiteSpace, and the R package "bibliometrix" were used to conduct this bibliometric analysis. Results From 2014 to 2023, a total of 1,502 documents from 78 countries on astrocytes in depression and treatment were analyzed from 169 journals, with the most co-cited journals being the Journal of Neuroscience and PNAS. China Medical University was the most productive institution. The analysis identified key authors like Verkhratsky Alexei and Baoman Li, and major co-cited references by Rajkowska and Liddelow. Keywords such as "synaptic plasticity," "astrocytes," and "neuroinflammation" revealed research trends focusing on molecular mechanisms, gut microbiota, and inflammation. Conclusion This is the first bibliometric study to comprehensively summarize the research trends and advancements regarding astrocytes in depression and its treatment. Through this bibliometric analysis, we aim to enhance the understanding of the significance of astrocytes in depression research and provide new perspectives and insights for future investigations. We hope that this study will facilitate a broader integration of basic and clinical research, offering novel approaches for the treatment of depression.
Collapse
Affiliation(s)
- Linsun Lin
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, Macao SAR, China
- Huizhou Health Sciences Polytechnic, Huizhou, China
| | - Ziyi Guo
- National Engineering Laboratory for Internet Medical Systems and Applications, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhuoyu Ren
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanchen Feng
- The First Clinical Medical School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Peigang Fang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Tao Wang
- Encephalopathy Center, The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Min Chen
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, Macao SAR, China
- Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
267
|
Qiu D, Wang L, Wang L, Dong Y. Human platelet lysate: a potential therapeutic for intracerebral hemorrhage. Front Neurosci 2025; 18:1517601. [PMID: 39881806 PMCID: PMC11774881 DOI: 10.3389/fnins.2024.1517601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Intracerebral hemorrhage (ICH) is a major public health challenge worldwide, and is associated with elevated rates of mortality, disability, and morbidity, especially in low- and middle-income nations. However, our knowledge of the detailed molecular processes involved in ICH remains insufficient, particularly those involved in the secondary injury stage, resulting in a lack of effective treatments for ICH. Human platelet lysates (HPL) are abundant in bioactive factors, and numerous studies have demonstrated their beneficial effects on neurological diseases, including their anti-neuroinflammatory ability, anti-oxidant effects, maintenance of blood-brain barrier integrity, and promotion of neurogenesis. In this review, we thoroughly explore the potential of HPL for treating ICH from three critical perspectives: the rationale for selecting HPL as a treatment for ICH, the mechanisms through which HPL contributes to ICH management, and the additional measures necessary for HPL as a treatment for ICH. We elucidate the role of platelets in ICH pathophysiology and highlight the limitations of the current treatment options and advancements in preclinical research on the application of HPL in neurological disorders. Furthermore, historical developments and preparation methods of HPL in the field of biomedicine are discussed. Additionally, we summarize the bioactive molecules present in HPL and their potential therapeutic effects in ICH. Finally, we outline the issues that must be addressed regarding utilizing HPL as a treatment modality for ICH.
Collapse
Affiliation(s)
- Dachang Qiu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Lin Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Lanlan Wang
- Department of Geriatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yongfei Dong
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
268
|
Kandror EK, Wang A, Carriere M, Peterson A, Liao W, Tjärnberg A, Fung JH, Mahbubani KT, Loper J, Pangburn W, Xu Y, Saeb-Parsy K, Rabadan R, Maniatis T, Rizvi AH. Enhancer Dynamics and Spatial Organization Drive Anatomically Restricted Cellular States in the Human Spinal Cord. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632483. [PMID: 39829819 PMCID: PMC11741326 DOI: 10.1101/2025.01.10.632483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Here, we report the spatial organization of RNA transcription and associated enhancer dynamics in the human spinal cord at single-cell and single-molecule resolution. We expand traditional multiomic measurements to reveal epigenetically poised and bivalent active transcriptional enhancer states that define cell type specification. Simultaneous detection of chromatin accessibility and histone modifications in spinal cord nuclei reveals previously unobserved cell-type specific cryptic enhancer activity, in which transcriptional activation is uncoupled from chromatin accessibility. Such cryptic enhancers define both stable cell type identity and transitions between cells undergoing differentiation. We also define glial cell gene regulatory networks that reorganize along the rostrocaudal axis, revealing anatomical differences in gene regulation. Finally, we identify the spatial organization of cells into distinct cellular organizations and address the functional significance of this observation in the context of paracrine signaling. We conclude that cellular diversity is best captured through the lens of enhancer state and intercellular interactions that drive transitions in cellular state. This study provides fundamental insights into the cellular organization of the healthy human spinal cord.
Collapse
Affiliation(s)
- Elena K. Kandror
- Department of Neuroscience and Waisman Center, University of Wisconsin-Madison
| | - Anqi Wang
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Medical Center
| | | | - Alexis Peterson
- Department of Neuroscience and Waisman Center, University of Wisconsin-Madison
| | | | - Andreas Tjärnberg
- Department of Neuroscience and Waisman Center, University of Wisconsin-Madison
| | - Jun Hou Fung
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Medical Center
| | - Krishnaa T. Mahbubani
- Cambridge Biorepository for Translational Medicine, Cambridge NIHR Biomedical Research Centre, Cambridge, UK
- Department of Surgery, University of Cambridge, Cambridge, UK
- Department of Haematology, Cambridge Stem Cell Institute, Cambridge, UK
| | - Jackson Loper
- Department of Statistics, University of Michigan Ann Arbor
| | - William Pangburn
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center
| | - Yuchen Xu
- Department of Neuroscience and Waisman Center, University of Wisconsin-Madison
| | - Kourosh Saeb-Parsy
- Cambridge Biorepository for Translational Medicine, Cambridge NIHR Biomedical Research Centre, Cambridge, UK
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Raul Rabadan
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Medical Center
| | - Tom Maniatis
- New York Genome Center
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center
| | - Abbas H. Rizvi
- Department of Neuroscience and Waisman Center, University of Wisconsin-Madison
- Lead contact
| |
Collapse
|
269
|
Wu C, Li M, Chen Z, Feng S, Deng Q, Duan R, Liu TCY, Yang L. Remote photobiomodulation ameliorates behavioral and neuropathological outcomes in a rat model of repeated closed head injury. Transl Psychiatry 2025; 15:8. [PMID: 39799140 PMCID: PMC11724958 DOI: 10.1038/s41398-025-03228-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 12/13/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025] Open
Abstract
Repeated closed-head injuries (rCHI) from activities like contact sports, falls, military combat, and traffic accidents pose a serious risk due to their cumulative impact on the brain. Often, rCHI is not diagnosed until symptoms of irreversible brain damage appear, highlighting the need for preventive measures. This study assessed the prophylactic efficacy of remote photobiomodulation (PBM) targeted at the lungs against rCHI-induced brain injury and associated behavioral deficits. Utilizing the "Marmarou" weight-drop model, rCHI was induced in rats on days 0, 5, and 10. Remote PBM, employing an 808 nm continuous wave laser, was administered daily in 2-min sessions per lung side over 20 days. Behavioral deficits were assessed through three-chamber social interaction, forced swim, grip strength, open field, elevated plus maze, and Barnes maze tests. Immunofluorescence staining and 3D reconstruction evaluated neuronal damage, apoptosis, degeneration, and the morphology of microglia and astrocytes, as well as astrocyte and microglia-mediated excessive synapse elimination. Additionally, 16S rDNA amplicon sequencing analyzed changes in the lung microbiome following remote PBM treatment. Results demonstrated that remote PBM significantly improved depressive-like behaviors, motor dysfunction, and social interaction impairment while enhancing grip strength and reducing neuronal damage, apoptosis, and degeneration induced by rCHI. Analysis of lung microbiome changes revealed an enrichment of lipopolysaccharide (LPS) biosynthesis pathways, suggesting a potential link to neuroprotection. Furthermore, remote PBM mitigated hyperactivation of cortical microglia and astrocytes and significantly reduced excessive synaptic phagocytosis by these cells, highlighting its potential as a preventive strategy for rCHI with neuroprotective effects.
Collapse
Affiliation(s)
- Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China
| | - Meng Li
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China
| | - Zhe Chen
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China
| | - Shu Feng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China
| | - Qianting Deng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China
| | - Rui Duan
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Timon Cheng-Yi Liu
- Laboratory of Laser Sports Medicine, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
270
|
Onat F, Andersson M, Çarçak N. The Role of Glial Cells in the Pathophysiology of Epilepsy. Cells 2025; 14:94. [PMID: 39851521 PMCID: PMC11763453 DOI: 10.3390/cells14020094] [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: 11/12/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
Epilepsy is a chronic neurological disorder marked by recurrent seizures, significantly impacting individuals worldwide. Current treatments are often ineffective for a third of patients and can cause severe side effects, necessitating new therapeutic approaches. Glial cells, particularly astrocytes, microglia, and oligodendrocytes, are emerging as crucial targets in epilepsy management. Astrocytes regulate neuronal homeostasis, excitability, and synaptic plasticity, playing key roles in maintaining the blood-brain barrier (BBB) and mediating neuroinflammatory responses. Dysregulated astrocyte functions, such as reactive astrogliosis, can lead to abnormal neuronal activity and seizure generation. They release gliotransmitters, cytokines, and chemokines that may exacerbate or mitigate seizures. Microglia, the innate immune cells of the CNS, contribute to neuroinflammation, glutamate excitotoxicity, and the balance between excitatory and inhibitory neurotransmission, underscoring their dual role in seizure promotion and protection. Meanwhile, oligodendrocytes, primarily involved in myelination, also modulate axonal excitability and contribute to the neuron-glia network underlying seizure pathogenesis. Understanding the dynamic interactions of glial cells with neurons provides promising avenues for novel epilepsy therapies. Targeting these cells may lead to improved seizure control and better clinical outcomes, offering hope for patients with refractory epilepsy.
Collapse
Affiliation(s)
- Filiz Onat
- Department of Medical Pharmacology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, 34684 Istanbul, Türkiye
- Institute of Health Sciences, Department of Neuroscience, Acibadem Mehmet Ali Aydinlar University, 34684 Istanbul, Türkiye
| | - My Andersson
- Department of Experimental Medicine, Faculty of Medicine, Lund University, 221 00 Lund, Sweden;
| | - Nihan Çarçak
- Institute of Health Sciences, Department of Neuroscience, Acibadem Mehmet Ali Aydinlar University, 34684 Istanbul, Türkiye
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, 34452 Istanbul, Türkiye
| |
Collapse
|
271
|
Wang P, Hu J, Chen C, Jiang Z, Zhang Y, Lin K, Liao L, Wang X. The immune regulatory mechanism of ketamine-induced psychiatric disorders: A new perspective on drug-induced psychiatric symptoms. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111194. [PMID: 39542202 DOI: 10.1016/j.pnpbp.2024.111194] [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: 08/13/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
Ketamine, a psychoactive substance strictly regulated by international drug conventions, is classified as a "new type drug" due to its excitatory, hallucinogenic, or inhibitory effects. The etiology of ketamine-induced psychiatric symptoms is multifaceted, with the immune regulatory mechanism being the most prominent among several explanatory theories. In recent years, the interaction between the immune system and nervous system have garnered significant attention in neuropsychiatric disorder research. Notably, the infiltration of peripheral lymphocytes into the central nervous system has emerged as an early hallmark of certain neuropsychiatric disorders. However, a notable gap exists in the current literature, regarding the immune regulatory mechanisms, specifically the peripheral immune alterations, associated with ketamine-induced psychiatric symptoms. To address this void, this article endeavors to provide a comprehensive overview of the pathophysiological processes implicated in psychiatric disorders or symptoms, encompassing those elicited by ketamine. This analysis delves into aspects such as nerve damage, alterations within the central immune system, and the regulation of the peripheral immune system. By emphasizing the intricate crosstalk between the peripheral immune system and the central nervous system, this study sheds light on their collaborative role in the onset and progression of psychiatric diseases or symptoms. This insight offers fresh perspectives on the underlying mechanisms, diagnosis and therapeutic strategies for mental disorders stemming from drug abuse.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Junmei Hu
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Congliang Chen
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Zihan Jiang
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yu Zhang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Kexin Lin
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Linchuan Liao
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| | - Xia Wang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
272
|
Zhang Z, Niu K, Huang T, Guo J, Xarbat G, Gong X, Gao Y, Liu F, Cheng S, Su W, Yang F, Liu Z, Ginhoux F, Zhang T. Microglia depletion reduces neurodegeneration and remodels extracellular matrix in a mouse Parkinson's disease model triggered by α-synuclein overexpression. NPJ Parkinsons Dis 2025; 11:15. [PMID: 39779738 PMCID: PMC11711755 DOI: 10.1038/s41531-024-00846-4] [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: 05/15/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Chronic neuroinflammation with sustained microglial activation occurs in Parkinson's disease (PD), yet the mechanisms and exact contribution of these cells to the neurodegeneration remains poorly understood. In this study, we induced progressive dopaminergic neuron loss in mice via rAAV-hSYN injection to cause the neuronal expression of α-synuclein, which produced neuroinflammation and behavioral alterations. We administered PLX5622, a colony-stimulating factor 1 receptor inhibitor, for 3 weeks prior to rAAV-hSYN injection, maintaining it for 8 weeks to eliminate microglia. This chronic treatment paradigm prevented the development of motor deficits and concomitantly preserved dopaminergic neuron cell and weakened α-synuclein phosphorylation. Gene expression profiles related to extracellular matrix (ECM) remodeling were increased after microglia depletion in PD mice, which were further validated on protein level. We demonstrated that microglia exert adverse effects during α-synuclein-overexpression-induced neuronal lesion formation, and their depletion remodels ECM and aids recovery following insult.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China
| | - Kun Niu
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China
| | - Taoying Huang
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China
| | - Jiali Guo
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China
| | - Gongbikai Xarbat
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China
| | - Xiaoli Gong
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing, 100069, China
| | - Yunke Gao
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China
| | - Feiyang Liu
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China
| | - Shan Cheng
- Department of Medical Genetics and Developmental Biology, Capital Medical University, Beijing, 100069, China
| | - Wenting Su
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
| | - Fei Yang
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Florent Ginhoux
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Republic of Singapore
- Gustave Roussy Cancer Campus, Villejuif, 94800, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Ting Zhang
- Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
273
|
Lee EJ, Lee MJ, Ryu YJ, Nam SH, Kim R, Song S, Park K, Park YJ, Kim JI, Koh SH, Chang MS. Neuroplasticity therapy using glia-like cells derived from human mesenchymal stem cells for the recovery of cerebral infarction sequelae. Mol Ther 2025; 33:356-374. [PMID: 39563032 PMCID: PMC11764092 DOI: 10.1016/j.ymthe.2024.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/12/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024] Open
Abstract
Despite a dramatic increase in ischemic stroke incidence worldwide, effective therapies for attenuating sequelae of cerebral infarction are lacking. This study investigates the use of human mesenchymal stem cells (hMSCs) induced toward glia-like cells (ghMSCs) to ameliorate chronic sequelae resulting from cerebral infarction. Transcriptome analysis demonstrated that ghMSCs exhibited astrocytic characteristics, and assessments conducted ex vivo using organotypic brain slice cultures demonstrated that ghMSCs exhibited superior neuroregenerative and neuroprotective activity against ischemic damage compared to hMSCs. The observed beneficial effects of ghMSCs were diminished by pre-treatment with a CXCR2 antagonist, indicating a direct role for CXCR2 signaling. Studies conducted in rats subjected to cerebral infarction demonstrated that ghMSCs restored neurobehavioral functions and reduced chronic brain infarction in a dose-dependent manner when transplanted at the subacute-to-chronic phase. These beneficial impacts were also inhibited by a CXCR2 antagonist. Molecular analyses confirmed that increased neuroplasticity contributed to ghMSCs' neuroregenerative effects. These data indicate that ghMSCs hold promise for treating refractory sequelae resulting from cerebral infarction by enhancing neuroplasticity and identify CXCR2 signaling as an important mediator of ghMSCs' mechanism of action.
Collapse
Affiliation(s)
- Eun Ji Lee
- Department of Translational Medical Science, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul 04763, Republic of Korea
| | - Min-Ju Lee
- Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy and Dental Research Institute, Seoul National University School of Dentistry, Seoul 03080, Republic of Korea
| | - Ye Jin Ryu
- Department of Translational Medical Science, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul 04763, Republic of Korea
| | - Sang-Hyeon Nam
- Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy and Dental Research Institute, Seoul National University School of Dentistry, Seoul 03080, Republic of Korea
| | - Rokhyun Kim
- Genomic Medicine Institute (GMI), Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sehyeon Song
- Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy and Dental Research Institute, Seoul National University School of Dentistry, Seoul 03080, Republic of Korea; Interdisciplinary Program in Neuroscience, Seoul National University College of Natural Sciences, Seoul 08826, Republic of Korea
| | - Kyunghyuk Park
- Genomic Medicine Institute (GMI), Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea
| | - Young Jun Park
- Genomic Medicine Institute (GMI), Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea
| | - Jong-Il Kim
- Genomic Medicine Institute (GMI), Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Seong-Ho Koh
- Department of Translational Medical Science, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul 04763, Republic of Korea; Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Gyeonggi-do 11923, Republic of Korea.
| | - Mi-Sook Chang
- Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy and Dental Research Institute, Seoul National University School of Dentistry, Seoul 03080, Republic of Korea; Interdisciplinary Program in Neuroscience, Seoul National University College of Natural Sciences, Seoul 08826, Republic of Korea; Neuroscience Research Institute, Seoul National University, Seoul 03080, Republic of Korea.
| |
Collapse
|
274
|
Labarta-Bajo L, Allen NJ. Astrocytes in aging. Neuron 2025; 113:109-126. [PMID: 39788083 PMCID: PMC11735045 DOI: 10.1016/j.neuron.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/05/2024] [Accepted: 12/11/2024] [Indexed: 01/12/2025]
Abstract
The mammalian nervous system is impacted by aging. Aging alters brain architecture, is associated with molecular damage, and can manifest with cognitive and motor deficits that diminish the quality of life. Astrocytes are glial cells of the CNS that regulate the development, function, and repair of neural circuits during development and adulthood; however, their functions in aging are less understood. Astrocytes change their transcriptome during aging, with astrocytes in areas such as the cerebellum, the hypothalamus, and white matter-rich regions being the most affected. While numerous studies describe astrocyte transcriptional changes in aging, many questions still remain. For example, how is astrocyte function altered by transcriptional changes that occur during aging? What are the mechanisms promoting astrocyte aged states? How do aged astrocytes impact brain function? This review discusses features of aged astrocytes and their potential triggers and proposes ways in which they may impact brain function and health span.
Collapse
Affiliation(s)
- Lara Labarta-Bajo
- Salk Institute for Biological Studies, Molecular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Nicola J Allen
- Salk Institute for Biological Studies, Molecular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
275
|
Groh J, Simons M. White matter aging and its impact on brain function. Neuron 2025; 113:127-139. [PMID: 39541972 DOI: 10.1016/j.neuron.2024.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/16/2024] [Accepted: 10/18/2024] [Indexed: 11/17/2024]
Abstract
Aging has a detrimental impact on white matter, resulting in reduced volume, compromised structural integrity of myelinated axons, and an increase in white matter hyperintensities. These changes are closely linked to cognitive decline and neurological disabilities. The deterioration of myelin and its diminished ability to regenerate as we age further contribute to the progression of neurodegenerative disorders. Understanding these changes is crucial for devising effective disease prevention strategies. Here, we will discuss the structural alterations in white matter that occur with aging and examine the cellular and molecular mechanisms driving these aging-related transformations. We highlight how the progressive disruption of white matter may initiate a self-perpetuating cycle of inflammation and neural damage.
Collapse
Affiliation(s)
- Janos Groh
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.
| |
Collapse
|
276
|
Gharibani P, Abramson E, Shanmukha S, Smith MD, Godfrey WH, Lee JJ, Hu J, Baydyuk M, Dorion MF, Deng X, Guo Y, Calle AJ, A Hwang S, Huang JK, Calabresi PA, Kornberg MD, Kim PM. The protein kinase C modulator bryostatin-1 therapeutically targets microglia to attenuate neuroinflammation and promote remyelination. Sci Transl Med 2025; 17:eadk3434. [PMID: 39772770 DOI: 10.1126/scitranslmed.adk3434] [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: 08/18/2023] [Revised: 09/19/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025]
Abstract
In multiple sclerosis (MS), microglia and macrophages within the central nervous system (CNS) play an important role in determining the balance among demyelination, neurodegeneration, and myelin repair. Phagocytic and regenerative functions of these CNS innate immune cells support remyelination, whereas chronic and maladaptive inflammatory activation promotes lesion expansion and disability, particularly in the progressive forms of MS. No currently approved drugs convincingly target microglia and macrophages within the CNS, contributing to the lack of therapies aimed at promoting remyelination and slowing disease progression for individuals with MS. Here, we found that the protein kinase C (PKC)-modulating drug bryostatin-1 (bryo-1), a CNS-penetrant compound with an established human safety profile, shifts the transcriptional programs of microglia and CNS-associated macrophages from a proinflammatory phenotype to a regenerative phenotype in vitro and in vivo. Treatment of microglia with bryo-1 stimulated scavenger pathways, phagocytosis, and secretion of factors that prevented the activation of neuroinflammatory reactive astrocytes while also promoting neuroaxonal health and oligodendrocyte differentiation. In line with these findings, systemic treatment of mice with bryo-1 augmented remyelination after a focal demyelinating injury. Our results demonstrate the potential of bryo-1 and possibly a wider class of PKC modulators as myelin-regenerative and supportive agents in MS and other neurologic diseases.
Collapse
Affiliation(s)
- Payam Gharibani
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Efrat Abramson
- Interdepartmental Neuroscience Program, Yale University School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Shruthi Shanmukha
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Matthew D Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Wesley H Godfrey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Judy J Lee
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jingwen Hu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Maryna Baydyuk
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Marie-France Dorion
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Xiaojing Deng
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yu Guo
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Andrew J Calle
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Soonmyung A Hwang
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jeffrey K Huang
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Michael D Kornberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Paul M Kim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
277
|
Lu E, Zhou K, Miao J, Zhu Y, Tang J, Du S, Feng Y, Jiang L, Jiang T, Huang T, Li P, Miao X, Han Q, Xiao J. A cryo-shocked M2 macrophages based treatment strategy promoting repair of spinal cord injury via immunomodulation and axonal regeneration effects. J Nanobiotechnology 2025; 23:8. [PMID: 39757205 DOI: 10.1186/s12951-024-03018-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/14/2024] [Indexed: 01/07/2025] Open
Abstract
Recovery from spinal cord injury (SCI) is often impeded by neuroinflammation, scar formation, and limited axonal regeneration. To tackle these issues, we developed an innovative biomimetic drug delivery system using liquid nitrogen-treated M2 macrophages (LNT M2) which internalized paclitaxel (PTX) nanoparticles beforehand. These were incorporated into a gelatin methacryloyl (GelMA) scaffold, creating a multifunctional, injectable treatment for single-dose administration. The LNT M2 inherited the inflammatory factor/chemokine receptors from the living M2 macrophages and thus possessing significant inflammatory neutralizing effect. In addition, the scaffold provides slow, sustained release of PTX, promoting axonal regeneration and suppressing scar formation in SCI rats. The LNT M2-based dual-functional scaffold significantly enhances motor function, reduces neuroinflammation, and accelerates axonal regeneration by modulating the inflammatory microenvironment and preventing the formation of glial and fibrotic scars. This approach combines the regenerative effects of low-dose PTX with the immunoregulatory properties of LNT M2, leading to remarkable neurological recovery in SCI rats. Moreover, the scaffold's straightforward preparation, ease of standardization, and "ready-to-use" nature make it a promising candidate for acute SCI intervention and future clinical applications.
Collapse
Affiliation(s)
- Ermei Lu
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Department of Emergency, Wenzhou Central Hospital, Wenzhou, 325099, Zhejiang, China
| | - Kecheng Zhou
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Jiansen Miao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yanlin Zhu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Department of Emergency and Critical Disease, Songjiang Research Institute, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
| | - Jiyao Tang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Department of Emergency and Critical Disease, Songjiang Research Institute, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
| | - Siting Du
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yanzhen Feng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Linyuan Jiang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Tianyao Jiang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ting Huang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ping Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xinjun Miao
- Department of Emergency, Wenzhou Central Hospital, Wenzhou, 325099, Zhejiang, China.
| | - Qi Han
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Department of Emergency and Critical Disease, Songjiang Research Institute, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jian Xiao
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, Zhejiang, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
278
|
Li M, Chen M, Li H, Gao D, Zhao L, Zhu M. Glial cells improve Parkinson's disease by modulating neuronal function and regulating neuronal ferroptosis. Front Cell Dev Biol 2025; 12:1510897. [PMID: 39830208 PMCID: PMC11739109 DOI: 10.3389/fcell.2024.1510897] [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: 10/14/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
The main characteristics of Parkinson's disease (PD) are the loss of dopaminergic (DA) neurons and abnormal aggregation of cytosolic proteins. However, the exact pathogenesis of PD remains unclear, with ferroptosis emerging as one of the key factors driven by iron accumulation and lipid peroxidation. Glial cells, including microglia, astrocytes, and oligodendrocytes, serve as supportive cells in the central nervous system (CNS), but their abnormal activation can lead to DA neuron death and ferroptosis. This paper explores the interactions between glial cells and DA neurons, reviews the changes in glial cells during the pathological process of PD, and reports on how glial cells regulate ferroptosis in PD through iron homeostasis and lipid peroxidation. This opens up a new pathway for basic research and therapeutic strategies in Parkinson's disease.
Collapse
Affiliation(s)
- Mengzhu Li
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Mengxuan Chen
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Haiyan Li
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Da Gao
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lijun Zhao
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Meiling Zhu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
279
|
Gong XR, You XR, Guo MR, Ding XY, Ma BX. Exploring the mechanism of Pujin oral liquid in the treatment of preterm white matter injury using network pharmacology and molecular docking. Medicine (Baltimore) 2025; 104:e40799. [PMID: 40184095 PMCID: PMC11709195 DOI: 10.1097/md.0000000000040799] [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: 11/10/2023] [Revised: 01/04/2024] [Accepted: 11/14/2024] [Indexed: 04/05/2025] Open
Abstract
We aimed to elucidate the pharmacological mechanisms of Pujin oral liquid in treating preterm white matter injury (PWMI). The Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform was used to identify Pujin oral liquid's active ingredients and predict their targets. The known targets related to treating PWMI were identified from the GeneCards, Online Mendelian Inheritance in Man, DisGeNet, PharmGKB, and CTD databases. A drug-disease intersecting protein-protein interaction network using a STRING database was built; gene ontology function and Kyoto Encyclopedia of Genes and Genomes signaling pathway enrichment analyses were performed on common target genes using the Metascape database. Molecular docking of the active ingredients and key targets was validated using the AutoDock Vina software. In total, 470 Pujin oral liquid targets and 13,290 disease targets were screened from multiple databases, and Venn analysis identified 407 common targets. Protein-protein interaction analysis showed that Pujin oral liquid may impact SRC, MAPK3, MAPK1, TP53, STAT3, AKT1, PIK3R1, JUN, RELA, CTNNB1, and ESR1. Moreover, gene ontology functional analysis revealed processes such as the response to inorganic substances, cellular response to organic cyclic compounds, response to xenobiotic stimuli, regulation of system processes, and protein phosphorylation. The main signaling pathways were neuroactive ligand-receptor interaction and the cGMP-PKG, JAK-STAT, and cAMP signaling pathways. Molecular docking showed that the active ingredients' small molecules bond strongly to target proteins. The therapeutic effect of Pujin oral liquid on PWMI is multifaceted, involving multiple targets and pathways. Its clinical application in treating preterm white matter injuries is promising.
Collapse
Affiliation(s)
- Xing-Ruo Gong
- Department of Pediatrics, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Rui You
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, China
| | - Mei-Ran Guo
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, China
| | - Xue-Ying Ding
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Bing-Xiang Ma
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| |
Collapse
|
280
|
Liu Q, Xia LX, Yi WZ, Wu YN, Gu SS, Chen JY, Liu TT, Lu YH, Cui YH, Meng J, Pan HW. Inhibition of Retinal Neovascularization by BEZ235: Targeting the Akt/4EBP1/Cyclin D1 Pathway in Endothelial Cells. Invest Ophthalmol Vis Sci 2025; 66:66. [PMID: 39888634 PMCID: PMC11784786 DOI: 10.1167/iovs.66.1.66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 12/17/2024] [Indexed: 02/01/2025] Open
Abstract
Purpose To investigate the therapeutic efficacy of BEZ235, a dual PI3K/mTOR inhibitor, in suppressing pathological neovascularization in an oxygen-induced retinopathy (OIR) mouse model and explore the role of cyclin D1 in endothelial cell cycle regulation. Methods Single-cell RNA sequencing was performed to analyze gene expression and cell-cycle alterations in retinal endothelial cells under normoxic and OIR conditions. The effects of BEZ235 on human umbilical vein endothelial cells (HUVECs) and human retinal microvascular endothelial cells (HRMECs) were evaluated by assessing cell viability, cell-cycle progression, proliferation, migration, and tube formation. In the OIR mouse model, retinal neovascularization was evaluated by retinal flatmount immunofluorescence staining, hematoxylin and eosin (H&E) staining, quantitative reverse-transcription polymerase chain reaction (RT-qPCR), and western blot analyses. The in vivo toxicity of BEZ235 was evaluated by electroretinography (ERG) and histological examination of the heart, liver, spleen, lungs, and kidneys. Results In vitro, BEZ235 significantly inhibited cell cycle progression by downregulating cyclin D1 at both mRNA and protein levels, inducing G0/G1 phase arrest. This led to significant reductions in cell viability, proliferation, migration, and tube formation. In the OIR model, BEZ235 substantially decreased neovascularization and improved vascular organization. BEZ235 mediates its effects by inhibiting the PI3K/Akt/mTOR pathway, reducing Akt and 4E-binding protein 1 (4EBP1) phosphorylation levels, thus downregulating cyclin D1 expression. ERG and histological examination suggested that BEZ235 did not induce evident retinal or systemic toxicity at the dosage used to inhibit retinal neovascularization. Conclusions BEZ235 effectively inhibits retinal neovascularization by downregulating cyclin D1 via 4EBP1 phosphorylation inhibition, highlighting its potential as a promising therapeutic agent for retinal neovascularization diseases.
Collapse
Affiliation(s)
- Qi Liu
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ling-Xiao Xia
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Wan-Zhao Yi
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ya-Ni Wu
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Shuo-Shuo Gu
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jian-Ying Chen
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ting-Ting Liu
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ying-Hui Lu
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yu-Hong Cui
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jing Meng
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
- The Affiliated Shunde Hospital of Jinan University, Foshan, China
| | - Hong-Wei Pan
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
281
|
Xu C, Jiang X, Yin W, Lyu L, Tan H, He W, Wang W, Zhou Z, Zhou H, Xu B, Yu Y, Wang Y, Zhou P, Liu Y, Xu J, Gou M, Hu Y. Microgel-encapsulated tetrandrine nanoparticles promote spinal cord repair by sustaining neuroinflammation inhibition. J Mater Chem B 2025; 13:683-694. [PMID: 39620256 DOI: 10.1039/d4tb02080d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Traumatic spinal cord injury (SCI) initiates an intricate secondary injury cascade, characterized by persistent inflammatory responses with neurotoxic microglia and astrocyte activation. Inhibition of neuroinflammation would significantly benefit SCI treatment. Here, tetrandrine with anti-neuroinflammatory activity was delivered into the intrathecal space for SCI treatment. The tetrandrine was encapsulated in MPEG-PDLLA nanoparticles and further loaded into GelMA microgels via a fast 3D printing process based on digital light. After intrathecal injection, the drug-loaded microgels could sustain the release of tetrandrine in the intrathecal space, resulting in efficient repair of the injured spinal cord with recovery of function. Its mechanisms were associated with the modulation of neurotoxic microglia and astrocytes as well as their crosstalk. This work demonstrates a tetrandrine-loaded microgel with potential application in SCI treatment via sustained inhibition of neuroinflammation.
Collapse
Affiliation(s)
- Chongxi Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Xuebing Jiang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wumeng Yin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Liang Lyu
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Huixin Tan
- Department of Rehabilitation, West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, China
| | - Wenbo He
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Wangyang Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhengyu Zhou
- Clinical Medicine School of Chongqing Medical University, Chongqing Medical University, Chongqing, 401331, China
| | - Hongyu Zhou
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610000, China
| | - Bin Xu
- Neurosurgery Department of West China Tianfu Hospital, Sichuan University, Chengdu, 610000, China
| | - Yang Yu
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Yuelong Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Peizhi Zhou
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Yi Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Jianguo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Maling Gou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Yu Hu
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| |
Collapse
|
282
|
Zhang I, Maysinger D, Beus M, Mravak A, Yu Z, Perić Bakulić M, Dion PA, Rouleau GA, Bonačić-Koutecký V, Antoine R, Sanader Maršić Ž. Gold nanoclusters Au 25AcCys 18 normalize intracellular ROS without increasing cytoplasmic alarmin acHMGB1 abundance in human microglia and neurons. NANOSCALE 2025; 17:1092-1104. [PMID: 39607703 DOI: 10.1039/d4nr03512g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
This study focuses on the modulatory effects of gold nanoclusters with 25 gold atoms and 18 acetyl cysteines (Au25AcCys18) in human microglia, human iPSC-derived neurons and SH-SY5Y differentiated human neuronal cells. The combination of chemical, biological, and computational methods shows the well-retained viability of these human cells treated with Au25AcCys18, interactions between Au25AcCys18 and transcription factor TFEB (computational approach), interactions between TFEB and HMGB1 (proximity ligation assay and molecular modeling using AlphaFold), modulation of the abundance and location of acHMGB1 by Au25AcCys18 (immunocytochemistry), and the reduction of ROS in cells treated with Au25AcCys18 (CellROX live imaging). These novel findings in human neural cells, particularly neurons, encourage further studies in experimental animal models of neurological disorders and/or human organoids to exploit the unique structural and photophysical properties of gold nanoclusters and to better understand their ability to modulate molecular mechanisms in human cells.
Collapse
Affiliation(s)
- Issan Zhang
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, H3G 1Y6 Montreal, Canada.
| | - Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, H3G 1Y6 Montreal, Canada.
| | - Maja Beus
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, H3G 1Y6 Montreal, Canada.
| | - Antonija Mravak
- Faculty of Science, University of Split, Ruđera Boškovića 33, 21000 Split, Croatia.
| | - Ziqi Yu
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Martina Perić Bakulić
- Faculty of Chemistry and Technology, University of Split, Ruđera Boškovića 35, 21000 Split, Croatia
- Center of Excellence for Science and Technology, Integration of Mediterranean Region (STIM), Faculty of Science, University of Split, Ruđera Boškovića 33, 21000 Split, Croatia
| | - Patrick A Dion
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Guy A Rouleau
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Vlasta Bonačić-Koutecký
- Center of Excellence for Science and Technology, Integration of Mediterranean Region (STIM), Faculty of Science, University of Split, Ruđera Boškovića 33, 21000 Split, Croatia
- Chemistry Department, Humboldt University of Berlin, Brook-Taylor-Strasse 2, 12489 Berlin, Germany
| | - Rodolphe Antoine
- Institut Lumière Matière, CNRS UMR 5306, Université Claude Bernard Lyon 1, Univ. Lyon, 69622 Villeurbanne Cedex, France
| | - Željka Sanader Maršić
- Faculty of Science, University of Split, Ruđera Boškovića 33, 21000 Split, Croatia.
- Center of Excellence for Science and Technology, Integration of Mediterranean Region (STIM), Faculty of Science, University of Split, Ruđera Boškovića 33, 21000 Split, Croatia
| |
Collapse
|
283
|
Tsering W, de la Rosa A, Ruan IY, Philips JL, Bathe T, Villareal JA, Prokop S. Preferential clustering of microglia and astrocytes around neuritic plaques during progression of Alzheimer's disease neuropathological changes. J Neurochem 2025; 169:e16275. [PMID: 39655787 PMCID: PMC11629606 DOI: 10.1111/jnc.16275] [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: 09/04/2024] [Revised: 10/28/2024] [Accepted: 11/17/2024] [Indexed: 12/13/2024]
Abstract
Neuroinflammation plays an important role in the pathological cascade of Alzheimer's disease (AD) along with aggregation of extracellular amyloid-β (Aβ) plaques and intracellular aggregates of tau protein. In animal models of amyloidosis, local immune activation is centered around Aβ plaques, which are usually of uniform morphology, dependent on the transgenic model used. In postmortem human brains a diversity of Aβ plaque morphologies is seen including diffuse plaques (non-neuritic plaques, non-NP), dense-core plaques, cotton-wool plaques, and NP. In a recent study, we demonstrated that during the progression of Alzheimer's disease neuropathologic changes (ADNC), a transformation of non-NP into NP occurs which is tightly linked to the emergence of cortical, but not hippocampal neurofibrillary tangle (NFT) pathology. This highlights the central role of NP in AD pathogenesis as well as brain region-specific differences in NP formation. In order to correlate the transformation of plaque types with local immune activation, we quantified the clustering and phenotype of microglia and accumulation of astrocytes around non-NP and NP during the progression of ADNC. We hypothesize that glial clustering occurs in response to formation of neuritic dystrophy around NP. First, we show that Iba1-positive microglia preferentially cluster around NP. Utilizing microglia phenotypic markers, we furthermore demonstrate that CD68-positive phagocytic microglia show a strong preference to cluster around NP in both the hippocampus and frontal cortex. A similar preferential clustering is observed for CD11c and ferritin-positive microglia in the frontal cortex, while this preference is less pronounced in the hippocampus, highlighting differences between hippocampal and cortical Aβ plaques. Glial fibrillary acidic protein-positive astrocytes showed a clear preference for clustering around NP in both the frontal cortex and hippocampus. These data support the notion that NP are intimately associated with the neuroimmune response in AD and underscore the importance of the interplay of protein deposits and the immune system in the pathophysiology of AD.
Collapse
Affiliation(s)
- Wangchen Tsering
- Center for Translational Research in Neurodegenerative Disease, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- Department of Neuroscience, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- McKnight Brain Institute, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Ana de la Rosa
- Center for Translational Research in Neurodegenerative Disease, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Isabelle Y. Ruan
- Center for Translational Research in Neurodegenerative Disease, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Jennifer L. Philips
- Center for Translational Research in Neurodegenerative Disease, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- Department of Pathology, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Tim Bathe
- Center for Translational Research in Neurodegenerative Disease, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- Department of Pathology, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Jonathan A. Villareal
- Center for Translational Research in Neurodegenerative Disease, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- Department of Pathology, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- McKnight Brain Institute, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- Department of Pathology, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- Norman Fixel Institute for Neurological DiseasesUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
284
|
Yuan Y, Liu H, Dai Z, He C, Qin S, Su Z. From Physiology to Pathology of Astrocytes: Highlighting Their Potential as Therapeutic Targets for CNS Injury. Neurosci Bull 2025; 41:131-154. [PMID: 39080102 PMCID: PMC11748647 DOI: 10.1007/s12264-024-01258-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/15/2024] [Indexed: 01/19/2025] Open
Abstract
In the mammalian central nervous system (CNS), astrocytes are the ubiquitous glial cells that have complex morphological and molecular characteristics. These fascinating cells play essential neurosupportive and homeostatic roles in the healthy CNS and undergo morphological, molecular, and functional changes to adopt so-called 'reactive' states in response to CNS injury or disease. In recent years, interest in astrocyte research has increased dramatically and some new biological features and roles of astrocytes in physiological and pathological conditions have been discovered thanks to technological advances. Here, we will review and discuss the well-established and emerging astroglial biology and functions, with emphasis on their potential as therapeutic targets for CNS injury, including traumatic and ischemic injury. This review article will highlight the importance of astrocytes in the neuropathological process and repair of CNS injury.
Collapse
Affiliation(s)
- Yimin Yuan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
- Department of Pain Medicine, School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Hong Liu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Ziwei Dai
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Cheng He
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Shangyao Qin
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China.
| | - Zhida Su
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
285
|
Xu L, Zhang Z, Feng Z, Niu S, Yang L, Xiao B, Jin XM, Ma C, Fan H, Xiao S, Chai Z. Study on the anti-Parkinson's disease activity mechanism and preparation of panaxadiol. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156296. [PMID: 39631295 DOI: 10.1016/j.phymed.2024.156296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/21/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Traditional Chinese medicine offers unique and valuable resources for the exploration of novel approaches to disease treatment. Panaxadiol, an active compound derived from the transformation of ginsenosides. It has a variety of pharmacological activities. However, there is a lack of research regarding its effects on Parkinson's disease (PD). PURPOSE AND STUDY DESIGN This study investigates the activity and mechanism of panaxadiol in the treatment of PD both in vivo and in vitro. In addition, a new method has been developed to extract panaxadiol. RESULTS The results showed that in vitro, panaxadiol can reduce the viability and cytotoxicity of BV2 cells induced by LPS, and inhibit the production of pro-inflammatory factors through the TLR4/MyD88/NF-κB pathway. In vivo, panaxadiol can improve motor and intestinal dysfunction in PD mice and repair the loss of DA neurons. Further studies have shown that panaxadiol treatment alleviates damage to the blood-brain barrier (BBB), inhibits neuroinflammation in the substantia nigra (SN), and further reduces damage to DA neurons. Subsequently, it was found that panaxadiol alleviated peripheral inflammation and significantly restored the intestinal microbial community. Further mechanistic studies found that panaxadiol treatment inhibited the TLR4/MyD88/NF-κB signaling pathway and its downstream pro-inflammatory products in the SN. In addition, Additionally, the response surface method was employed to identify the optimal conditions for extracting panaxadiol using a deep eutectic solvent (NADES). CONCLUSION In summary, this study has found for the first time that panaxadiol can effectively improve the symptoms of PD by regulating neuroinflammation, repairing the BBB, and gut microbiota. Meanwhile, adopting a green extraction process significantly increases the content of panaxadiol, providing a new direction for the development of PD candidate drugs.
Collapse
Affiliation(s)
- Lei Xu
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Zhongjin Zhang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Ziqi Feng
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Sixiang Niu
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Lixia Yang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Baoguo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200025, China
| | - Xiao-Ming Jin
- Department of Anatomy and Cell Biology, Department of Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute. Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Cungen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Huijie Fan
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China.
| | - Shengnan Xiao
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China.
| | - Zhi Chai
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China.
| |
Collapse
|
286
|
Li S, Chen Y, Chen G. Cognitive disorders: Potential astrocyte-based mechanism. Brain Res Bull 2025; 220:111181. [PMID: 39725239 DOI: 10.1016/j.brainresbull.2024.111181] [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: 08/25/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Cognitive disorders are a common clinical manifestation, including a deterioration in the patient's memory ability, attention, executive power, language, and other functions. The contributing factors of cognitive disorders are numerous and diverse in nature, including organic diseases and other mental disorders. Neurodegenerative diseases are a common type of organic disease related to the pathology of neuronal death and disruption of glial cell balance, ultimately accompanied with cognitive impairment. Thus, cognitive disorder frequently serves as an extremely critical indicator of neurodegenerative disorders. Cognitive impairments negatively affect patients' daily lives. However, our understanding of the precise pathogenic pathways of cognitive defects remains incomplete. The most prevalent kind of glial cells in the central nervous system are called astrocytes. They have a unique significance in cerebral function because of their wide range of functions in maintaining homeostasis in the central nervous system, regulating synaptic plasticity, and so on. Dysfunction of astrocytes is intimately linked to cognitive disorders, and we are attempting to understand this phenomenon predominantly from those perspectives: neuroinflammation, astrocytic senescence, connexin, Ca2 + signaling, mitochondrial dysfunction, and the glymphatic system.
Collapse
Affiliation(s)
- Shiyu Li
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yeru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
287
|
Frick LR. Neuroglia in Tourette syndrome and obsessive-compulsive disorder. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:325-334. [PMID: 40148053 DOI: 10.1016/b978-0-443-19102-2.00005-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
In recent years, neuroglia have drawn the attention of researchers in the fields of neurology and psychiatry. Besides their well-known functions providing support to neurons, myelinating axons, and clearing up debris, a constantly growing of evidence indicates that glial cells are key contributors to the pathophysiology of neuropsychiatric disorders. Alterations in microglia, astrocytes, and oligodendrocytes have been described in Tourette syndrome (TS) and obsessive-compulsive disorder (OCD). The sudden onset of tics and OCD-like symptoms after infection in children (Pediatric Autoimmune Neuropsychiatric Disorder Associated with Streptococcal Infections) suggests a connection with the immune system; in fact, neuroinflammation has been reported. Many imaging studies revealed abnormal myelination in the brain of TS and OCD patients, highlighting the implication of oligodendroglia in the connectivity alterations. Moreover, animal models have unveiled a cell-autonomous role of microglia and astrocytes in the etiology of pathologic grooming, which links these glial cells to the related disorder trichotillomania. This chapter reviews the state of the art and current gaps in the literature, proposing possible pathomechanisms and future research directions.
Collapse
Affiliation(s)
- Luciana R Frick
- Departments of Neurology and Medicine, Neuroscience Graduate Program, Jacobs School of Medicine & Biomedical Sciences, State University of New York at Buffalo, Clinical and Translational Research Center, Buffalo, NY, United States.
| |
Collapse
|
288
|
Kalaki-Jouybari F, Shirzad M, Javan M, Ghasemi-Kasman M, Pouramir M. Co-administration of Naringin and NLRP3 Inhibitor Improves Myelin Repair and Mitigates Oxidative Stress in Cuprizone-Induced Demyelination Model. Curr Neuropharmacol 2025; 23:475-491. [PMID: 40123459 DOI: 10.2174/1570159x23666241206102022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/26/2024] [Accepted: 05/30/2024] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Naringin and MCC950 as an inflammasome inhibitor have exhibited numerous pharmacological activities, including antioxidant and anti-inflammatory effects. The present study has examined the combined impacts of naringin and MCC950 on the levels of oxidative stress, demyelination, and inflammation in the cuprizone (CPZ)-induced demyelination model. METHODS In order to induce demyelination, CPZ (0.2% w/w) was added to the normal diet of mice for 42 days. Subsequently, the male C57BL/6 mice received naringin (oral administration), MCC950 (intraperitoneal injection), or their combination for 14 days. Working memory was tested by the Y maze. FluoroMyelin staining, MOG, and GFAP immunostaining assessed the demyelination extent, myelin intensity, and astrocyte activation, respectively. Oxidant/antioxidant biomarkers were measured using colorimetric techniques. The expression levels of MBP, PDGFRα, Olig2, Nrf2, HO-1, NQO-1, GSK3β, IL1α, and IL18 were assessed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). RESULTS Our results indicated that the co-administration of naringin and MCC950 improved working memory and antioxidant capacity. A significant reduction was found in the extent of demyelination and inflammatory mediatorsin naringin and MCC950-treated mice. In addition, co-administration of naringin and MCC950 elevated the expression levels of pro-myelinating and antioxidant markers. CONCLUSION These findings indicated improvement of the working memory through co-administration of naringin and MCC950, which might be partly mediated by enhancing antioxidant capacity, promoting remyelination, and mitigating inflammation in the CPZ-induced demyelination model.
Collapse
Affiliation(s)
- Fatemeh Kalaki-Jouybari
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Moein Shirzad
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Physiology, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mehdi Pouramir
- Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
289
|
Agner SC, Brier LM, Hill JD, Liu EY, Bice A, Rahn RM, Chen S, Culver JP, Klein RS. Zika virus encephalitis causes transient reduction of functional cortical connectivity. NEUROPHOTONICS 2025; 12:S14603. [PMID: 39610883 PMCID: PMC11603678 DOI: 10.1117/1.nph.12.s1.s14603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 10/16/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024]
Abstract
Significance Determining the long-term cognitive impact of infections is clinically challenging. Using functional cortical connectivity, we demonstrate that interhemispheric cortical connectivity is decreased in individuals with acute Zika virus (ZIKV) encephalitis. This correlates with decreased presynaptic terminals in the somatosensory cortex. During recovery from ZIKV infection, presynaptic terminals recover, which is associated with recovered interhemispheric connectivity. This supports the contribution of synapses in the cortex to functional networks in the brain, which can be detected by widefield optical imaging. Although myeloid cell and astrocyte numbers are still increased during recovery, RNA transcription of multiple proinflammatory cytokines that increase during acute infection decreases to levels comparable to mock-infected mice during recovery. These findings also suggest that the immune response and cytokine-mediated neuroinflammation play significant roles in the integrity of brain networks during and after viral encephalitis. Aim We hypothesized that widefield optical imaging would allow us to assess functional cortical network disruption by ZIKV, including hippocampal-cortical networks. Approach We use widefield optical imaging to measure cortical functional connectivity (FC) in mice during acute infection with, and recovery from, intracranial infection with a mouse-adapted strain of ZIKV. Results Acute ZIKV infection leads to high levels of myeloid cell activation, with loss of neurons and presynaptic termini in the cerebral cortex and associated loss of FC primarily within the somatosensory cortex. During recovery, neuron numbers, synapses, and FC recover to levels near those of healthy mice. However, hippocampal injury and impaired spatial cognition persist. The magnitude of activated myeloid cells during acute infection predicted both recovery of synapses and the degree of FC recovery after recovery from ZIKV infection. Conclusions These findings suggest that a robust inflammatory response may contribute to the health of functional brain networks after recovery from infection.
Collapse
Affiliation(s)
- Shannon C. Agner
- Washington University School of Medicine, Center for Neuroimmunology and Neuroinfectious Diseases, St. Louis, Missouri, United States
- Washington University School of Medicine, Department of Neurology, St. Louis, Missouri, United States
| | - Lindsey M. Brier
- Washington University School of Medicine, Department of Radiology, St. Louis, Missouri, United States
| | - Jeremy D. Hill
- Washington University School of Medicine, Center for Neuroimmunology and Neuroinfectious Diseases, St. Louis, Missouri, United States
- Washington University School of Medicine, Department of Medicine, St. Louis, Missouri, United States
| | - Ethan Y. Liu
- Washington University School of Medicine, Department of Neurology, St. Louis, Missouri, United States
| | - Annie Bice
- Washington University School of Medicine, Department of Radiology, St. Louis, Missouri, United States
| | - Rachel M. Rahn
- Washington University School of Medicine, St. Louis, Missouri, United States
| | - Shengxuan Chen
- Washington University School of Medicine, St. Louis, Missouri, United States
| | - Joseph P. Culver
- Washington University School of Medicine, Department of Radiology, St. Louis, Missouri, United States
- Washington University School of Medicine, St. Louis, Missouri, United States
- Washington University School of Medicine, Departments of Physics, Biomedical Engineering, and Electrical and Systems Engineering, St. Louis, Missouri, United States
| | - Robyn S. Klein
- Washington University School of Medicine, Center for Neuroimmunology and Neuroinfectious Diseases, St. Louis, Missouri, United States
- Western University, Departments of Medicine, Microbiology & Immunology, Western Institute of Neuroscience, London, Ontario, Canada
| |
Collapse
|
290
|
Sun M, Song Y, Hu X, Zhang Z, Tan R, Cai Z, Wang X, Fu Y, You H, Cui S, Zhao W, An J, Chen X, Lu H. Leptin reduces LPS-induced A1 reactive astrocyte activation and inflammation via inhibiting p38-MAPK signaling pathway. Glia 2025; 73:25-37. [PMID: 39310943 DOI: 10.1002/glia.24611] [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: 10/25/2023] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 12/21/2024]
Abstract
Neurotoxic A1 reactive astrocytes are induced by inflammatory stimuli. Leptin has been confirmed to have neuroprotective properties. However, its effect on the activation of A1 astrocytes in infectious inflammation is unclear. In the current study, astrocytes cultured from postnatal day 1 Sprague-Dawley rats were stimulated with lipopolysaccharide (LPS) to induce an acute in vitro inflammatory response. Leptin was applied 6 h later to observe its protective effects. The viability of the astrocytes was assessed. A1 astrocyte activation was determined by analyzing the gene expression of C3, H2-D1, H2-T23, and Serping 1 and secretion of pro-inflammatory cytokines IL-6 and TNF-α. The levels of phospho-p38 (pp38) and nuclear factor-κB (NF-κB) phosphor-p65 (pp65) were measured to explore the possible signaling pathways. Additionally, an LPS-induced inflammatory animal model was established to investigate the in vivo effects of leptin on A1 astrocytic activation. Results showed that in the in vitro culture system, LPS stimulation caused elevated expression of A1 astrocyte-specific genes and the secretion of pro-inflammatory cytokines, indicating the activation of A1 astrocytes. Leptin treatment significantly reversed the LPS induced upregulation in a dose-dependent manner. Similarly, LPS upregulated pp38, NF-κB pp65 protein and inflammatory cytokines were successfully reduced by leptin. In the LPS-induced animal model, the amelioratory effect of leptin on A1 astrocyte activation and inflammation was further confirmed, showed by the reduced sickness behaviors, A1 astrocyte genesis and inflammatory cytokines in vivo. Our results demonstrate that leptin efficiently inhibits LPS-induced neurotoxic activation of A1 astrocytes and neuroinflammation by suppressing p38-MAPK signaling pathway.
Collapse
Affiliation(s)
- Meiqi Sun
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yiqun Song
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xiaoxuan Hu
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
- Department of Human Anatomy and Histoembryology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Zixuan Zhang
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
- Department of Human Anatomy and Histoembryology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Ruolan Tan
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
- Department of Human Anatomy and Histoembryology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Zhenlu Cai
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xinyi Wang
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
- Department of Human Anatomy and Histoembryology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yali Fu
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
- Department of Human Anatomy and Histoembryology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Hongli You
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Simeng Cui
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Wanting Zhao
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jing An
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xinlin Chen
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Haixia Lu
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
291
|
Ferrer RM, Jaspers YRJ, Dijkstra IME, Breeuwsma N, van Klinken J, Romero C, Engelen M, Kemp S, Heine VM. Altered lipid profile and reduced neuronal support in human induced pluripotent stem cell-derived astrocytes from adrenoleukodystrophy patients. J Inherit Metab Dis 2025; 48:e12832. [PMID: 39704488 PMCID: PMC11660744 DOI: 10.1002/jimd.12832] [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: 09/04/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024]
Abstract
X-linked adrenoleukodystrophy (ALD) is a peroxisomal disorder resulting from pathogenic variants in the ABCD1 gene that primarily affects the nervous system and is characterized by progressive axonal degeneration in the spinal cord and peripheral nerves and leukodystrophy. Dysfunction of peroxisomal very long-chain fatty acid (VLCFA) degradation has been implicated in ALD pathology, but the impact on astrocytes, which critically support neuronal function, remains poorly understood. Fibroblasts from four ALD patients were reprogrammed to generate human-induced pluripotent stem cells (hiPSC). hiPSC-derived astrocytes were generated to study the impact of ALD on astrocytic fatty acid homeostasis. Our study reveals significant changes in the lipidome of ALD hiPSC-derived astrocytes, characterized by an enrichment of VLCFAs across multiple lipid classes, including triacylglycerols, cholesteryl esters, and phosphatidylcholines. Importantly, ALD hiPSC-derived astrocytes not only exhibit intrinsic lipid dysregulation but also affect the dendritic tree complexity of neurons in co-culture systems. These findings highlight the cell-autonomous effects of pathogenic variants in the ABCD1 protein on astrocytes and their microenvironment, shed light on potential mechanisms underlying ALD neuropathology, and underscore the critical role of astrocytes in neuronal health.
Collapse
Affiliation(s)
- Roberto Montoro Ferrer
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Pediatric NeurologyEmma Children's Hospital, Amsterdam UMC, Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, University of AmsterdamAmsterdamThe Netherlands
- Department of Complex Trait GeneticsCentre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Yorrick R. J. Jaspers
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
| | - Inge M. E. Dijkstra
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
| | - Nicole Breeuwsma
- Department of Child and Adolescence PsychiatryEmma Children's Hospital, Amsterdam UMC Location, Vrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Jan‐Bert van Klinken
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
- Core Facility MetabolomicsAmsterdam UMC Location, University of AmsterdamAmsterdamThe Netherlands
| | - Cato Romero
- Department of Complex Trait GeneticsCentre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of Child and Adolescence PsychiatryEmma Children's Hospital, Amsterdam UMC Location, Vrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Marc Engelen
- Department of Pediatric NeurologyEmma Children's Hospital, Amsterdam UMC, Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, University of AmsterdamAmsterdamThe Netherlands
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
| | - Vivi M. Heine
- Department of Complex Trait GeneticsCentre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of Child and Adolescence PsychiatryEmma Children's Hospital, Amsterdam UMC Location, Vrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamThe Netherlands
| |
Collapse
|
292
|
Yang H, Mo N, Tong L, Dong J, Fan Z, Jia M, Yue J, Wang Y. Microglia lactylation in relation to central nervous system diseases. Neural Regen Res 2025; 20:29-40. [PMID: 38767474 PMCID: PMC11246148 DOI: 10.4103/nrr.nrr-d-23-00805] [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: 05/15/2023] [Revised: 08/09/2023] [Accepted: 01/08/2024] [Indexed: 05/22/2024] Open
Abstract
The development of neurodegenerative diseases is closely related to the disruption of central nervous system homeostasis. Microglia, as innate immune cells, play important roles in the maintenance of central nervous system homeostasis, injury response, and neurodegenerative diseases. Lactate has been considered a metabolic waste product, but recent studies are revealing ever more of the physiological functions of lactate. Lactylation is an important pathway in lactate function and is involved in glycolysis-related functions, macrophage polarization, neuromodulation, and angiogenesis and has also been implicated in the development of various diseases. This review provides an overview of the lactate metabolic and homeostatic regulatory processes involved in microglia lactylation, histone versus non-histone lactylation, and therapeutic approaches targeting lactate. Finally, we summarize the current research on microglia lactylation in central nervous system diseases. A deeper understanding of the metabolic regulatory mechanisms of microglia lactylation will provide more options for the treatment of central nervous system diseases.
Collapse
Affiliation(s)
- Hui Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Nan Mo
- Department of Clinical Laboratory, The Fourth Clinical Medical College of Zhejiang University of Traditional Chinese Medicine (Hangzhou First People’s Hospital), Hangzhou, Zhejiang Province, China
| | - Le Tong
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jianhong Dong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Ziwei Fan
- Department of Orthopedics (Spine Surgery), the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Mengxian Jia
- Department of Orthopedics (Spine Surgery), the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Juanqing Yue
- Department of Pathology, Affiliated Hangzhou First People’s Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Ying Wang
- Department of Clinical Research Center, Affiliated Hangzhou First People’s Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
293
|
An J, Chen B, Zhang R, Tian D, Shi K, Zhang L, Zhang G, Wang J, Yang H. Therapeutic Potential of Mesenchymal Stem Cell-Derived Exosomes in Spinal Cord Injury. Mol Neurobiol 2025; 62:1291-1315. [PMID: 39312070 DOI: 10.1007/s12035-024-04490-0] [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: 09/11/2023] [Accepted: 09/10/2024] [Indexed: 01/04/2025]
Abstract
Spinal cord injury (SCI) can lead to severe motor and sensory dysfunction, with a high rate of disability and mortality. Due to the complicated pathological process of SCI, there is no effective clinical treatment strategy at present. Although mesenchymal stem cells (MSCs) are effective in the treatment of SCI, their application is limited by factors such as low survival rate, cell dedifferentiation, tumorigenesis, blood-brain barrier, and immune rejection. Fortunately, there is growing evidence that most of the biological and therapeutic effects of MSCs may be mediated by the release of paracrine factors, which are extracellular vesicles called exosomes. Exosomes are small endosomal vesicles with bilaminar membranes that have recently been recognized as key mediators for communication between cells and tissues through the transfer of proteins, lipids, nucleic acids, cytokines, and growth factors. Mesenchymal stem cell-derived exosomes (MSC-exos) play a critical role in SCI repair by promoting angiogenesis and axonal growth, regulating inflammation and immune response, inhibiting apoptosis, and maintaining the integrity of the blood-spinal cord barrier. Furthermore, they can be used to transport genetic material or drugs to target cells, and their relatively small size allows them to permeate the blood-brain barrier. Studies have demonstrated that some exosomal miRNAs derived from MSCs play a significant role in the treatment of SCI. In this review, we summarize recent research advances in MSC-exos and exosomal miRNAs in SCI therapy to better understand this emerging cell-free therapeutic strategy and discuss the advantages and challenges of MSC-exos in future clinical applications.
Collapse
Affiliation(s)
- Jing An
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China
| | - Bo Chen
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China.
| | - Rui Zhang
- Department of Medical Technology, Guiyang Healthcare Vocational University, Guiyang, 550081, Guizhou, China
| | - Ding Tian
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China
| | - Kuohao Shi
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China
| | - Lingling Zhang
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China
| | - Gaorong Zhang
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China
| | - Jingchao Wang
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China
| | - Hao Yang
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China.
| |
Collapse
|
294
|
Boulton M, Al-Rubaie A. Neuroinflammation and neurodegeneration following traumatic brain injuries. Anat Sci Int 2025; 100:3-14. [PMID: 38739360 PMCID: PMC11725545 DOI: 10.1007/s12565-024-00778-2] [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: 12/20/2023] [Accepted: 05/05/2024] [Indexed: 05/14/2024]
Abstract
Traumatic brain injuries (TBI) commonly occur following head trauma. TBI may result in short- and long-term complications which may lead to neurodegenerative consequences, including cognitive impairment post-TBI. When investigating the neurodegeneration following TBI, studies have highlighted the role reactive astrocytes have in the neuroinflammation and degeneration process. This review showcases a variety of markers that show reactive astrocyte presence under pathological conditions, including glial fibrillary acidic protein (GFAP), Crystallin Alpha-B (CRYA-B), Complement Component 3 (C3) and S100A10. Astrocyte activation may lead to white-matter inflammation, expressed as white-matter hyperintensities. Other white-matter changes in the brain following TBI include increased cortical thickness in the white matter. This review addresses the gaps in the literature regarding post-mortem human studies focussing on reactive astrocytes, alongside the potential uses of these proteins as markers in the future studies that investigate the proportions of astrocytes in the post-TBI brain has been discussed. This research may benefit future studies that focus on the role reactive astrocytes play in the post-TBI brain and may assist clinicians in managing patients who have suffered TBI.
Collapse
Affiliation(s)
- Matthew Boulton
- School of Health Sciences, Swinburne University of Technology, Hawthorn, VIC, 3122, Australia
| | - Ali Al-Rubaie
- School of Health Sciences, Swinburne University of Technology, Hawthorn, VIC, 3122, Australia.
| |
Collapse
|
295
|
Maupin EA, Adams KL. Cellular Senescence in Glial Cells: Implications for Multiple Sclerosis. J Neurochem 2025; 169:e16301. [PMID: 39831743 PMCID: PMC11745082 DOI: 10.1111/jnc.16301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
Aging is the most common risk factor for Multiple Sclerosis (MS) disease progression. Cellular senescence, the irreversible state of cell cycle arrest, is the main driver of aging and has been found to accumulate prematurely in neurodegenerative diseases, including Alzheimer's and Parkinson's disease. Cellular senescence in the central nervous system of MS patients has recently gained attention, with several studies providing evidence that demyelination induces cellular senescence, with common hallmarks of p16INK4A and p21 expression, oxidative stress, and senescence-associated secreted factors. Here we discuss the current evidence of cellular senescence in animal models of MS and different glial populations in the central nervous system, highlighting the major gaps in the field that still remain. As premature senescence in MS may exacerbate demyelination and inflammation, resulting in inhibition of myelin repair, it is critical to increase understanding of cellular senescence in vivo, the functional effects of senescence on glial cells, and the impact of removing senescent cells on remyelination and MS. This emerging field holds promise for opening new avenues of treatment for MS patients.
Collapse
Affiliation(s)
- Elizabeth A. Maupin
- Department of Biological SciencesUniversity of Notre DameNotre DameIndianaUSA
| | - Katrina L. Adams
- Department of Biological SciencesUniversity of Notre DameNotre DameIndianaUSA
- The Center for Stem Cells and Regenerative MedicineUniversity of Notre DameNotre DameIndianaUSA
| |
Collapse
|
296
|
Estrella LD, Manganaro JE, Sheldon L, Roland N, Snyder AD, George JW, Emanuel K, Lamberty BG, Stauch KL. Chronic glial activation and behavioral alterations induced by acute/subacute pioglitazone treatment in a mouse model of traumatic brain injury. Brain Behav Immun 2025; 123:64-80. [PMID: 39242055 DOI: 10.1016/j.bbi.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/15/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Abstract
Traumatic brain injury (TBI) is a disabling neurotraumatic condition and the leading cause of injury-related deaths and disability in the United States. Attenuation of neuroinflammation early after TBI is considered an important treatment target; however, while these inflammatory responses can induce secondary brain injury, they are also involved in the repair of the nervous system. Pioglitazone, which activates peroxisome proliferator-activated receptor gamma, has been shown to decrease inflammation acutely after TBI, but the long-term consequences of its use remain unknown. For this reason, the impacts of treatment with pioglitazone during the acute/subacute phase (30 min after injury and each subsequent 24 h for 5 days) after TBI were interrogated during the chronic phase (30- and 274-days post-injury (DPI)) in mice using the controlled cortical impact model of experimental TBI. Acute/subacute pioglitazone treatment after TBI results in long-term deleterious consequences, including disruption of tau homeostasis, chronic glial cell activation, neuronal pathology, and worsened injury severity particularly at 274 DPI, with male mice being more susceptible than female mice. Further, male pioglitazone-treated TBI mice exhibited increased dominant and offensive-like behavior while having a decreased non-social exploring behavior at 274 DPI. After TBI, both sexes exhibited glial activation at 30 DPI when treated with pioglitazone; however, while injury severity was increased in females it was not impacted in male mice. This work reveals that although pioglitazone has been shown to lead to attenuated TBI outcomes acutely, sex-based differences, timing and long-term consequences of treatment with glitazones must be considered and further studied prior to their clinical use for TBI therapy.
Collapse
Affiliation(s)
- L Daniel Estrella
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Jane E Manganaro
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Lexi Sheldon
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Nashanthea Roland
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Austin D Snyder
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Joseph W George
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Katy Emanuel
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Benjamin G Lamberty
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Kelly L Stauch
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA.
| |
Collapse
|
297
|
Sekiya T, Holley MC. The Glial Scar: To Penetrate or Not for Motor Pathway Restoration? Cell Transplant 2025; 34:9636897251315271. [PMID: 40152462 PMCID: PMC11951902 DOI: 10.1177/09636897251315271] [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: 12/25/2024] [Accepted: 01/08/2025] [Indexed: 03/29/2025] Open
Abstract
Although notable progress has been made, restoring motor function from the brain to the muscles continues to be a substantial clinical challenge in motor neuron diseases/disorders such as spinal cord injury (SCI). While cell transplantation has been widely explored as a potential therapeutic method for reconstructing functional motor pathways, there remains considerable opportunity for enhancing its therapeutic effectiveness. We reviewed studies on motor pathway regeneration to identify molecular and ultrastructural cues that could enhance the efficacy of cell transplantation. While the glial scar is often cited as an intractable barrier to axon regeneration, this mainly applies to axons trying to penetrate its "core" to reach the opposite side. However, the glial scar exhibits a "duality," with an anti-regenerative core and a pro-regenerative "surface." This surface permissiveness is attributed to pro-regenerative molecules, such as laminin in the basement membrane (BM). Transplanting donor cells onto the BM, which forms plastically after injury, may significantly enhance the efficacy of cell transplantation. Specifically, forming detour pathways between transplanted cells and endogenous propriospinal neurons on the pro-regenerative BM may efficiently bypass the intractable scar core and promote motor pathway regeneration. We believe harnessing the tissue's innate repair capacity is crucial, and targeting post-injury plasticity in astrocytes and Schwann cells, especially those associated with the BM that has predominantly been overlooked in the field of SCI research, can advance motor system restoration to a new stage. A shift in cell delivery routes-from the traditional intra-parenchymal (InP) route to the transplantation of donor cells onto the pro-regenerative BM via the extra-parenchymal (ExP) route-may signify a transformative step forward in neuro-regeneration research. Practically, however, the complementary use of both InP and ExP methods may offer the most substantial benefit for restoring motor pathways. We aim for this review to deepen the understanding of cell transplantation and provide a framework for evaluating the efficacy of this therapeutic modality in comparison to others.
Collapse
Affiliation(s)
- Tetsuji Sekiya
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Neurological Surgery, Hikone Chuo Hospital, Hikone, Japan
| | - Matthew C. Holley
- Department of Biomedical Science, University of Sheffield, Sheffield, England
| |
Collapse
|
298
|
Yanev P, Martin-Jimenez C, Vesga-Jimenez DJ, Zvinys L, Weinrich N, Cree MA, Preuss TM, Zhang X, Yepes M. Plasminogen activator inhibitor-1 mediates cerebral ischemia-induced astrocytic reactivity. J Cereb Blood Flow Metab 2025; 45:102-114. [PMID: 39113414 PMCID: PMC11572231 DOI: 10.1177/0271678x241270445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 11/20/2024]
Abstract
Although ischemia increases the abundance of plasminogen activator inhibitor-1 (PAI-1), its source and role in the ischemic brain remain unclear. We detected PAI-1-immunoreactive cells with morphological features of reactive astrocytes in the peri-ischemic cortex of mice after an experimentally-induced ischemic lesion, and of a chimpanzee that suffered a naturally-occurring stroke. We found that although the abundance of PAI-1 increases 24 hours after the onset of the ischemic injury in a non-reperfusion murine model of ischemic stroke, at that time-point there is no difference in astrocytic reactivity and the volume of the ischemic lesion between wild-type (Wt) animals and in mice either genetically deficient (PAI-1-/-) or overexpressing PAI-1 (PAI-1Tg). In contrast, 72 hours later astrocytic reactivity and the volume of the ischemic lesion were decreased in PAI-1-/- mice and increased in PAI-1Tg animals. Our immunoblottings and fractal analysis studies show that the abundance of astrocytic PAI-1 rises during the recovery phase from a hypoxic injury, which in turn increases the abundance of glial fibrillary acidic protein (GFAP) and triggers morphological features of reactive astrocytes. These studies indicate that cerebral ischemia-induced release of astrocytic PAI-1 triggers astrocytic reactivity associated with enlargement of the necrotic core.
Collapse
Affiliation(s)
- Pavel Yanev
- Division of Neuropharmacology and Neurologic Diseases, Emory Primate Research Center, Atlanta, GA, USA
| | - Cynthia Martin-Jimenez
- Division of Neuropharmacology and Neurologic Diseases, Emory Primate Research Center, Atlanta, GA, USA
| | | | - Laura Zvinys
- Division of Neuropharmacology and Neurologic Diseases, Emory Primate Research Center, Atlanta, GA, USA
| | - Nicholas Weinrich
- Division of Neuropharmacology and Neurologic Diseases, Emory Primate Research Center, Atlanta, GA, USA
| | - Mary Ann Cree
- Division of Neuropharmacology and Neurologic Diseases, Emory Primate Research Center, Atlanta, GA, USA
| | - Todd M Preuss
- Division of Neuropharmacology and Neurologic Diseases, Emory Primate Research Center, Atlanta, GA, USA
| | - Xiaodong Zhang
- Imaging Center, Emory National Primate Research Center, Atlanta, GA, USA
| | - Manuel Yepes
- Division of Neuropharmacology and Neurologic Diseases, Emory Primate Research Center, Atlanta, GA, USA
- Department of Neurology & Center for Neurodegenerative Disease, Emory University, Atlanta, GA, USA
- Department of Neurology, Veterans Affairs Medical Center, Atlanta, GA, USA
| |
Collapse
|
299
|
Xing F, Su HY, Zhong HY, Li YZ, Zhang YY, Chen L, Zhou XL. Synthesis and biological evaluation of lappaconitine analogues as potential anti-neuroinflammatory agents by side chain modification and scaffold hopping strategy. Bioorg Med Chem 2025; 117:118012. [PMID: 39608210 DOI: 10.1016/j.bmc.2024.118012] [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: 08/16/2024] [Revised: 11/10/2024] [Accepted: 11/17/2024] [Indexed: 11/30/2024]
Abstract
Neuroinflammation mediated by microglia is widely recognized as a key pathophysiological mechanism in neurodegenerative diseases. Lappaconitine (LA) is a natural C18-diterpenoid alkaloid isolated from Aconitum sinomontanum Nakai, and previous study showed that LA and its derivatives inhibited lipopolysaccharide (LPS)-induced nitric oxide (NO) production in RAW264.7 cells. However, the anti-neuroinflammatory effects of LA and its derivatives on microglia are still not clear. Here, LA analogues were designed and synthesized, and the anti-neuroinflammatory activity of the synthesized compounds was screened using LPS-induced overexpression of NO in BV-2 microglia. The screening results showed that compound 10 displayed the highest ability to inhibit NO production (IC50 = 9.98 ± 1.6 µM). Mechanistic investigations revealed that compound 10 attenuated LPS-activated neuroinflammation through suppression of TLR4/MyD88/NF-κB pathway in BV-2 microglia. Acute toxicity assays showed that compound 10 (LD50 = 508.1 mg/kg) was safer relative to LA (LD50 = 30.6 mg/kg). Collectively, our findings show that compound 10 could have potential as anti-neuroinflammatory agents.
Collapse
Affiliation(s)
- Feng Xing
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China; Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Hong-Yi Su
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - He-Yang Zhong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yu-Zhu Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yin-Yong Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Lin Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Xian-Li Zhou
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China; Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China; School of Life Science and Engineering, Yibin Institute of Southwest Jiaotong University, Yibin 644000, China.
| |
Collapse
|
300
|
Endo F. Deciphering the spectrum of astrocyte diversity: Insights into molecular, morphological, and functional dimensions in health and neurodegenerative diseases. Neurosci Res 2025; 210:1-10. [PMID: 39098767 DOI: 10.1016/j.neures.2024.07.008] [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: 03/29/2024] [Revised: 07/11/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024]
Abstract
Astrocytes are the most abundant and morphologically complex glial cells that play active roles in the central nervous system (CNS). Recent research has identified shared and region-specific astrocytic genes and functions, elucidated the cellular origins of their regional diversity, and uncovered the molecular networks for astrocyte morphology, which are essential for their functional complexity. Reactive astrocytes exhibit a wide range of functional diversity in a context-specific manner in CNS disorders. This review discusses recent advances in understanding the molecular and morphological diversity of astrocytes in healthy individuals and those with neurodegenerative diseases, such as Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Fumito Endo
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8601, Japan.
| |
Collapse
|