1
|
Gu H, Zhang Y, Sun J, Liu L, Liu Z. Exploring the effect and mechanism of action of Jinlida granules (JLD) in the treatment of diabetes-associated cognitive impairment based on network pharmacology with experimental validation. Ann Med 2025; 57:2445181. [PMID: 39723533 DOI: 10.1080/07853890.2024.2445181] [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: 04/18/2024] [Revised: 08/19/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
OBJECTIVES To explore the effect and the probable mechanisms of JLD in the treatment of type 2 diabetes mellitus (T2DM) - associated cognitive impairment (TDACI). METHODS The effect of JLD in combating TDACI was assessed in T2DM model mice by conducting Morris water maze (MWM) behaviour testing. Active components and their putative targets, as well as TDACI-related targets, were collected from public databases. Protein-protein interactions (PPIs), Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses and molecular docking were then utilized to explore potential molecular network mechanisms. Finally, the main targets were verified in animal model experiments. RESULTS MWM test showed that JLD improved aspects of behaviour in T2DM model mice. JLD improved glucose intolerance, tissue insulin sensitivity, lipid metabolism and enhanced synapse-associated protein expression in hippocampus tissue. Network pharmacology revealed 185 active components, 337 targets of JLD, and 7998 TDACI related targets were obtained . PPI network analyses revealed 39 core targets. GO and KEGG analyses suggested that JLD might improve TDACI by regulating gene expression, apoptotic processes and inflammatory responses mainly via PI3K-AKT and AGE-RAGE signaling pathways. Molecular docking revealed strong binding of the main components to core targets. JLD reduced hippocampus tissue expression of the inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL6), core targets of treatment of TDACI. CONCLUSIONS The findings suggested that JLD has the potential to improve TDACI through multiple components, multiple targets and multiple pathways. JLD may be a promising treatment for diabetic cognitive impairment.
Collapse
Affiliation(s)
- Haiyan Gu
- Department of Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
- Department of Shijiazhuang Technology Innovation Center of Precision Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
| | - Yuxin Zhang
- Department of Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
- Department of Shijiazhuang Technology Innovation Center of Precision Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
| | - Jinghua Sun
- Department of Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
- Department of Shijiazhuang Technology Innovation Center of Precision Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
| | - Lipeng Liu
- Department of Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
- Department of Shijiazhuang Technology Innovation Center of Precision Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
| | - Zanchao Liu
- Department of Hebei Provincial Key Laboratory of Basic Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
- Department of Shijiazhuang Technology Innovation Center of Precision Medicine for Diabetes, The Shijiazhuang Second Hospital, Shijiazhuang, China
| |
Collapse
|
2
|
Yang M, Bai M, Zhuang Y, Lu S, Ge Q, Li H, Deng Y, Wu H, Xu X, Niu F, Dong X, Zhang B, Liu B. High-dose dexamethasone regulates microglial polarization via the GR/JAK1/STAT3 signaling pathway after traumatic brain injury. Neural Regen Res 2025; 20:2611-2623. [PMID: 39314167 PMCID: PMC11801282 DOI: 10.4103/nrr.nrr-d-23-01772] [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: 10/29/2023] [Revised: 02/18/2024] [Accepted: 08/22/2024] [Indexed: 09/25/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202509000-00023/figure1/v/2024-11-05T132919Z/r/image-tiff Although microglial polarization and neuroinflammation are crucial cellular responses after traumatic brain injury, the fundamental regulatory and functional mechanisms remain insufficiently understood. As potent anti-inflammatory agents, the use of glucocorticoids in traumatic brain injury is still controversial, and their regulatory effects on microglial polarization are not yet known. In the present study, we sought to determine whether exacerbation of traumatic brain injury caused by high-dose dexamethasone is related to its regulatory effects on microglial polarization and its mechanisms of action. In vitro cultured BV2 cells and primary microglia and a controlled cortical impact mouse model were used to investigate the effects of dexamethasone on microglial polarization. Lipopolysaccharide, dexamethasone, RU486 (a glucocorticoid receptor antagonist), and ruxolitinib (a Janus kinase 1 antagonist) were administered. RNA-sequencing data obtained from a C57BL/6 mouse model of traumatic brain injury were used to identify potential targets of dexamethasone. The Morris water maze, quantitative reverse transcription-polymerase chain reaction, western blotting, immunofluorescence and confocal microscopy analysis, and TUNEL, Nissl, and Golgi staining were performed to investigate our hypothesis. High-throughput sequencing results showed that arginase 1, a marker of M2 microglia, was significantly downregulated in the dexamethasone group compared with the traumatic brain injury group at 3 days post-traumatic brain injury. Thus dexamethasone inhibited M1 and M2 microglia, with a more pronounced inhibitory effect on M2 microglia in vitro and in vivo . Glucocorticoid receptor plays an indispensable role in microglial polarization after dexamethasone treatment following traumatic brain injury. Additionally, glucocorticoid receptor activation increased the number of apoptotic cells and neuronal death, and also decreased the density of dendritic spines. A possible downstream receptor signaling mechanism is the GR/JAK1/STAT3 pathway. Overactivation of glucocorticoid receptor by high-dose dexamethasone reduced the expression of M2 microglia, which plays an anti-inflammatory role. In contrast, inhibiting the activation of glucocorticoid receptor reduced the number of apoptotic glia and neurons and decreased the loss of dendritic spines after traumatic brain injury. Dexamethasone may exert its neurotoxic effects by inhibiting M2 microglia through the GR/JAK1/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Mengshi Yang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Miao Bai
- Department of Neurology, The First Hospital of Tsinghua University, Beijing, China
| | - Yuan Zhuang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shenghua Lu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qianqian Ge
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hao Li
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yu Deng
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hongbin Wu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaojian Xu
- Department of Neurotrauma and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Fei Niu
- Department of Neurotrauma and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xinlong Dong
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurotrauma and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Bin Zhang
- Department of Critical Care Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Baiyun Liu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurotrauma and Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Center for Nerve Injury and Repair, Beijing Institute of Brain Disorders, China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
3
|
Henningfield CM, Ngo M, Murray KM, Kwang NE, Tsourmas KI, Neumann J, Pashkutz ZA, Kawauchi S, Swarup V, Lane TE, MacGregor GR, Green KN. Generation of an Inducible Destabilized-Domain Cre Mouse Line to Target Disease Associated Microglia. Glia 2025; 73:1272-1287. [PMID: 39988890 DOI: 10.1002/glia.70004] [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: 09/24/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025]
Abstract
The function of microglia during progression of Alzheimer's disease (AD) can be investigated using mouse models that enable genetic manipulation of microglial subpopulations in a temporal manner. We developed mouse lines that express either Cre recombinase (Cre) for constitutive targeting, or destabilized-domain Cre recombinase (DD-Cre) for inducible targeting from the Cst7 locus (Cst7 DD-Cre) to specifically manipulate disease associated microglia (DAM) and crossed with Ai14 tdTomato cre-reporter line mice. Cst7Cre was found to target all brain resident myeloid cells, due to transient developmental expression of Cst7, but no expression was found in the inducible Cst7 DD-Cre mice. Further crossing of this line with 5xFAD mice combined with dietary administration of trimethoprim to induce DD-Cre activity produces long-term labeling in DAM without evidence of leakiness, with tdTomato-expression restricted to cells surrounding plaques. Using this model, we found that DAMs are a subset of plaque-associated microglia (PAMs) and their transition to DAM increases with age and disease stage. Spatial transcriptomic analysis revealed that tdTomato+ cells show higher expression of disease and inflammatory genes compared to other microglial populations, including non-labeled PAMs. These models allow either complete cre-loxP targeting of all brain myeloid cells (Cst7Cre), or inducible targeting of DAMs, without leakiness (Cst7 DD-Cre).
Collapse
Affiliation(s)
- Caden M Henningfield
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Minh Ngo
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Kaitlin M Murray
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Nellie E Kwang
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Kate I Tsourmas
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Jonathan Neumann
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
| | - Zachary A Pashkutz
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
| | - Shimako Kawauchi
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Thomas E Lane
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
- Center for Virus Research, University of California, Irvine, USA
| | - Grant R MacGregor
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
- Department of Developmental and Cell Biology, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Kim N Green
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| |
Collapse
|
4
|
Sharma P, Daksh R, Khanna S, Mudgal J, Lewis SA, Arora D, Nampoothiri M. Microglial cannabinoid receptor 2 and epigenetic regulation: Implications for the treatment of depression. Eur J Pharmacol 2025; 995:177422. [PMID: 39988094 DOI: 10.1016/j.ejphar.2025.177422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/20/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Depression, often stress-induced, is closely related to neuroinflammation, in which microglia, the brain's immune cells, are the leading players. Microglia shift between a quiescent and an active state, promoting both pro- and anti-inflammatory responses. Cannabinoid type 2 (CB2) receptor encoded by the CNR2 gene is a key player to modulate inflammatory activity. CB2 receptor is highly controlled at the epigenetic level, especially in response to stressful stimuli, positioning it between stress, neuroinflammation, and depression. The following review addresses how epigenetic regulation of CNR2 expression affects depression and the dissection, further, of molecular pathways driving neuroinflammation-related depressive states. The present study emphasizes the therapeutic potential of CB2 receptor agonists that selectively interact with activated microglia and opens a new avenue for the treatment of depression associated with neuroinflammation. The review, therefore, provides a framework of underlying mechanisms for developing novel therapeutic strategies that focus on relieving symptoms by modulating the neuroinflammatory response. Finally, this review underlines the possibilities of therapeutic interventions taking into account CB2 receptors in combating depression.
Collapse
Affiliation(s)
- Pratyasha Sharma
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Rajni Daksh
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Saumya Khanna
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Shaila A Lewis
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Devinder Arora
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, 4222, Australia
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
5
|
Augusto-Oliveira M, Arrifano GDP, Leal-Nazaré CG, Chaves-Filho A, Santos-Sacramento L, Lopes-Araujo A, Tremblay MÈ, Crespo-Lopez ME. Morphological diversity of microglia: Implications for learning, environmental adaptation, ageing, sex differences and neuropathology. Neurosci Biobehav Rev 2025; 172:106091. [PMID: 40049541 DOI: 10.1016/j.neubiorev.2025.106091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/21/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
Microglia are the brain resident macrophages that respond rapidly to any insult. These non-neuroectodermal cells are decorated with plenty of receptors allowing them to recognise and respond precisely to a multitude of stimuli. To do so, microglia undergo structural and functional changes aiming to actively keep the brain's homeostasis. However, some microglial responses, when sustained or exacerbated, can contribute to neuropathology and neurodegeneration. Many microglial molecular and cellular changes were identified that display a strong correlation with neuronal damage and neuroinflammation/disease status, as well as present key sex-related differences that modulate microglial outcomes. Nevertheless, the relationship between microglial structural and functional features is just beginning to be unravelled. Several reports show that microglia undergo soma and branch remodelling in response to environmental stimuli, ageing, neurodegenerative diseases, trauma, and systemic inflammation, suggesting a complex form and function link. Also, it is reasonable overall to suppose that microglia diminishing their process length and ramification also reduce their monitoring activity of synapses, which is critical for detecting any synaptic disturbance and performing synaptic remodelling. Elucidating the complex interactions between microglial morphological plasticity and its functional implications appears essential for the understanding of complex cognitive and behavioural processes in health and neuropathological conditions.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER).
| | - Gabriela de Paula Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Caio Gustavo Leal-Nazaré
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Adriano Chaves-Filho
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, British Columbia, Canada; Women's Health Research Institute, British Columbia, Canada
| | - Leticia Santos-Sacramento
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Amanda Lopes-Araujo
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada; Department of Molecular Medicine, Université Laval, Québec, Qubec, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, Québec, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, British Columbia, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, British Columbia, Canada; Women's Health Research Institute, British Columbia, Canada; College Member of the Royal Society of Canada, Canada.
| | - Maria Elena Crespo-Lopez
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER).
| |
Collapse
|
6
|
Zheng Y, Ren Z, Liu Y, Yan J, Chen C, He Y, Shi Y, Cheng F, Wang Q, Li C, Wang X. T cell interactions with microglia in immune-inflammatory processes of ischemic stroke. Neural Regen Res 2025; 20:1277-1292. [PMID: 39075894 PMCID: PMC11624874 DOI: 10.4103/nrr.nrr-d-23-01385] [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: 08/19/2023] [Revised: 01/17/2024] [Accepted: 03/07/2024] [Indexed: 07/31/2024] Open
Abstract
The primary mechanism of secondary injury after cerebral ischemia may be the brain inflammation that emerges after an ischemic stroke, which promotes neuronal death and inhibits nerve tissue regeneration. As the first immune cells to be activated after an ischemic stroke, microglia play an important immunomodulatory role in the progression of the condition. After an ischemic stroke, peripheral blood immune cells (mainly T cells) are recruited to the central nervous system by chemokines secreted by immune cells in the brain, where they interact with central nervous system cells (mainly microglia) to trigger a secondary neuroimmune response. This review summarizes the interactions between T cells and microglia in the immune-inflammatory processes of ischemic stroke. We found that, during ischemic stroke, T cells and microglia demonstrate a more pronounced synergistic effect. Th1, Th17, and M1 microglia can co-secrete pro-inflammatory factors, such as interferon-γ, tumor necrosis factor-α, and interleukin-1β, to promote neuroinflammation and exacerbate brain injury. Th2, Treg, and M2 microglia jointly secrete anti-inflammatory factors, such as interleukin-4, interleukin-10, and transforming growth factor-β, to inhibit the progression of neuroinflammation, as well as growth factors such as brain-derived neurotrophic factor to promote nerve regeneration and repair brain injury. Immune interactions between microglia and T cells influence the direction of the subsequent neuroinflammation, which in turn determines the prognosis of ischemic stroke patients. Clinical trials have been conducted on the ways to modulate the interactions between T cells and microglia toward anti-inflammatory communication using the immunosuppressant fingolimod or overdosing with Treg cells to promote neural tissue repair and reduce the damage caused by ischemic stroke. However, such studies have been relatively infrequent, and clinical experience is still insufficient. In summary, in ischemic stroke, T cell subsets and activated microglia act synergistically to regulate inflammatory progression, mainly by secreting inflammatory factors. In the future, a key research direction for ischemic stroke treatment could be rooted in the enhancement of anti-inflammatory factor secretion by promoting the generation of Th2 and Treg cells, along with the activation of M2-type microglia. These approaches may alleviate neuroinflammation and facilitate the repair of neural tissues.
Collapse
Affiliation(s)
- Yuxiao Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zilin Ren
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Juntang Yan
- Library, Beijing University of Chinese Medicine, Beijing, China
| | - Congai Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yanhui He
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuyu Shi
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Fafeng Cheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qingguo Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Changxiang Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xueqian Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
7
|
Strackeljan L, Baidoe-Ansah D, Mirzapourdelavar H, Jia S, Kaushik R, Cangalaya C, Dityatev A. Partial microglial depletion through inhibition of colony-stimulating factor 1 receptor improves synaptic plasticity and cognitive performance in aged mice. Exp Neurol 2025; 387:115186. [PMID: 39956381 DOI: 10.1016/j.expneurol.2025.115186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Microglia depletion, followed by repopulation, improves cognitive functions in the aged mouse brain. However, even temporal ablation of microglia puts the brain at a high risk of infection. Hence, in the present work, we studied if the partial reduction of microglia with PLX3397 (pexidartinib), an inhibitor of the colony-stimulating factor 1 receptor (CSF1R), could bring similar benefits as reported for microglia ablation. Aged (two-years-old) mice were treated with PLX3397 for a total of 6 weeks, which reduced microglia numbers in the hippocampus and retrosplenial cortex (RSC) to the levels seen in young mice and resulted in layer-specific ablation in the expression of microglial complement protein C1q mediating synaptic remodeling. This treatment boosted long-term potentiation in the CA1 region and improved performance in the hippocampus-dependent novel object location recognition task. Although PLX3397 treatment did not alter the number or total intensity of Wisteria floribunda agglutinin-positive perineuronal nets (PNNs) in the CA1 region of the hippocampus, it changed the fine structure of PNNs. It also elevated the expression of perisynaptic proteoglycan brevican, presynaptic vGluT1 at excitatory synapses, and vGAT in inhibitory ones in the CA1 stratum radiatum. Thus, targeting the CSF1R may provide a safe and efficient strategy to boost synaptic and cognitive functions in the aged brain.
Collapse
Affiliation(s)
- Luisa Strackeljan
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - David Baidoe-Ansah
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Hadi Mirzapourdelavar
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Shaobo Jia
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Rahul Kaushik
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Carla Cangalaya
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Alexander Dityatev
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany; Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany.
| |
Collapse
|
8
|
Zimmermann S, Mathew A, Bondareva O, Elwakiel A, Jiang S, Rana R, Bechmann I, Goldschmidt J, Klöting N, Sheikh BN, Isermann B. Noncanonical microglial IL-1β maturation in chronic kidney disease. Nephrol Dial Transplant 2025; 40:929-942. [PMID: 39496522 DOI: 10.1093/ndt/gfae239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Organ transplantation reverses cognitive impairment in chronic kidney disease (CKD), indicating that cognitive impairment driven by CKD is therapeutically amendable. We recently demonstrated that impaired cognition in CKD is linked to interleukin-1β (IL-1β) release from microglia and IL-1 receptor type 1 signalling in neuronal cells, thereby identifying a signalling pathway that can be exploited therapeutically. However, the mechanism of IL-1β maturation in microglia in CKD remains unknown. We hypothesized that microglia cells require caspase-1 for CKD-driven cognitive impairment. METHODS We used a combination of single-cell analyses, in situ analyses, genetically modified mouse models (including newly generated Cre-LoxP mouse models) and in vitro models. The current study builds on a recently identified intercellular cross-talk between microglia and neurons that impairs cognition in CKD. RESULTS Here we show that despite NLRP3 inflammasome activation in the brain and protection of mice with constitutive NLRP3 deficiency from CKD-induced cognitive impairment, caspase-1 is not required for IL-1β maturation in microglia and targeted caspase-1 deficiency in microglia does not improve cognition in CKD mice. These data indicate that IL-1β maturation in microglia is independent of the NLRP3-caspase-1 interaction in CKD. Indeed, microglia activation in CKD induces noncanonical, cathepsin C-caspase-8-mediated IL-1β maturation. Depletion of cathepsin C or caspase-8 blocks IL-1β maturation in microglia. Preliminary analyses suggest that noncanonical microglia IL-1β maturation occurs also in diabetes mellitus. CONCLUSION These results identify a noncanonical IL-1β-maturation pathway as a potential therapeutic target to combat microglia-induced neuronal dysfunction in CKD and possibly other peripheral diseases.
Collapse
Affiliation(s)
- Silke Zimmermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, Leipzig, Germany
| | - Akash Mathew
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, Leipzig, Germany
| | - Olga Bondareva
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (Hl-MAG) of the Helmholtz Center Munich, Leipzig, Germany
| | - Ahmed Elwakiel
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, Leipzig, Germany
| | - Shihai Jiang
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, Leipzig, Germany
| | - Rajiv Rana
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, Leipzig, Germany
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | | | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (Hl-MAG) of the Helmholtz Center Munich, Leipzig, Germany
| | - Bilal N Sheikh
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (Hl-MAG) of the Helmholtz Center Munich, Leipzig, Germany
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, Leipzig, Germany
| |
Collapse
|
9
|
Damiani F, Giuliano MG, Cornuti S, Putignano E, Tognozzi A, Suckow V, Kalscheuer VM, Pizzorusso T, Tognini P. Multi-site investigation of gut microbiota in CDKL5 deficiency disorder mouse models: Targeting dysbiosis to improve neurological outcomes. Cell Rep 2025; 44:115546. [PMID: 40220293 PMCID: PMC12014524 DOI: 10.1016/j.celrep.2025.115546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/31/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Cyclin-dependent kinase-like 5 (CDKL5) deficiency disorder (CDD) is a rare neurodevelopmental disorder often associated with gastrointestinal (GI) issues and subclinical immune dysregulation, suggesting a link to the gut microbiota. We analyze the fecal microbiota composition in two CDKL5 knockout (KO) mouse models at postnatal days (P) 25, 32 (youth), and 70 (adulthood), revealing significant microbial imbalances, particularly during juvenile stages. To investigate the role of the intestinal microbiota in CDD and assess causality, we administer antibiotics, which lead to improved visual cortical responses and reduce hyperactivity. Additionally, microglia morphology changes, indicative of altered surveillance and activation states, are reversed. Strikingly, fecal transplantation from CDKL5 KO to wild-type (WT) recipient mice successfully transfers both visual response deficits and hyperactive behavior. These findings show that gut microbiota alterations contribute to the severity of neurological symptoms in CDD, shedding light on the interplay between microbiota, microglia, and neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Francesca Damiani
- Laboratory of Biology BIO@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Maria Grazia Giuliano
- Laboratory of Biology BIO@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy; Health Science Interdisciplinary Center, Sant'Anna School of Advanced Studies, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
| | - Sara Cornuti
- Laboratory of Biology BIO@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Elena Putignano
- Institute of Neuroscience, National Research Council, Via G. Moruzzi 1, 56124 Pisa, Italy
| | - Andrea Tognozzi
- Laboratory of Biology BIO@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy; PhD Program in Clinical and Translational Science, University of Pisa, Via Savi 10, 56126 Pisa, Italy
| | - Vanessa Suckow
- Max Planck Institute for Molecular Genetics, Ihnestraße 63, 14195 Berlin, Germany
| | - Vera M Kalscheuer
- Max Planck Institute for Molecular Genetics, Ihnestraße 63, 14195 Berlin, Germany
| | - Tommaso Pizzorusso
- Laboratory of Biology BIO@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy; Institute of Neuroscience, National Research Council, Via G. Moruzzi 1, 56124 Pisa, Italy
| | - Paola Tognini
- Health Science Interdisciplinary Center, Sant'Anna School of Advanced Studies, Piazza Martiri della Libertà 33, 56127 Pisa, Italy.
| |
Collapse
|
10
|
Menarchek BJ, Bridi MCD. Latent mechanisms of plasticity are upregulated during sleep. Curr Opin Neurobiol 2025; 93:103029. [PMID: 40267630 DOI: 10.1016/j.conb.2025.103029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/25/2025]
Abstract
Sleep is thought to serve an important role in learning and memory, but the mechanisms by which sleep promotes plasticity remain unclear. Even in the absence of plastic changes in neuronal function, many molecular, cellular, and physiological processes linked to plasticity are upregulated during sleep. Therefore, sleep may be a state in which latent plasticity mechanisms are poised to respond following novel experiences during prior wake. Many of these plasticity-related processes can promote both synaptic strengthening and weakening. Signaling pathways activated during sleep may interact with complements of proteins, determined by the content of prior waking experience, to establish the polarity of plasticity. Furthermore, precise reactivation of neuronal spiking patterns during sleep may interact with ongoing neuromodulatory, dendritic, and network activity to strengthen and weaken synapses. In this review, we will discuss the idea that sleep elevates latent plasticity mechanisms, which drive bidirectional plasticity depending on prior waking experience.
Collapse
Affiliation(s)
- Benjamin J Menarchek
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA
| | - Michelle C D Bridi
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA.
| |
Collapse
|
11
|
Malhotra S, Donneger F, Farrell JS, Dudok B, Losonczy A, Soltesz I. Integrating endocannabinoid signaling, CCK interneurons, and hippocampal circuit dynamics in behaving animals. Neuron 2025:S0896-6273(25)00188-6. [PMID: 40267911 DOI: 10.1016/j.neuron.2025.03.016] [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: 07/12/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 04/25/2025]
Abstract
The brain's endocannabinoid signaling system modulates a diverse range of physiological phenomena and is also involved in various psychiatric and neurological disorders. The basic components of the molecular machinery underlying endocannabinoid-mediated synaptic signaling have been known for decades. However, limitations associated with the short-lived nature of endocannabinoid lipid signals had made it challenging to determine the spatiotemporal specificity and dynamics of endocannabinoid signaling in vivo. Here, we discuss how novel technologies have recently enabled unprecedented insights into endocannabinoid signaling taking place at specific synapses in behaving animals. In this review, we primarily focus on cannabinoid-sensitive inhibition in the hippocampus in relation to place cell properties to illustrate the potential of these novel methodologies. In addition, we highlight implications of these approaches and insights for the unraveling of cannabinoid regulation of synapses in vivo in other brain circuits in both health and disease.
Collapse
Affiliation(s)
- Shreya Malhotra
- Department of Neurosurgery, Stanford University, Stanford, CA, USA.
| | - Florian Donneger
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Jordan S Farrell
- Department of Neurology, Harvard Medical School, Boston, MA, USA; Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA; F.M. Kirby Neurobiology Center, Harvard Medical School, Boston, MA, USA
| | - Barna Dudok
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, NY, USA; Kavli Institute for Brain Sciences, Columbia University, New York, NY, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| |
Collapse
|
12
|
Wang C, He T, Qin J, Jiao J, Ji F. The roles of immune factors in neurodevelopment. Front Cell Neurosci 2025; 19:1451889. [PMID: 40276707 PMCID: PMC12018394 DOI: 10.3389/fncel.2025.1451889] [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: 06/20/2024] [Accepted: 03/28/2025] [Indexed: 04/26/2025] Open
Abstract
The development of the nervous system is a highly complex process orchestrated by a multitude of factors, including various immune elements. These immune components play a dual role, not only regulating the immune response but also actively influencing brain development under both physiological and pathological conditions. The brain's immune barrier includes microglia in the brain parenchyma, which act as resident macrophages, astrocytes that support neuronal function and contribute to the inflammatory response, as well as circulating immune cells that reside at the brain's borders, including the choroid plexus, meninges, and perivascular spaces. Cytokines-soluble signaling molecules released by immune cells-play a crucial role in mediating communication between immune cells and the developing nervous system. Cytokines regulate processes such as neurogenesis, synaptic pruning, and inflammation, helping to shape the neural environment. Dysregulation of these immune cells, astrocytes, or cytokine signaling can lead to alterations in neurodevelopment, potentially contributing to neurodevelopmental abnormalities. This article reviews the central role of microglia, astrocytes, cytokines, and other immune factors in neurodevelopment, and explores how neuroinflammation can lead to the onset of neurodevelopmental disorders, shedding new light on their pathogenesis.
Collapse
Affiliation(s)
- Chong Wang
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Tingting He
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jie Qin
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jianwei Jiao
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Fen Ji
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| |
Collapse
|
13
|
Shimamura T, Kitashiba M, Nishizawa K, Hattori Y. Physiological roles of embryonic microglia and their perturbation by maternal inflammation. Front Cell Neurosci 2025; 19:1552241. [PMID: 40260079 PMCID: PMC12009865 DOI: 10.3389/fncel.2025.1552241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
The interplay between the nervous and immune systems is well documented in the context of adult physiology and disease. Recent advances in understanding immune cell development have highlighted a significant interaction between neural lineage cells and microglia, the resident brain macrophages, during developmental stages. Throughout development, particularly from the embryonic to postnatal stages, diverse neural lineage cells are sequentially generated, undergo fate determination, migrate dynamically to their appropriate locations while maturing, and establish connections with their surroundings to form neural circuits. Previous studies have demonstrated that microglia contribute to this highly orchestrated process, ensuring the proper organization of brain structure. These findings underscore the need to further investigate how microglia behave and function within a broader framework of neurodevelopment. Importantly, recent epidemiological studies have suggested that maternal immune activation (MIA), triggered by various factors, such as viral or bacterial infections, environmental stressors, or other external influences, can affect neurogenesis and neural circuit formation, increasing the risk of neurodevelopmental disorders (NDDs) in offspring. Notably, many studies have revealed that fetal microglia undergo significant changes in response to MIA. Given their essential roles in neurogenesis and vascular development, inappropriate activation or disruption of microglial function may impair these critical processes, potentially leading to abnormal neurodevelopment. This review highlights recent advances in rodent models and human studies that have shed light on the behaviors and multifaceted roles of microglia during brain development, with a particular focus on the embryonic stage. Furthermore, drawing on insights from rodent MIA models, this review explores how MIA disrupts microglial function and how such disturbances may impair brain development, ultimately contributing to the onset of NDDs.
Collapse
Affiliation(s)
| | | | | | - Yuki Hattori
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
14
|
Vicario R, Fragkogianni S, Pokrovskii M, Meyer C, Lopez-Rodrigo E, Hu Y, Ogishi M, Alberdi A, Baako A, Ay O, Plu I, Sazdovitch V, Heritier S, Cohen-Aubart F, Shor N, Miyara M, Nguyen-Khac F, Viale A, Idbaih A, Amoura Z, Rosenblum MK, Zhang H, Karnoub ER, Sashittal P, Jakatdar A, Iacobuzio-Donahue CA, Abdel-Wahab O, Tabar V, Socci ND, Elemento O, Diamond EL, Boisson B, Casanova JL, Seilhean D, Haroche J, Donadieu J, Geissmann F. Role of clonal inflammatory microglia in histiocytosis-associated neurodegeneration. Neuron 2025; 113:1065-1081.e13. [PMID: 40081365 DOI: 10.1016/j.neuron.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/28/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025]
Abstract
Langerhans cell histiocytosis (LCH) and Erdheim-Chester disease (ECD) are clonal myeloid disorders associated with mitogen-activated protein (MAP)-kinase-activating mutations and an increased risk of neurodegeneration. We found microglial mutant clones in LCH and ECD patients, whether or not they presented with clinical symptoms of neurodegeneration, associated with microgliosis, astrocytosis, and neuronal loss, predominantly in the rhombencephalon gray nuclei. Neurological symptoms were associated with PU.1+ clone size (p = 0.0003) in patients with the longest evolution of the disease, indicating a phase of subclinical incipient neurodegeneration. Genetic barcoding analysis suggests that clones may originate from definitive or yolk sac hematopoiesis, depending on the patients. In a mouse model, disease topography was attributable to a local clonal proliferative advantage, and microglia depletion by a CSF1R-inhibitor limited neuronal loss and improved survival. These studies characterize a neurodegenerative disease associated with clonal proliferation of inflammatory microglia. The long preclinical stage represents a therapeutic window before irreversible neuronal depletion.
Collapse
Affiliation(s)
- Rocio Vicario
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stamatina Fragkogianni
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maria Pokrovskii
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Carina Meyer
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Estibaliz Lopez-Rodrigo
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yang Hu
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell, New York, NY 10021, USA
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Araitz Alberdi
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ann Baako
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Oyku Ay
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Isabelle Plu
- Department of Neuropathology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Véronique Sazdovitch
- Department of Neuropathology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Sebastien Heritier
- French Langerhans cell histiocytosis registry, Department of Pediatric Hematology and Oncology, Trousseau Hospital, AP-HP, Paris, France
| | - Fleur Cohen-Aubart
- Department of Internal Medicine & Institut E3M, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Natalia Shor
- Department of Neuroradiology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Makoto Miyara
- Center for Immunology and Infectious Diseases (CIMI-PARIS), Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Florence Nguyen-Khac
- Department of Hematology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Agnes Viale
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, MSKCC, New York, NY 10065, USA
| | - Ahmed Idbaih
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié-Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, 75013 Paris, France
| | - Zahir Amoura
- Department of Neuroradiology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | | | - Haochen Zhang
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | | | - Palash Sashittal
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Akhil Jakatdar
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Christine A Iacobuzio-Donahue
- Department of Pathology, MSKCC, New York, NY 10065, USA; Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Viviane Tabar
- Department of Neurosurgery, and Center for Stem Cell Biology, MSKCC, New York, NY, USA; Department of Neurology, MSKCC, New York, NY 10065, USA
| | - Nicholas D Socci
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, MSKCC, New York, NY 10065, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell, New York, NY 10021, USA
| | - Eli L Diamond
- Department of Neurosurgery, and Center for Stem Cell Biology, MSKCC, New York, NY, USA
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Danielle Seilhean
- Department of Neuropathology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Julien Haroche
- Department of Internal Medicine & Institut E3M, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France.
| | - Jean Donadieu
- French Langerhans cell histiocytosis registry, Department of Pediatric Hematology and Oncology, Trousseau Hospital, AP-HP, Paris, France
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
15
|
Lawrence AB, Brown SM, Bradford BM, Mabbott NA, Bombail V, Rutherford KMD. Non-neuronal brain biology and its relevance to animal welfare. Neurosci Biobehav Rev 2025; 173:106136. [PMID: 40185375 DOI: 10.1016/j.neubiorev.2025.106136] [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: 07/10/2024] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Non-neuronal cells constitute a significant portion of brain tissue and are seen as having key roles in brain homeostasis and responses to challenges. This review illustrates how non-neuronal biology can bring new perspectives to animal welfare through understanding mechanisms that determine welfare outcomes and highlighting interventions to improve welfare. Most obvious in this respect is the largely unrecognised relevance of neuroinflammation to animal welfare which is increasingly found to have roles in determining how animals respond to challenges. We start by introducing non-neuronal cells and review their involvement in affective states and cognition often seen as core psychological elements of animal welfare. We find that the evidence for a causal involvement of glia in cognition is currently more advanced than the corresponding evidence for affective states. We propose that translational research on affective disorders could usefully apply welfare science derived approaches for assessing affective states. Using evidence from translational research, we illustrate the involvement of non-neuronal cells and neuroinflammatory processes as mechanisms modulating resilience to welfare challenges including disease, pain, and social stress. We review research on impoverished environments and environmental enrichment which suggests that environmental conditions which improve animal welfare also improve resilience to challenges through balancing pro- and anti-inflammatory non-neuronal processes. We speculate that non-neuronal biology has relevance to animal welfare beyond neuro-inflammation including facilitating positive affective states. We acknowledge the relevance of neuronal biology to animal welfare whilst proposing that non-neuronal biology provides additional and relevant insights to improve animals' lives.
Collapse
Affiliation(s)
- Alistair B Lawrence
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK; Scotland's Rural College (SRUC), Edinburgh EH9 3JG, UK.
| | - Sarah M Brown
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Barry M Bradford
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Neil A Mabbott
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | | | | |
Collapse
|
16
|
Zhang P, Cheng RJ, Yang QL, Gong Y, Xu Y, Chen LM, Zhou L, Jiang CL. Mitophagy impairment drives microglia activation and results in cognitive deficits in neonatal mice following sevoflurane exposure. Toxicol Lett 2025; 406:20-30. [PMID: 39955081 DOI: 10.1016/j.toxlet.2025.02.008] [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: 06/18/2024] [Revised: 02/02/2025] [Accepted: 02/13/2025] [Indexed: 02/17/2025]
Abstract
Sevoflurane exposure induces cognitive deficits in neonatal mice. Mitophagy was closely correlated to sevoflurane inhalation induced neurotoxicity in developing brains. However, the underlying mechanisms have not been fully elucidated. In this study, we attempted to clarify the role of mitophagy in neonatal mice undergoing sevoflurane exposure. BV2 microglial cells were cultured, and mcherry-EGFP-LC3B adenovirus were transfected. The results showed that the fluorescence intensity of GFP was markedly increased after sevoflurane exposure, and rapamycin administration could mitigate this effect. The mitophagy flux test showed that sevoflurane exposure reduced the degree of colocalization between Mito-Traker and Lyso-Traker fluorescent, while which was elevated by rapamycin treatment. The immunofluorescence assay suggested that sevoflurane inhalation resulted in the significant decrease of autolysosome formation, which was sharply enhanced in SEV group after rapamycin treatment. Meanwhile, sevoflurane inhalation shifted microglial M1/M2 phenotypic polarization, and rapamycin administration reversed this status. Moreover, the degree of colocalization among Iba-1, Synaptophysin (Syn) and lysosomal-associated membrane protein 1 (Lamp1) was increased after sevoflurane exposure, and that was reduced following rapamycin treatment. The behavioral performance was worse after sevoflurane inhalation in neonatal mice, and rapamycin treatment effectively improved the cognitive outcome. Collectively, these findings demonstrated that mitophagy impairment induced by sevoflurane exposure promoted microglia M1 phenotypic polarization and enlarged phagocytosis, and resulted in cognitive deficits, while rapamycin administration effectively reversed this tendency.
Collapse
Affiliation(s)
- Piao Zhang
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Rui-Juan Cheng
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qiao-Ling Yang
- Reproductive Medicine Center, SiChuan Provincial Maternity and Child Health Care Hospital,the Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan 610045, China
| | - Yan Gong
- Reproductive Medicine Center, SiChuan Provincial Maternity and Child Health Care Hospital,the Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan 610045, China
| | - Yan Xu
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ling-Min Chen
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Li Zhou
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Chun-Ling Jiang
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
17
|
Nazari S, Niknamfar S, Ghazvini H, Rafaiee R, Allahverdy A, Khazaie H, Tamijani S. Transcutaneous Auricular Vagus Nerve Stimulation Restores Cognitive Impairment in Morphine-Withdrawn Rats: Role of BDNF and Glial Cells in the Hippocampus. Addict Biol 2025; 30:e70016. [PMID: 40183201 PMCID: PMC11969173 DOI: 10.1111/adb.70016] [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: 03/24/2024] [Revised: 10/29/2024] [Accepted: 01/07/2025] [Indexed: 04/05/2025]
Abstract
Opioid use disorder (OUD) is a significant mental health problem, with prolonged usage potentially resulting in tolerance, addiction and cognitive decline, including learning and memory deficiency. At present, pharmacotherapy serves as the primary treatment approach for OUD. However, despite its status as a cornerstone of treatment, pharmacotherapy has certain limitations, thereby mandating the exploration of alternative modalities. This study evaluated the efficacy of transcutaneous auricular vagus nerve stimulation (taVNS) in multiple cognitive domains in morphine-withdrawn rats. To induce morphine dependence, the rats were administered 10 mg/kg morphine for 10 consecutive days. taVNS was administered to the left ear of each rat and continued for 2 weeks. After electrical stimulation, various cognitive and emotional functions were assessed through related behavioural tasks, including open field, Y-maze, novel object recognition and elevated plus maze tests. GFAP, Iba1 and BDNF expression levels in the hippocampus were determined via quantitative polymerase chain reaction (qPCR). Our investigation revealed that taVNS ameliorated the impairment of working and recognition memory induced by morphine in behavioural tests. Additionally, it exerts an anxiolytic effect. Moreover, taVNS counteracted the decreased concentration of brain-derived neurotrophic factor (BDNF) and elevated levels of glial fibrillary acidic protein (GFAP) caused by morphine. Nonetheless, taVNS applied only at a frequency of 100 Hz has the potential to lower Iba1 levels independently of prior exposure to morphine. taVNS has been shown to exert a neuroprotective effect on morphine-withdrawn rats. This outcome indicates that taVNS can be employed as a supplementary therapy with other pharmacological interventions for OUD.
Collapse
Affiliation(s)
- Somayeh Nazari
- Student Research Committee, School of Advanced Technologies in MedicineMazandaran University of Medical SciencesSariIran
| | - Saba Niknamfar
- Student Research Committee, School of Advanced Technologies in MedicineMazandaran University of Medical SciencesSariIran
| | - Hamed Ghazvini
- Department of Neuroscience, School of Advanced Technologies in MedicineMazandaran University of Medical SciencesSariIran
- Psychiatry and Behavioral Sciences Research Center, Addiction InstituteMazandaran University of Medical SciencesSariIran
| | - Raheleh Rafaiee
- Department of Neuroscience, School of Advanced Technologies in MedicineMazandaran University of Medical SciencesSariIran
| | - Armin Allahverdy
- Neurodevelopmental Disorders Prevention CenterPerinatal Institute, Cincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Habibolah Khazaie
- Department of Psychiatry, Faculty of MedicineKermanshah University of Medical SciencesKermanshahIran
| | - Seyedeh Masoumeh Seyedhosseini Tamijani
- Department of Neuroscience, School of Advanced Technologies in MedicineMazandaran University of Medical SciencesSariIran
- Psychiatry and Behavioral Sciences Research Center, Addiction InstituteMazandaran University of Medical SciencesSariIran
| |
Collapse
|
18
|
Zhou B, Li Q, Su M, Liao P, Luo Y, Luo R, Yu Y, Luo M, Lei F, Li X, Jiao J, Yi L, Wang J, Yang L, Liao D, Zhou C, Zhang X, Xiao H, Zuo Y, Liu J, Zhu T, Jiang R. Astrocyte morphological remodeling regulates consciousness state transitions induced by inhaled general anesthesia. Mol Psychiatry 2025:10.1038/s41380-025-02978-2. [PMID: 40169801 DOI: 10.1038/s41380-025-02978-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 04/03/2025]
Abstract
General anesthetics (GAs) are conventionally thought to induce loss of consciousness (LOC) by acting on pre- and post-synaptic targets. However, the mechanism underlying the involvement of astrocytes in LOC remains unclear. Here we report that inhaled GAs cause reversible impairments in the fine processes of astrocytes within the somatosensory cortex, mediated by regulating the phosphorylation level of Ezrin, a protein critical for the fine morphology of astrocytes. Genetically deleting Ezrin or disrupting its phosphorylation was sufficient to decrease astrocyte-synapse interaction and enhance sensitivity to sevoflurane (Sevo) in vivo. Moreover, we show that disrupting astrocytic Ezrin phosphorylation boosted the inhibitory effect of Sevo on pyramidal neurons by enhancing tonic GABA and lowering excitability under anesthesia. Our work reveals previously unappreciated phosphorylation-dependent morphological dynamics, which enable astrocytes to regulate neuronal activity during the transition between two brain consciousness states.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qingran Li
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mengchan Su
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Pain Management, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ping Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuncheng Luo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rong Luo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunqing Yu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meiyan Luo
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fan Lei
- Institute of Brain Science and Diseases, West China Hospital, Sichuan University, Chengdu, 610213, China
| | - Xin Li
- Institute of Brain Science and Diseases, West China Hospital, Sichuan University, Chengdu, 610213, China
| | - Jiao Jiao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Limei Yi
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Wang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Linghui Yang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Daqing Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cheng Zhou
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xia Zhang
- Institute of Brain Science and Diseases, West China Hospital, Sichuan University, Chengdu, 610213, China
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong Xiao
- Department of Pain Management, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunxia Zuo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Ruotian Jiang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
19
|
Cui J, Wang XR, Yu J, Zhang BR, Shi YF, So KF, Zhang L, Wei JA. Neuropeptide-mediated activation of astrocytes improves stress resilience in mice by modulating cortical neural synapses. Acta Pharmacol Sin 2025; 46:867-879. [PMID: 39643639 PMCID: PMC11950203 DOI: 10.1038/s41401-024-01420-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/28/2024] [Indexed: 12/09/2024]
Abstract
Astrocytes are known to modulate synaptogenesis or neuronal activities, thus participating in mental functions. It has been shown that astrocytes are involved in the antidepressant mechanism. In this study we investigated the potential hormonal mediator governing the astrocyte-neuron interplay for stress-coping behaviors. Mice were subjected to chronic restraint stress (CRS) for 14 days, and then brain tissue was harvested for analyses. We found that the expression of pituitary adenylate cyclase activating polypeptide (PACAP) and its receptor PAC1 was significantly decreased in astrocytes of the prelimbic (PrL) cortex. By conducting a combination of genetics, in vivo imaging and behavioral assays we demonstrated that PAC1 in cortical astrocytes was necessary for maintaining normal resilience of mice against chronic environmental stress like restraint stress. Furthermore, we showed the enhancement of de novo cortical spine formation and synaptic activity under PACAP-mediated astrocytic activation possibly via the ATP release. The molecular mechanisms suggested that the vesicle homeostasis mediated by PACAP-PAC1 axis in astrocytes was involved in regulating synaptic functions. This study identifies a previously unrecognized route by which neuropeptide modulates cortical functions via local regulation of astrocytes.
Collapse
Affiliation(s)
- Jing Cui
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiao-Ran Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jie Yu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Bo-Rui Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Ya-Fei Shi
- College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Kwok-Fai So
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- State Key Laboratory of Brain and Cognitive Science, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, 266114, China
- Center for Exercise and Brain Science, School of Psychology, Shanghai University of Sport, Shanghai, 200438, China
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, China
| | - Li Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, 266114, China.
- Center for Exercise and Brain Science, School of Psychology, Shanghai University of Sport, Shanghai, 200438, China.
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, China.
| | - Ji-An Wei
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
20
|
Vear A, Heneka MT, Clemmensen C. Incretin-based therapeutics for the treatment of neurodegenerative diseases. Nat Metab 2025; 7:679-696. [PMID: 40211045 DOI: 10.1038/s42255-025-01263-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/06/2025] [Indexed: 04/12/2025]
Abstract
Neurodegenerative diseases (NDDs) represent a heterogeneous group of disorders characterized by progressive neuronal loss, which results in significant deficits in memory, cognition, motor skills, and sensory functions. As the prevalence of NDDs rises, there is an urgent unmet need for effective therapies. Current drug development approaches primarily target single pathological features of the disease, which could explain the limited efficacy observed in late-stage clinical trials. Originally developed for the treatment of obesity and diabetes, incretin-based therapies, particularly long-acting GLP-1 receptor (GLP-1R) agonists and GLP-1R-gastric inhibitory polypeptide receptor (GIPR) dual agonists, are emerging as promising treatments for NDDs. Despite limited conclusive preclinical evidence, their pleiotropic ability to reduce neuroinflammation, enhance neuronal energy metabolism and promote synaptic plasticity positions them as potential disease-modifying NDD interventions. In anticipation of results from larger clinical trials, continued advances in next-generation incretin mimetics offer the potential for improved brain access and enhanced neuroprotection, paving the way for incretin-based therapies as a future cornerstone in the management of NDDs.
Collapse
Affiliation(s)
- Anika Vear
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
21
|
Gao Y, Chen Y, Wang N, Meng Q. Albiflorin ameliorates neuroinflammation and exerts neuroprotective effects in Parkinson's disease models. Immunopharmacol Immunotoxicol 2025; 47:201-212. [PMID: 39924948 DOI: 10.1080/08923973.2025.2457960] [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: 03/25/2024] [Accepted: 01/18/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Albiflorin isolated from Paeoniae Alba Radix can cross the blood-brain barrier (BBB) and possesses analgesia, anticonvulsant, anti-inflammatory, and hepatoprotective properties. This study investigates albiflorin functions and related mechanisms in Parkinson's disease (PD) pathogenesis. METHODS Cellular and animal models of PD were constructed. Cell viability and apoptosis were detected by CCK-8 assays. Levels of Iba-1 and TH were measured by immunofluorescence staining, western blotting, and immunohistochemistry staining. Levels of pro-inflammatory mediators and pathway-related genes were measured by western blotting and RT-qPCR. Locomotor activity of mice was examined by open field test, rod climbing test, and rod rotating test. RESULTS For in vitro analysis, albiflorin inhibited LPS-induced microglial activation and neuroinflammation. Additionally, albiflorin inactivated NF-κB and MAPK pathways in LPS-treated BV2 cells. Moreover, albiflorin attenuated neurotoxicity mediated by LPS-stimulated microglia. For in vivo analysis, albiflorin improved MPTP-induced locomotor activity deficits and reduced MPTP-induced dopaminergic neuron loss. In parallel, albiflorin inhibited activated microglia-mediated neuroinflammation in MPTP-treated mice. CONCLUSION Albiflorin mitigates neuronal apoptosis and improves behavioral impairments in MPTP-induced PD mouse model through inhibition of activated microglia-mediated neuroinflammation via the NF-κB and MAPK pathways.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Neurology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yanmei Chen
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Ning Wang
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Qiang Meng
- Department of Neurology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
22
|
Hao K, Chen F, Xu S, Xiong Y, Xu R, Huang H, Shu C, Lv Y, Wang G, Wang H. Cognitive impairment following maternal separation in rats mediated by the NAD +/SIRT3 axis via modulation of hippocampal synaptic plasticity. Transl Psychiatry 2025; 15:112. [PMID: 40159484 PMCID: PMC11955552 DOI: 10.1038/s41398-025-03318-2] [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: 04/25/2024] [Revised: 01/16/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
Maternal separation (MS) during early life can induce behaviors in adult animals that resemble those seen in schizophrenia, manifesting cognitive deficits. These cognitive deficits may be indicative of oxidative stress linked to mitochondrial dysfunction. However, there is limited understanding of the molecular mechanisms regulating mitochondria in neural circuits that govern cognitive impairment relevant to schizophrenia, and their impact on neuronal structure and function. A 24-h MS rat model was utilized to simulate features associated with schizophrenia. Schizophrenia-associated behaviors and cognitive impairment were assessed using the open field test, pre-pulse inhibition, novel object recognition test, and Barnes maze test. The levels of mitochondrial proteins were measured using western blot analysis. Additionally, alterations in mitochondrial morphology, reduced hippocampal neuronal spine density, and impaired LTP in the hippocampus were observed. Nicotinamide (NAM) supplementation, administration of honokiol (HNK) (a SIRT3 activator), or overexpression of SIRT3 could inhibit cognitive deficits and cellular dysfunction. Conversely, administration of 3-TYP (a SIRT3 inhibitor) or knocking down SIRT3 expression in control rats led to deficits in behavioral and hippocampal neuronal phenotype. Our results suggest a causal role for the NAD+/SIRT3 axis in modulating cognitive behaviors via effects on hippocampal neuronal synaptic plasticity. The NAD+/SIRT3 axis could be a promising therapeutic target for addressing cognitive dysfunctions, such as those seen in schizophrenia.
Collapse
Affiliation(s)
- Keke Hao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Psychiatry, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fashuai Chen
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shilin Xu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ying Xiong
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rui Xu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Huan Huang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chang Shu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yisheng Lv
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Institute of Neurology and Psychiatry Research, Wuhan, China
| | - Huiling Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China.
- Department of Psychiatry, Hubei Provincial Clinical Research Center for Psychiatry, Wuhan, China.
| |
Collapse
|
23
|
Ma K, Niu J, Zeng L, Tian J, Zhang Y. SAM Alleviates Neuroinflammation by Regulating M1/M2 Polarization of Microglia Through α7nAChR/Nrf2/HO-1 Signaling Pathway. Neurochem Res 2025; 50:131. [PMID: 40156718 DOI: 10.1007/s11064-025-04373-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/29/2025] [Accepted: 03/10/2025] [Indexed: 04/01/2025]
Abstract
Microglia are the drivers of neuroinflammation. Microglia activation plays a critical role in the pathogenesis of aging. However, the mechanisms underlying microglial activation during aging are still not fully understood. Here, we investigated the role of S-adenosylmethionine (SAM) and its interplay with microglial activation in aging. In this study, we investigated the effect of SAM on BV2 cells treated with D-galactose (D-gal) and its molecular mechanism by Cell Counting Kit-8 (CCK8) assay, Senescence-associated β-Galactosidase (SA-β-gal) staining, western blot and immunofluorescence. We found that D-gal could induce microglia senescence. SAM intervention induced a significant decrease in the levels of inducible nitric oxide synthase (iNOS), tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) and increased arginase-1 (Arg1), α7 nicotinic acetylcholine receptor (α7nAChR), nuclear factor erythrocyte 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1) expression. Moreover, after administration of α7nAChR selective antagonist methyllycaconitine citrate (MLA), our results showed that SAM enhanced expression of α7nAChR, Nrf2 and HO-1, promoted the transformation of microglia from M1 to M2 subtype, and decreased the proinflammatory cytokines compared with MLA + D-gal group. These results suggest that SAM attenuates neuroinflammation by inhibiting microglia polarization through the α7nAChR/Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Kang Ma
- School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, Shandong Province, 266071, China
| | - Jiandong Niu
- The General Hospital of Ningxia Medical University, Yinchuan, China
| | - Liang Zeng
- The General Hospital of Ningxia Medical University, Yinchuan, China
| | - Jianying Tian
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia Hui Autonomous Region, 750004, China.
| | - Yawen Zhang
- School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, Shandong Province, 266071, China.
| |
Collapse
|
24
|
Martinez MX, Mahler SV. Potential roles for microglia in drug addiction: Adolescent neurodevelopment and beyond. J Neuroimmunol 2025; 404:578600. [PMID: 40199197 DOI: 10.1016/j.jneuroim.2025.578600] [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: 01/16/2025] [Revised: 03/14/2025] [Accepted: 03/27/2025] [Indexed: 04/10/2025]
Abstract
Adolescence is a sensitive period for development of addiction-relevant brain circuits, and it is also when people typically start experimenting with drugs. Unfortunately, such substance use may cause lasting impacts on the brain, and might increase vulnerability to later-life addictions. Microglia are the brain's immune cells, but their roles in shaping neural connectivity and synaptic plasticity, especially in developmental sensitive periods like adolescence, may also contribute to addiction-related phenomena. Here, we overview how drugs of abuse impact microglia, and propose that they may play poorly-understood, but important roles in addiction vulnerability and progression.
Collapse
Affiliation(s)
- Maricela X Martinez
- Department of Neurobiology and Behavior, University of California, 2221 McGaugh Hall, Irvine, CA 92697, USA.
| | - Stephen V Mahler
- Department of Neurobiology and Behavior, University of California, 2221 McGaugh Hall, Irvine, CA 92697, USA
| |
Collapse
|
25
|
Chen K, Li F, Zhang S, Chen Y, Ikezu TC, Li Z, Martens YA, Qiao W, Meneses A, Zhu Y, Xhafkollari G, Bu G, Zhao N. Enhancing TREM2 expression activates microglia and modestly mitigates tau pathology and neurodegeneration. J Neuroinflammation 2025; 22:93. [PMID: 40122810 PMCID: PMC11931752 DOI: 10.1186/s12974-025-03420-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
TREM2, a microglia-specific receptor, is strongly associated with Alzheimer's disease (AD) risk, mediating microglial responses to amyloid pathology critical to AD development. However, its role in tau pathology and neurodegeneration remains unclear. Using the PS19 tauopathy mouse model with inducible overexpression of human wild-type TREM2 (TREM2-WT) or the R47H variant (TREM2-R47H), we show that increasing TREM2-WT expression modestly reduces soluble phosphorylated tau levels and mildly preserves neuronal integrity. Single-cell RNA sequencing reveals that TREM2-WT robustly enhances microglial activation, characterized by a disease-associated microglia (DAM) signature. In contrast, TREM2-R47H overexpression exhibits a loss-of-function phenotype, with no significant impact on tau levels, neurodegeneration, or microglial activation. These findings highlight the role of TREM2 in modulating microglial activity and its influence on tau pathology and neurodegeneration, providing important insights for the future development of therapies targeting TREM2 or microglial pathways in AD or other tauopathies.
Collapse
Affiliation(s)
- Kai Chen
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Fuyao Li
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Shuwen Zhang
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Yixing Chen
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Tadafumi C Ikezu
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Zonghua Li
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Yuka A Martens
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Wenhui Qiao
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Axel Meneses
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- Clinical and Translational Science Graduate Program, Mayo Clinic, Jacksonville, FL, USA
| | - Yiyang Zhu
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Gisela Xhafkollari
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
- Clinical and Translational Science Graduate Program, Mayo Clinic, Jacksonville, FL, USA.
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
26
|
Mazzitelli M, Kiritoshi T, Presto P, Hurtado Z, Antenucci N, Ji G, Neugebauer V. BDNF Signaling and Pain Modulation. Cells 2025; 14:476. [PMID: 40214430 PMCID: PMC11987912 DOI: 10.3390/cells14070476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is an important neuromodulator of nervous system functions and plays a key role in neuronal growth and survival, neurotransmission, and synaptic plasticity. The effects of BDNF are mainly mediated by the activation of tropomyosin receptor kinase B (TrkB), expressed in both the peripheral and central nervous system. BDNF has been implicated in several neuropsychiatric conditions such as schizophrenia and anxio-depressive disorders, as well as in pain states. This review summarizes the evidence for a critical role of BDNF throughout the pain system and describes contrasting findings of its pro- and anti-nociceptive effects. Different cellular sources of BDNF, its influence on neuroimmune signaling in pain conditions, and its effects in different cell types and regions are described. These and endogenous BDNF levels, downstream signaling mechanisms, route of administration, and approaches to manipulate BDNF functions could explain the bidirectional effects in pain plasticity and pain modulation. Finally, current knowledge gaps concerning BDNF signaling in pain are discussed, including sex- and pathway-specific differences.
Collapse
Affiliation(s)
- Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Takaki Kiritoshi
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Peyton Presto
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Zachary Hurtado
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Nico Antenucci
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
27
|
Hsu CH, Hsu YY, Chang BM, Raffensperger K, Kadden M, Ton HT, Ette EA, Lin S, Brooks J, Burke MW, Lee YJ, Wang PC, Shoykhet M, Tu TW. StainAI: quantitative mapping of stained microglia and insights into brain-wide neuroinflammation and therapeutic effects in cardiac arrest. Commun Biol 2025; 8:462. [PMID: 40114030 PMCID: PMC11926354 DOI: 10.1038/s42003-025-07926-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Microglia, the brain's resident macrophages, participate in development and influence neuroinflammation, which is characteristic of multiple brain pathologies. Diverse insults cause microglia to alter their morphology from "resting" to "activated" shapes, which vary with stimulus type, brain location, and microenvironment. This morphologic diversity commonly restricts microglial analyses to specific regions and manual methods. We introduce StainAI, a deep learning tool that leverages 20x whole-slide immunohistochemistry images for rapid, high-throughput analysis of microglial morphology. StainAI maps microglia to a brain atlas, classifies their morphology, quantifies morphometric features, and computes an activation score for any region of interest. As a proof of principle, StainAI was applied to a rat model of pediatric asphyxial cardiac arrest, accurately classifying millions of microglia across multiple slices, surpassing current methods by orders of magnitude, and identifying both known and novel activation patterns. Extending its application to a non-human primate model of simian immunodeficiency virus infection further demonstrated its generalizability beyond rodent datasets, providing new insights into microglial responses across species. StainAI offers a scalable, high-throughput solution for microglial analysis from routine immunohistochemistry images, accelerating research in microglial biology and neuroinflammation.
Collapse
Affiliation(s)
- Chao-Hsiung Hsu
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
| | - Yi-Yu Hsu
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
- Miin Wu School of Computing, National Cheng Kung University, Tainan City, Taiwan
| | - Be-Ming Chang
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
| | - Katherine Raffensperger
- Center for Neuroscience Research, Children's National Research Institute, Washington, DC, USA
| | - Micah Kadden
- Center for Neuroscience Research, Children's National Research Institute, Washington, DC, USA
- Pediatric Critical Care Medicine, Children's National Hospital, Washington, DC, USA
| | - Hoai T Ton
- Center for Neuroscience Research, Children's National Research Institute, Washington, DC, USA
| | - Essiet-Adidiong Ette
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
| | - Stephen Lin
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
| | - Janiya Brooks
- Department of Physiology and Biophysics, Howard University, Washington, DC, USA
| | - Mark W Burke
- Department of Physiology and Biophysics, Howard University, Washington, DC, USA
| | - Yih-Jing Lee
- School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Paul C Wang
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
- Department of Physics, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Michael Shoykhet
- Center for Neuroscience Research, Children's National Research Institute, Washington, DC, USA
- Pediatric Critical Care Medicine, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Tsang-Wei Tu
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA.
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
28
|
Fan CY, McAllister BB, Stokes-Heck S, Harding EK, Pereira de Vasconcelos A, Mah LK, Lima LV, van den Hoogen NJ, Rosen SF, Ham B, Zhang Z, Liu H, Zemp FJ, Burkhard R, Geuking MB, Mahoney DJ, Zamponi GW, Mogil JS, Ousman SS, Trang T. Divergent sex-specific pannexin-1 mechanisms in microglia and T cells underlie neuropathic pain. Neuron 2025; 113:896-911.e9. [PMID: 39892387 DOI: 10.1016/j.neuron.2025.01.005] [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: 05/08/2024] [Revised: 11/25/2024] [Accepted: 01/08/2025] [Indexed: 02/03/2025]
Abstract
Chronic pain is a leading cause of disability, affecting more women than men. Different immune cells contribute to this sexual divergence, but the mechanisms, especially in females, are not well defined. We show that pannexin-1 (Panx1) channels on microglia and T cells differentially cause mechanical allodynia, a debilitating symptom of neuropathic pain. In male rodents, Panx1 drives vascular endothelial growth factor-A (VEGF-A) release from microglia. Cell-specific knockdown of microglial Panx1 or pharmacological blockade of the VEGF receptor attenuated allodynia in nerve-injured males. In females, nerve injury increased spinal CD8+ T cells and leptin levels. Leptin release from female-derived CD8+ T cells was Panx1 dependent, and intrathecal leptin-neutralizing antibody injection sex-specifically reversed allodynia. Adoptive transfer of female-derived CD8+ T cells caused robust allodynia, which was prevented by a leptin-neutralizing antibody or leptin small interfering RNA (siRNA) knockdown. Panx1-targeted approaches may alleviate neuropathic pain in both sexes, while T cell- and leptin-directed treatments could have sex-dependent benefits for women.
Collapse
Affiliation(s)
- Churmy Y Fan
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Brendan B McAllister
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Sierra Stokes-Heck
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Erika K Harding
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada; Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Aliny Pereira de Vasconcelos
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Laura K Mah
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute For Chronic Diseases, University of Calgary, Calgary, Canada
| | - Lucas V Lima
- Departments of Psychology and Anesthesia and Faculty of Dentistry, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Nynke J van den Hoogen
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Sarah F Rosen
- Departments of Psychology and Anesthesia and Faculty of Dentistry, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Boram Ham
- Departments of Psychology and Anesthesia and Faculty of Dentistry, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Zizhen Zhang
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Hongrui Liu
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada; Annie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada
| | - Franz J Zemp
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada; Annie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
| | - Regula Burkhard
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute For Chronic Diseases, University of Calgary, Calgary, Canada
| | - Markus B Geuking
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute For Chronic Diseases, University of Calgary, Calgary, Canada
| | - Douglas J Mahoney
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute For Chronic Diseases, University of Calgary, Calgary, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada; Annie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Jeffrey S Mogil
- Departments of Psychology and Anesthesia and Faculty of Dentistry, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Shalina S Ousman
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada; Department of Cell Biology & Anatomy, University of Calgary, Calgary, Canada
| | - Tuan Trang
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada.
| |
Collapse
|
29
|
d'Errico P, Früholz I, Meyer-Luehmann M, Vlachos A. Neuroprotective and plasticity promoting effects of repetitive transcranial magnetic stimulation (rTMS): A role for microglia. Brain Stimul 2025; 18:810-821. [PMID: 40118248 DOI: 10.1016/j.brs.2025.03.012] [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: 01/06/2025] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a non-invasive brain stimulation technique used to modulate neocortical excitability, with expanding applications in neurological and psychiatric disorders. However, the cellular and molecular mechanisms underlying its effects, particularly the role of microglia -the resident immune cells of the central nervous system- remain poorly understood. This review synthesizes recent findings on how different rTMS protocols influence microglial function under physiological conditions and in disease models. Emerging evidence indicates that rTMS modulates microglial activation, promoting neuroprotective and plasticity-enhancing processes not only in models of brain disorders, such as Alzheimer's and Parkinson's disease, but also in healthy neural circuits. While much of the current research has focused on the inflammatory profile of microglia, critical aspects such as activity-dependent synaptic remodeling, phagocytic activity, and process motility remain underexplored. Given the substantial heterogeneity of microglial responses across brain regions, age, and sex, as well as their differential roles in health and disease, a deeper understanding of their involvement in rTMS-induced plasticity is essential. Future studies should integrate selective microglial manipulation and advanced structural, functional, and molecular profiling techniques to clarify their causal involvement. Addressing these gaps will be pivotal in optimizing rTMS protocols and maximizing its therapeutic potential across a spectrum of neurological and neuropsychiatric conditions.
Collapse
Affiliation(s)
- Paolo d'Errico
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
| | - Iris Früholz
- Department of Neurology, Medical Center - University of Freiburg, Freiburg, Germany
| | | | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany; Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
30
|
Elser BA, Hing B, Eliasen S, Afrifa MA, Meurice N, Rimi F, Chimenti M, Schulz LC, Dailey ME, Gibson-Corley KN, Stevens HE. Maternal α-cypermethrin and permethrin exert differential effects on fetal growth, placental morphology, and fetal neurodevelopment in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.16.643434. [PMID: 40166261 PMCID: PMC11956951 DOI: 10.1101/2025.03.16.643434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Pyrethroid insecticides represent a broad class of chemicals used widely in agriculture and household applications. Human studies show mixed effects of maternal pyrethroid exposure on fetal growth and neurodevelopment. Assessment of shared pyrethroid metabolites as a biomarker for exposure obscures effects of specific chemicals within this broader class. To better characterize pyrethroid effects on fetal development, we investigated maternal exposure to permethrin, a type I pyrethroid, and α-cypermethrin, a type II pyrethroid, on fetal development in mice. Pregnant CD1 mice were exposed to permethrin (1.5, 15, or 50 mg/kg), α-cypermethrin (0.3, 3, or 10 mg/kg), or corn oil vehicle via oral gavage on gestational days (GD) 6-16. Effects on fetal growth, placental toxicity, and neurodevelopment were evaluated at GD 16. Cypermethrin, but not permethrin, significantly reduced fetal growth and altered placental layer morphology. Placental RNAseq analysis revealed downregulation of genes involved in extracellular matrix remodeling in response to α-cypermethrin. Both pyrethroids induced shifts in fetal dorsal forebrain microglia morphology from ramified to ameboid states; however, effects of α-cypermethrin were more pronounced. The α-cypermethrin transcriptome of fetal dorsal forebrain implicated altered glutamate receptor signaling, synaptogenesis, and c-AMP signaling. Coregulated gene modules in individual placenta and fetal dorsal forebrain pairs were correlated and overlapped in biological processes characterizing synapses, mitotic cell cycle, and chromatin organization, suggesting placenta-fetal brain shared mechanisms with α-cypermethrin exposure. In summary, maternal type II pyrethroid α-cypermethrin exposure but not type I pyrethroid permethrin significantly affected placental development, fetal growth, and neurodevelopment, and these effects were linked.
Collapse
Affiliation(s)
- Benjamin A Elser
- Interdisciplinary Graduate Program in Human Toxicology, Graduate College, The University of Iowa, Iowa City, Iowa, USA
- Department of Psychiatry, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Benjamin Hing
- Department of Psychiatry, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Samuel Eliasen
- Department of Psychiatry, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Malik A Afrifa
- Department of Psychiatry, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Naomi Meurice
- Department of Psychiatry, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Farzana Rimi
- Interdisciplinary Graduate Program in Human Toxicology, Graduate College, The University of Iowa, Iowa City, Iowa, USA
| | - Michael Chimenti
- Iowa Institute of Human Genetics, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Laura C Schulz
- Division of Reproductive and Perinatal Research, Department of Obstetrics, Gynecology, and Women's Health, University of Missouri, Columbia, Missouri, USA
| | - Michael E Dailey
- Department of Biology, University of Iowa College of Liberal Arts and Sciences, Iowa City, IA, USA
| | - Katherine N Gibson-Corley
- Division of Comparative Medicine, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hanna E Stevens
- Interdisciplinary Graduate Program in Human Toxicology, Graduate College, The University of Iowa, Iowa City, Iowa, USA
- Department of Psychiatry, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
31
|
Walter E, Angst G, Bollinger J, Truong L, Ware E, Wohleb ES, Fan Y, Wang C. Atg5 in microglia regulates sex-specific effects on postnatal neurogenesis in Alzheimer's disease. NPJ AGING 2025; 11:18. [PMID: 40091054 PMCID: PMC11911432 DOI: 10.1038/s41514-025-00209-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Female Alzheimer's disease (AD) patients display greater cognitive deficits and worse AD pathology as compared to male AD patients. In this study, we found that conditional knockout (cKO) of Atg5 in female microglia failed to obtain disease-associated microglia (DAM) gene signatures in familiar AD mouse model (5xFAD). Next, we analyzed the maintenance and neurogenesis of neural stem cells (NSCs) in the hippocampus and subventricular zone (SVZ) from 5xFAD mice with Atg5 cKO. Our data indicated that Atg5 cKO reduced the NSC number in hippocampus of female but not male 5xFAD mice. However, in the SVZ, Atg5 cKO only impaired NSCs in male 5xFAD mice. Interestingly, female 5xFAD;Fip200 cKO mice and 5xFAD;Atg14 cKO mice did not show NSC defects. These autophagy genes cKO 5xFAD mice exhibited a higher neurogenesis activity in their SVZ. Together, our data indicate a sex-specific role for microglial Atg5 in postnatal neurogenesis in AD mice.
Collapse
Affiliation(s)
- Ellen Walter
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Gabrielle Angst
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute and College of Medicine at The Ohio State University, Columbus, OH, USA
- Center for Cancer Metabolism, James Comprehensive Cancer Center at The Ohio State University, Columbus, OH, USA
| | - Justin Bollinger
- Department of Pharmacology & Systems Physiology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Linh Truong
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Elena Ware
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Yanbo Fan
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Chenran Wang
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA.
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute and College of Medicine at The Ohio State University, Columbus, OH, USA.
- Center for Cancer Metabolism, James Comprehensive Cancer Center at The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
32
|
Shlomi-Loubaton S, Nitzan K, Rivkin-Natan M, Sabbah S, Toledano R, Franko M, Bentulila Z, David D, Frenkel D, Doron R. Chronic stress leads to earlier cognitive decline in an Alzheimer's mouse model: The role of neuroinflammation and TrkB. Brain Behav Immun 2025; 127:303-314. [PMID: 40096896 DOI: 10.1016/j.bbi.2025.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 03/19/2025] Open
Abstract
While most Alzheimer's disease (AD) studies focus on the cognitive aspects of the disease, less focus is given to affective symptoms. In this study, we investigated the long-term consequences of exposure to chronic stress. 5xFAD AD model mice were exposed to unpredictable chronic mild stress, and cognitive and emotional aspects were examined at 3-time points (up to 4 months after exposure to stress). We found that exposure to chronic stress accelerates neuropathology outcomes in the 5xFAD mouse model in adulthood, accompanied by changes in the neurotrophic system. Specifically, we found that chronic stress accelerated the appearance of short-term spatial memory deficits in the 5xFAD mice and decreased tyrosine kinase B full receptor (TrkB.FL) expression levels. In vitro, we showed that corticosterone impairs the ability of microglia to uptake Aβ and reduces microglial activation. To conclude, our study may shed light on the mechanisms through which mild chronic stress might contribute to the onset and progression of Alzheimer's disease symptoms.
Collapse
Affiliation(s)
- Shir Shlomi-Loubaton
- Department of Neurobiology, School of Neurobiology Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Keren Nitzan
- Department of Education and Psychology, The Open University, Raanana 43107, Israel
| | - Maria Rivkin-Natan
- Department of Neurobiology, School of Neurobiology Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Salomé Sabbah
- Department of Neurobiology, School of Neurobiology Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Roni Toledano
- Department of Education and Psychology, The Open University, Raanana 43107, Israel
| | - Motty Franko
- Department of Education and Psychology, The Open University, Raanana 43107, Israel
| | - Ziv Bentulila
- Department of Education and Psychology, The Open University, Raanana 43107, Israel
| | - Dekel David
- Department of Education and Psychology, The Open University, Raanana 43107, Israel
| | - Dan Frenkel
- Department of Neurobiology, School of Neurobiology Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Ravid Doron
- Department of Education and Psychology, The Open University, Raanana 43107, Israel.
| |
Collapse
|
33
|
Barraco M, Kudova E, Bucolo C, Ciranna L, Sortino MA, Chisari M. Cholesterol metabolites modulate ionotropic P2X4 and P2X7 receptor current in microglia cells. Neuropharmacology 2025; 266:110294. [PMID: 39755203 DOI: 10.1016/j.neuropharm.2024.110294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025]
Abstract
The central nervous system is a well-known steroidogenic tissue producing, among others, cholesterol metabolites such as neuroactive steroids, oxysterols and steroid hormones. It is well known that these endogenous molecules affect several receptor classes, including ionotropic GABAergic and NMDA glutamatergic receptors in neurons. It has been shown that also ionotropic purinergic (P2X) receptors are cholesterol metabolites' targets. Among P2X receptors, purinergic P2X4 and P2X7 receptors are expressed in microglia, the innate immune cells involved in the brain inflammatory response. In this study, we explore the ionotropic purinergic receptors modulation by cholesterol metabolites in microglia. Patch-clamp experiments were performed in BV2 cells, a murine microglia cell line, to evaluate effects of cholesterol metabolites using micro- and nanomolar concentrations. About P2X4 receptor, we found that testosterone butyrate (20 μM and 200 nM) and allopregnanolone (10 μM and 100 nM) both potentiated its current, while neither 25-hydroxycholesterol (10 μM and 100 nM) nor 17β-estradiol (1 μM) showed any effects. On the other hand, P2X7 receptor current was potentiated by allopregnanolone (10 μM) and 25-hydroxycholesterol (10 μM and 100 nM). Taken together, our data show that modulation of either P2X4 and P2X7 current is affected differently by cholesterol metabolites, suggesting a structure-activity relationship among these players. Identifying the possible link between purinergic transmission, microglia and cholesterol metabolites will allow to define new targets for drug development to treat neuroinflammation.
Collapse
Affiliation(s)
- Michele Barraco
- Dept. of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Eva Kudova
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Claudio Bucolo
- Dept. of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Lucia Ciranna
- Dept. of Biomedical and Biotechnological Sciences, Physiology, University of Catania, Catania, Italy
| | - Maria Angela Sortino
- Dept. of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Mariangela Chisari
- Dept. of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy.
| |
Collapse
|
34
|
Belančić A, Janković T, Gkrinia EMM, Kristić I, Rajič Bumber J, Rački V, Pilipović K, Vitezić D, Mršić-Pelčić J. Glial Cells in Spinal Muscular Atrophy: Speculations on Non-Cell-Autonomous Mechanisms and Therapeutic Implications. Neurol Int 2025; 17:41. [PMID: 40137462 PMCID: PMC11944370 DOI: 10.3390/neurolint17030041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by homozygous deletions or mutations in the SMN1 gene, leading to progressive motor neuron degeneration. While SMA has been classically viewed as a motor neuron-autonomous disease, increasing evidence indicates a significant role of glial cells-astrocytes, microglia, oligodendrocytes, and Schwann cells-in the disease pathophysiology. Astrocytic dysfunction contributes to motor neuron vulnerability through impaired calcium homeostasis, disrupted synaptic integrity, and neurotrophic factor deficits. Microglia, through reactive gliosis and complement-mediated synaptic stripping, exacerbate neurodegeneration and neuroinflammation. Oligodendrocytes exhibit impaired differentiation and metabolic support, while Schwann cells display abnormalities in myelination, extracellular matrix composition, and neuromuscular junction maintenance, further compromising motor function. Dysregulation of pathways such as NF-κB, Notch, and JAK/STAT, alongside the upregulation of complement proteins and microRNAs, reinforces the non-cell-autonomous nature of SMA. Despite the advances in SMN-restorative therapies, they do not fully mitigate glial dysfunction. Targeting glial pathology, including modulation of reactive astrogliosis, microglial polarization, and myelination deficits, represents a critical avenue for therapeutic intervention. This review comprehensively examines the multifaceted roles of glial cells in SMA and highlights emerging glia-targeted strategies to enhance treatment efficacy and improve patient outcomes.
Collapse
Affiliation(s)
- Andrej Belančić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Tamara Janković
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | | | - Iva Kristić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Jelena Rajič Bumber
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Valentino Rački
- Department of Neurology, Clinical Hospital Centre Rijeka, Krešimirova 42, 51000 Rijeka, Croatia;
| | - Kristina Pilipović
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Dinko Vitezić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Jasenka Mršić-Pelčić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| |
Collapse
|
35
|
Rodriguez-Baena FJ, Marquez-Galera A, Ballesteros-Martinez P, Castillo A, Diaz E, Moreno-Bueno G, Lopez-Atalaya JP, Sanchez-Laorden B. Microglial reprogramming enhances antitumor immunity and immunotherapy response in melanoma brain metastases. Cancer Cell 2025; 43:413-427.e9. [PMID: 39919736 DOI: 10.1016/j.ccell.2025.01.008] [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: 04/11/2024] [Revised: 11/04/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025]
Abstract
Melanoma is one of the tumor types with the highest risk of brain metastasis. However, the biology of melanoma brain metastasis and the role of the brain immune microenvironment in treatment responses are not yet fully understood. Using preclinical models and single-cell transcriptomics, we have identified a mechanism that enhances antitumor immunity in melanoma brain metastasis. We show that activation of the Rela/Nuclear Factor κB (NF-κB) pathway in microglia promotes melanoma brain metastasis. Targeting this pathway elicits microglia reprogramming toward a proinflammatory phenotype, which enhances antitumor immunity and reduces brain metastatic burden. Furthermore, we found that proinflammatory microglial markers in melanoma brain metastasis are associated with improved responses to immune checkpoint inhibitors in patients and targeting Rela/NF-κB pathway in mice improves responses to these therapies in the brain, suggesting a strategy to enhance antitumor immunity and responses to immune checkpoint inhibitors in patients with melanoma brain metastasis.
Collapse
Affiliation(s)
| | | | | | - Alba Castillo
- Instituto de Neurociencias (CSIC-UMH), San Juan de Alicante, Spain
| | - Eva Diaz
- MD Anderson Cancer Center International Foundation, Madrid, Spain
| | - Gema Moreno-Bueno
- MD Anderson Cancer Center International Foundation, Madrid, Spain; Instituto de Investigaciones Biomédicas "Sols-Morreale" CSIC-UAM, Madrid, Spain; CIBERONC Centro de Investigación Biomédica en Red de Cancer, ISCIII, Madrid, Spain; Translational Cancer Research Group, Area 3 Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | | | | |
Collapse
|
36
|
Schoot Uiterkamp FE, Maes ME, Alamalhoda MA, Firoozi A, Colombo G, Siegert S. Optic Nerve Crush Does Not Induce Retinal Ganglion Cell Loss in the Contralateral Eye. Invest Ophthalmol Vis Sci 2025; 66:49. [PMID: 40126507 PMCID: PMC11951053 DOI: 10.1167/iovs.66.3.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 02/16/2025] [Indexed: 03/25/2025] Open
Abstract
Purpose Optic nerve crush (ONC) is a model for studying optic nerve trauma. Unilateral ONC induces massive retinal ganglion cell (RGC) degeneration in the affected eye, leading to vision loss within a month. A common assumption has been that the non-injured contralateral eye is unaffected due to the minimal retino-retinal projections of the RGCs at the chiasm. Yet, recently, microglia, the brain-resident macrophages, have shown a responsive phenotype in the contralateral eye after ONC. Whether RGC loss accompanies this phenotype is still controversial. Methods Using the available RGCode algorithm and developing our own RGC-Quant deep-learning-based tool, we quantify RGC's total number and density across the entire retina after ONC. Results We confirm a short-term microglia response in the contralateral eye after ONC, but this did not affect the microglia number. Furthermore, we cannot confirm the previously reported RGC loss between naïve and contralateral retinas 5 weeks after ONC induction across the commonly used Cx3cr1creERT2 and C57BL6/J mouse models. Neither sex nor the direct comparison of the RGC markers Brn3a and RBPMS, with Brn3a co-labeling, on average, 89% of the RBPMS+-cells, explained this discrepancy, suggesting that the early microglia-responsive phenotype does not have immediate consequences on the RGC number. Conclusions Our results corroborate that unilateral optic nerve injury elicits a microglial response in the uninjured contralateral eye but without RGC loss. Therefore, the contralateral eye should be treated separately and not as an ONC control.
Collapse
Affiliation(s)
| | - Margaret E. Maes
- Institute of Science and Technology Austria (ISTA), Am Campus 1, Klosterneuburg, Austria
| | | | - Arsalan Firoozi
- Institute of Science and Technology Austria (ISTA), Am Campus 1, Klosterneuburg, Austria
| | - Gloria Colombo
- Institute of Science and Technology Austria (ISTA), Am Campus 1, Klosterneuburg, Austria
| | - Sandra Siegert
- Institute of Science and Technology Austria (ISTA), Am Campus 1, Klosterneuburg, Austria
| |
Collapse
|
37
|
Wang J, Gao S, Fu S, Li Y, Su L, Li X, Wu G, Jiang J, Zhao Z, Yang C, Wang X, Cui K, Sun X, Qi X, Wang C, Sun H, Shao S, Tian Y, Gong T, Luo J, Zheng J, Cui S, Liao F, Liu F, Wang D, Wong CCL, Yi M, Wan Y. Irisin reprograms microglia through activation of STAT6 and prevents cognitive dysfunction after surgery in mice. Brain Behav Immun 2025; 125:68-91. [PMID: 39701329 DOI: 10.1016/j.bbi.2024.12.019] [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: 05/19/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024] Open
Abstract
Postoperative cognitive dysfunction (POCD) is common in the aged population and associated with poor clinical outcomes. Irisin, an endogenous molecule that mediates the beneficial effects of exercise, has shown neuroprotective potential in several models of neurological diseases. Here we show that preoperative serum level of irisin is reduced in dementia patients over the age of 70. Comprehensive proteomics analysis reveals that deletion of irisin affects the nervous and immune systems, and reduces the expression of complement proteins. Systemically administered irisin penetrates the blood-brain barrier in mice, targets the microglial integrin αVβ5 receptor, activates signal transducer and activator of transcription 6 (STAT6), induces microglia reprogramming to the M2 phenotype, and improves immune microenvironment in LPS-induced neuroinflammatory mice. Finally, prophylactic administration of irisin prevents POCD-like behavior, particularly early cognitive dysfunction. Our findings provide new insights into the direct regulation of the immune microenvironment by irisin, and reveal that recombinant irisin holds great promise as a novel therapy for preventing POCD and other neuroinflammatory disorders. SUMMARY: Our findings reveal molecular and cellular mechanisms of irisin on neuroinflammation, and show that prophylactic administration of irisin prevents POCD-like behavior, particularly early cognitive dysfunction.
Collapse
Affiliation(s)
- Jiaxin Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Shuaixin Gao
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China; Human Nutrition Program, Department of Human Sciences & James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Su Fu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Yawei Li
- Department of Anesthesiology, Peking University First Hospital, Beijing 10034, China
| | - Li Su
- Peking University Medical and Health Analysis Center, Peking University, Beijing 10034, China
| | - Xiaoman Li
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Guanghao Wu
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Jiankuo Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Zifang Zhao
- Department of Pain Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Chaojuan Yang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Xiaoyi Wang
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Kun Cui
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China; Beijing Life Science Academy, Beijing 102209, China
| | - Xiaoyan Sun
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Xuetao Qi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Cheng Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China; Changping Laboratory, Beijing 102206, China
| | - Haojie Sun
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China; UCL School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Shan Shao
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Yue Tian
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Tingting Gong
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Jianyuan Luo
- Department of Medical Genetics, Center for Medical Genetics, Peking University Health Science Center, Beijing 100191, China
| | - Jie Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Shuang Cui
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Feifei Liao
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Fengyu Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China.
| | - Dongxin Wang
- Department of Anesthesiology, Peking University First Hospital, Beijing 10034, China.
| | - Catherine C L Wong
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China.
| | - Ming Yi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China; Medical Innovation Center (Taizhou) of Peking University, Taizhou 225316, China.
| | - You Wan
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China; Medical Innovation Center (Taizhou) of Peking University, Taizhou 225316, China.
| |
Collapse
|
38
|
Bai J, Yue Y, Zhang Z, Wang K, Jin Y, Wang J, Zou L. Effects of vagus nerve stimulation on microglia inhibit heroin-induced conditional place preference. Brain Behav Immun 2025; 127:45-56. [PMID: 40032204 DOI: 10.1016/j.bbi.2025.02.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/12/2025] [Accepted: 02/22/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Heroin addiction remains a significant global health challenge with limited effective treatments. Vagus nerve stimulation (VNS) has shown promise in treating various neurological disorders, but its potential in addiction treatment is unexplored. OBJECTIVE To investigate the effects of VNS on heroin-induced conditioned place preference (CPP) and elucidate the underlying neurobiological mechanisms. METHODS We employed bilateral subphrenic vagotomy and VNS models in mice. Heroin-induced CPP was assessed following transcervical (nVNS) or transcutaneous auricular (taVNS) stimulation. Microglial activation in the nucleus accumbens (NAc) was evaluated using immunofluorescence and ELISA. The role of α7 nicotinic acetylcholine receptors (α7nAChRs) was investigated using the antagonist methyllycaconitine. RESULTS Both nVNS and taVNS significantly attenuated heroin-induced CPP. VNS reversed heroin-induced microglial activation in the NAc, reducing pro-inflammatory markers and cytokines while increasing anti-inflammatory markers. These effects were mediated by α7nAChRs, as antagonist administration abolished VNS efficacy. Notably, subphrenic vagotomy did not affect VNS efficacy, suggesting a primarily central mechanism of action. CONCLUSION VNS inhibits heroin-induced CPP, likely through modulation of NAc microglia via α7nAChRs. taVNS, while less effective than nVNS, offers a promising non-invasive approach to addiction treatment. These findings provide a rationale for further clinical investigation of VNS, particularly taVNS, as an adjunct therapy for heroin addiction. BRIEF ABSTRACT Vagus nerve stimulation (VNS) attenuates heroin-induced conditioned place preference in mice by modulating microglial activation in the nucleus accumbens via α7 nicotinic acetylcholine receptors. Both invasive and non-invasive VNS show efficacy, with the latter offering potential as a novel addiction treatment approach. These findings warrant further investigation of VNS in clinical settings for heroin addiction management.
Collapse
Affiliation(s)
- Jianhua Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yingbiao Yue
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Zunyue Zhang
- Yunnan Province Drug Dependence Treatment Technology Innovation Center, Yunnan University, Kunming, Yunnan, China
| | - Kunhua Wang
- Yunnan Province Drug Dependence Treatment Technology Innovation Center, Yunnan University, Kunming, Yunnan, China
| | - Yun Jin
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Junfeng Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Lei Zou
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China.
| |
Collapse
|
39
|
Zhao S, Wang L, Kleidonas D, Qi F, Liang Y, Zheng J, Umpierre AD, Wu LJ. Chemogenetic activation of microglial Gi signaling decreases microglial surveillance and impairs neuronal synchronization. SCIENCE ADVANCES 2025; 11:eado7829. [PMID: 40020068 PMCID: PMC11870068 DOI: 10.1126/sciadv.ado7829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 01/29/2025] [Indexed: 03/03/2025]
Abstract
Microglia actively survey the brain and dynamically interact with neurons to maintain brain homeostasis. Microglial Gi protein-coupled receptors (Gi-GPCRs) play a critical role in microglia-neuron communications. However, the impact of temporally activating microglial Gi signaling on microglial dynamics and neuronal activity in the homeostatic brain remains largely unknown. In this study, we used Gi-based designer receptors exclusively activated by designer drugs (Gi-DREADD) to selectively and temporally modulate microglial Gi signaling pathway. By integrating this chemogenetic approach with in vivo two-photon imaging, we observed that exogenous activation of microglial Gi signaling transiently inhibited microglial process dynamics, reduced neuronal activity, and impaired neuronal synchronization. These altered neuronal functions were associated with a decrease in interactions between microglia and neuron somata. Together, this study demonstrates that acute, exogenous activation of microglial Gi signaling regulates neuronal circuit function, offering a potential pharmacological target for the neuromodulation through microglia.
Collapse
Affiliation(s)
- Shunyi Zhao
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
- Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Lingxiao Wang
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Dimitrios Kleidonas
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Fangfang Qi
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yue Liang
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | | | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
40
|
Mendoza-Romero HN, Biddinger JE, Bedenbaugh MN, Simerly RB. Microglia are Required for Developmental Specification of AgRP Innervation in the Hypothalamus of Offspring Exposed to Maternal High-Fat Diet During Lactation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.12.607566. [PMID: 39185162 PMCID: PMC11343114 DOI: 10.1101/2024.08.12.607566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Agouti-related peptide (AgRP) neurons in the arcuate nucleus of the hypothalamus respond to multiple metabolic signals and distribute neuroendocrine information to other brain regions such as the paraventricular hypothalamic nucleus (PVH), which plays a central role in metabolic homeostasis. Neural projections from AgRP neurons to the PVH form during the postnatal lactational period in mice and these projections are reduced in offspring of dams that consumed a high-fat diet (HFD) during lactation (MHFD-L). Here we used immunohistochemistry to visualize microglial morphology in MHFD-L offspring and identified changes that were regionally localized to the PVH and appeared temporally restricted to the period when AgRP neurons innervate this region. In addition, axon labeling experiments revealed that microglia engulf AgRP terminals in the PVH, and that the density of AgRP innervation to the PVH in MHFD-L offspring may be dependent on microglia, because microglial depletion blocked the decrease in PVH AgRP innervation observed in MHFD-L offspring, as well as prevented the increased body weight exhibited at weaning. Together, these findings suggest that microglia are activated by exposure to MHFD-L and interact directly with AgRP axons during postnatal development to permanently alter innervation of the PVH, with implications for developmental programming of metabolic phenotype.
Collapse
Affiliation(s)
| | - Jessica E. Biddinger
- Dept of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Michelle N. Bedenbaugh
- Dept of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Richard B. Simerly
- Dept of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
41
|
Flury A, Aljayousi L, Park HJ, Khakpour M, Mechler J, Aziz S, McGrath JD, Deme P, Sandberg C, González Ibáñez F, Braniff O, Ngo T, Smith S, Velez M, Ramirez DM, Avnon-Klein D, Murray JW, Liu J, Parent M, Mingote S, Haughey NJ, Werneburg S, Tremblay MÈ, Ayata P. A neurodegenerative cellular stress response linked to dark microglia and toxic lipid secretion. Neuron 2025; 113:554-571.e14. [PMID: 39719704 DOI: 10.1016/j.neuron.2024.11.018] [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: 04/11/2024] [Revised: 10/22/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024]
Abstract
The brain's primary immune cells, microglia, are a leading causal cell type in Alzheimer's disease (AD). Yet, the mechanisms by which microglia can drive neurodegeneration remain unresolved. Here, we discover that a conserved stress signaling pathway, the integrated stress response (ISR), characterizes a microglia subset with neurodegenerative outcomes. Autonomous activation of ISR in microglia is sufficient to induce early features of the ultrastructurally distinct "dark microglia" linked to pathological synapse loss. In AD models, microglial ISR activation exacerbates neurodegenerative pathologies and synapse loss while its inhibition ameliorates them. Mechanistically, we present evidence that ISR activation promotes the secretion of toxic lipids by microglia, impairing neuron homeostasis and survival in vitro. Accordingly, pharmacological inhibition of ISR or lipid synthesis mitigates synapse loss in AD models. Our results demonstrate that microglial ISR activation represents a neurodegenerative phenotype, which may be sustained, at least in part, by the secretion of toxic lipids.
Collapse
Affiliation(s)
- Anna Flury
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Leen Aljayousi
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Hye-Jin Park
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | | | - Jack Mechler
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biochemistry, CUNY Graduate Center, New York, NY 10016, USA
| | - Siaresh Aziz
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Jackson D McGrath
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Michigan Medicine, Ann Arbor, MI 48105, USA
| | - Pragney Deme
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Colby Sandberg
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C4, Canada
| | | | - Olivia Braniff
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C4, Canada
| | - Thi Ngo
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Simira Smith
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Matthew Velez
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Denice Moran Ramirez
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Dvir Avnon-Klein
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - John W Murray
- Columbia Center for Human Development, Center for Stem Cell Therapies, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jia Liu
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Martin Parent
- CERVO Brain Research Center, Québec City, QC G1E 1T2, Canada
| | - Susana Mingote
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Norman J Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sebastian Werneburg
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Michigan Medicine, Ann Arbor, MI 48105, USA; Michigan Neuroscience Institute, Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C4, Canada; Department of Molecular Medicine, Université Laval, Québec City, QC G1V 0A6, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, QC H3A 2B4, Canada; Canada Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 2A1, Canada; Centre for Advanced Materials and Related Technology and Institute on Aging and Lifelong Health, University of Victoria, Victoria, BC V8N 5M8, Canada
| | - Pinar Ayata
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA; Graduate Program in Biochemistry, CUNY Graduate Center, New York, NY 10016, USA.
| |
Collapse
|
42
|
Cao ZL, Zhu LX, Wang HM, Zhu LJ. Microglial Regulation of Neural Networks in Neuropsychiatric Disorders. Neuroscientist 2025:10738584251316558. [PMID: 39932233 DOI: 10.1177/10738584251316558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2025]
Abstract
Microglia serve as vital innate immune cells in the central nervous system, playing crucial roles in the generation and development of brain neurons, as well as mediating a series of immune and inflammatory responses. The morphologic transitions of microglia are closely linked to their function. With the advent of single-cell sequencing technology, the diversity of microglial subtypes is increasingly recognized. The intricate interactions between microglia and neuronal networks have significant implications for psychiatric disorders and neurodegenerative diseases. A deeper investigation of microglia in neurologic diseases such as Alzheimer disease, depression, and epilepsy can provide valuable insights in understanding the pathogenesis of diseases and exploring novel therapeutic strategies, thereby addressing issues related to central nervous system disorders.
Collapse
Affiliation(s)
- Zi-Lin Cao
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| | - Li-Xia Zhu
- Patent Examination Cooperation (JIANGSU) Center of the Patent Office, China National Intellectual Property Administration (CNIPA), Suzhou, China
| | - Hong-Mei Wang
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| | - Li-Juan Zhu
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
43
|
Hafeez MT, Gao H, Ju F, Qi F, Li T, Zhang S. Transcriptomic Analysis Divulges Differential Expressions of Microglial Genes After Microglial Repopulation in Mice. Int J Mol Sci 2025; 26:1494. [PMID: 40003960 PMCID: PMC11855859 DOI: 10.3390/ijms26041494] [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: 12/12/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Microglia are key immune cells in the central nervous system (CNS) and maintain hemostasis in physiological conditions. Microglial depletion leads to rapid repopulation, but the gene expression and signaling pathways related to repopulation remain unclear. Here, we used RNA sequencing (RNA-Seq) analysis to profile the transcriptome of microglia-depleted tissue by taking advantage of a conditional genetic microglial depletion model (CX3CR1CreER/+ system). Differential gene expression (DGE) sequencing analysis showed that 1226 genes were differentially up- and downregulated in both groups compared to control. Our data demonstrated that many microglial genes were highly regulated on day 3 after depletion but the numbers of differentially expressed genes were reduced by day 7. Gene ontology (GO) analysis categorized these differentially expressed genes on day 3 and day 7 to the specific biological processes, such as cell proliferation, cell activation, and cytokine and chemokine production. DGE analysis indicated that specific genes related to proliferation were regulated after depletion. Consistent with the changes in transcriptome, the histological analysis of transgenic mice revealed that the microglia after depletion undergo proliferation and activation from day 3 to day 7. Collectively, these results suggest that transcriptomic changes in microglial genes during depletion have a profound implication for the renewal and activation of microglia and may help to understand the regulatory mechanism of microglial activation in disease conditions.
Collapse
Affiliation(s)
| | | | | | | | | | - Shengxiang Zhang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
44
|
Chen J, Bai Y, He X, Xiao W, Chen L, Wong YK, Wang C, Gao P, Cheng G, Xu L, Yang C, Liao F, Han G, Sun J, Xu C, Wang J. The spatiotemporal transcriptional profiling of murine brain during cerebral malaria progression and after artemisinin treatment. Nat Commun 2025; 16:1540. [PMID: 39934099 DOI: 10.1038/s41467-024-52223-7] [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: 09/04/2023] [Accepted: 08/28/2024] [Indexed: 02/13/2025] Open
Abstract
Cerebral malaria (CM) is a severe encephalopathy caused by Plasmodium parasite infection, resulting in thousands of annual deaths and neuro-cognitive sequelae even after anti-malarial drugs treatment. Despite efforts to dissect the mechanism, the cellular transcriptomic reprogramming within the spatial context remains elusive. Here, we constructed single-cell and spatial transcriptome atlases of experimental CM (ECM) male murine brain tissues with or without artesunate (ART) treatment. We identified activated inflammatory endothelial cells during ECM, characterized by a disrupted blood-brain barrier, increased antigen presentation, and leukocyte adhesion. We also observed that inflammatory microglia enhance antigen presentation pathway such as MHC-I to CD8+ cytotoxic T cells. The latter underwent an inflammatory state transition with up-regulated cytokine expression and cytotoxic activity. Multi-omics analysis revealed that the activated interferon-gamma response of injured neurons during ECM and persisted after ART treatment. Overall, our research provides valuable resources for understanding malaria parasite-host interaction mechanisms and adjuvant therapy development.
Collapse
Affiliation(s)
- Jiayun Chen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China
| | - Yunmeng Bai
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Xueling He
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wei Xiao
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China
- Department of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Lina Chen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yin Kwan Wong
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Chen Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Peng Gao
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, Henan, China
| | - Guangqing Cheng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Liting Xu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chuanbin Yang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Fulong Liao
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Guang Han
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, Henan, China
| | - Jichao Sun
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| | - Chengchao Xu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| | - Jigang Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China.
- Department of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, Henan, China.
| |
Collapse
|
45
|
Zhao Q, Jaiswal J, Chang E, Shahsavari A, Zhang Y, Yu V, Zheng R, Liu F. The ontogeny of mouse salivary gland macrophages is distinct between genders. RESEARCH SQUARE 2025:rs.3.rs-5903499. [PMID: 39975913 PMCID: PMC11838738 DOI: 10.21203/rs.3.rs-5903499/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Sexual dimorphism is found in gene expression and polarization of macrophages in mammals but remains unclear in the ontogeny of tissue-resident macrophages. Salivary glands show remarkable gender differences and macrophages here are essential for the maintenance of immune quiescence and tissue repair after damages. Therefore, we compared origins of macrophages in salivary glands between male and female mice using conditional Cx3cr1 and Ccr2 lineage tracing approaches. We found most salivary gland macrophages in adult males are locally maintained and derived from yolk sac progenitors or postnatal monocytes, whereas these cells in adult females are mostly short-lived and continuously replenished by monocytes. Female salivary glands also contain more monocytes and polarized macrophages. Cx3cr1 knockout made the macrophage ontogeny in adult male salivary glands like females. These gender differences appear related to female-dominant salivary gland phenotypes in a common autoimmune disease, Sjogren's syndrome, and need be considered in developing macrophage-targeting therapies of dry mouth caused by autoimmunity or radiation.
Collapse
Affiliation(s)
- Qingguo Zhao
- Cell Biology and Genetics Department, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Juhi Jaiswal
- Cell Biology and Genetics Department, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Ethan Chang
- Cell Biology and Genetics Department, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Arash Shahsavari
- Cell Biology and Genetics Department, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Yu Zhang
- Cell Biology and Genetics Department, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Victor Yu
- Cell Biology and Genetics Department, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Ronald Zheng
- Cell Biology and Genetics Department, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Fei Liu
- Cell Biology and Genetics Department, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| |
Collapse
|
46
|
Soylu KO, Yemisci M, Karatas H. The link between spreading depolarization and innate immunity in the central nervous system. J Headache Pain 2025; 26:25. [PMID: 39901107 PMCID: PMC11792447 DOI: 10.1186/s10194-024-01938-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/12/2024] [Indexed: 02/05/2025] Open
Abstract
Spreading depolarization (SD) is a complex event that induces significant cellular stress in the central nervous system, leading to a robust inflammatory response without causing cell death in healthy tissues which may be called as neuro-parainflammation. Research has established a clear link between SD and the activation of pro-inflammatory pathways, particularly through the release of cytokines like interleukin-1β and tumor necrosis factor-α, and the involvement of inflammatory mediators such as cyclooxygenase-2 and high mobility group box 1 (HMGB1). Mechanistically, the opening of pannexin-1 (Panx1) channels and the activation of the (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome play critical roles in this process, facilitating the release of inflammatory signals that can exacerbate conditions like migraine. Furthermore, the interplay between neurons and glial cells, particularly astrocytes and microglia, underscores the intricate nature of neuroinflammation triggered by SD. Importantly, these findings indicate that these inflammatory processes may also have systemic implications, affecting immune responses beyond the central nervous system. Overall, this body of work highlights the need for further exploration of the mechanisms underlying SD-induced inflammation and potential therapeutic targets to mitigate neuroinflammatory disorders. Inflammation extends beyond the central nervous system to peripheral structures, including the meninges and trigeminovascular system, which are critical for headache initiation. Genetic factors, particularly familial hemiplegic migraine (FHM), exacerbate neuroinflammatory responses to SD, leading to increased susceptibility and prolonged headache behaviors. Collectively, these findings underscore the complex cellular interactions and innate inflammatory processes underlying SD and their relevance to migraine mechanisms, suggesting potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Kadir Oguzhan Soylu
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Türkiye
| | - Muge Yemisci
- Institute of Neurological Sciences and Psychiatry, Faculty of Medicine, Department of Neurology, Hacettepe University, Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Türkiye
| | - Hulya Karatas
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Türkiye.
| |
Collapse
|
47
|
Xiong M, Xia D, Yu H, Meng L, Zhang X, Chen J, Tian Y, Yuan X, Niu X, Nie S, Zhang Z, Liu C, Chen Q, Ye K, Zhang Z. Microglia Process α-Synuclein Fibrils and Enhance their Pathogenicity in a TREM2-Dependent Manner. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413451. [PMID: 39665233 PMCID: PMC11831461 DOI: 10.1002/advs.202413451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/17/2024] [Indexed: 12/13/2024]
Abstract
Parkinson's disease (PD) is characterized by the deposition of misfolded α-synuclein (α-syn) in the brain. Converging evidence indicates that the intracellular transmission and subsequent templated amplification of α-syn are involved in the onset and progression of PD. However, the molecular mechanisms underlying the cell-to-cell transmission of pathological α-syn remain poorly understood. Microglia is highly activated in the brains of PD patients. Here, it is shown that depletion of microglia slows the spread of pathological α-syn pathology in mice injected with α-syn fibrils. Microglia phagocytose α-syn fibrils and transform them into more toxic species. The phagocytosis of α-syn fibrils by microglia is partially mediated by triggering a receptor expressed on myeloid cells 2 (TREM2), a transmembrane protein expressed on the surface of microglia. The endocytosed α-syn fibrils are then cleaved by the lysosomal proteinase asparagine endopeptidase (AEP) to generate truncated α-syn 1-103 fibrils with enhanced seeding activity. Knockout of TREM2 and AEP impedes the endocytosis and cleavage of α-syn fibrils, respectively. The results demonstrate that TREM2-mediated phagocytosis of α-syn fibrils by microglia and subsequent AEP-mediated cleavage of α-syn fibrils contribute to the spread of α-syn in the brain. Blocking either of these two steps attenuates the progression of α-syn pathology.
Collapse
Affiliation(s)
- Min Xiong
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Danhao Xia
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Honglu Yu
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Lanxia Meng
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Xingyu Zhang
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Jiehui Chen
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Ye Tian
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Xin Yuan
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Xuan Niu
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Shuke Nie
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Zhaohui Zhang
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Chaoyang Liu
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Qiang Chen
- Frontier Science Center for Immunology and MetabolismMedical Research InstituteWuhan UniversityWuhan430071China
| | - Keqiang Ye
- Faculty of Life and Health SciencesShenzhen Institute of Advanced TechnologyShenzhen518035China
| | - Zhentao Zhang
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhan430060China
- TaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430000China
| |
Collapse
|
48
|
Duffy AS, Eyo UB. Microglia and Astrocytes in Postnatal Neural Circuit Formation. Glia 2025; 73:232-250. [PMID: 39568399 DOI: 10.1002/glia.24650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
Over the past two decades, microglia and astrocytes have emerged as critical mediators of neural circuit formation. Particularly during the postnatal period, both glial subtypes play essential roles in orchestrating nervous system development through communication with neurons. These functions include regulating synapse elimination, modulating neuronal density and activity, mediating synaptogenesis, facilitating axon guidance and organization, and actively promoting neuronal survival. Despite the vital roles of both microglia and astrocytes in ensuring homeostatic brain development, the extent to which the postnatal functions of these cells are regulated by sex and the manner in which these glial cells communicate with one another to coordinate nervous system development remain less well understood. Here, we review the critical functions of both microglia and astrocytes independently and synergistically in mediating neural circuit formation, focusing our exploration on the postnatal period from birth to early adulthood.
Collapse
Affiliation(s)
- Abigayle S Duffy
- Brain Immunology and Glia Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA
| | - Ukpong B Eyo
- Brain Immunology and Glia Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
49
|
Peña-Ortega F. Microglial modulation of neuronal network function and plasticity. J Neurophysiol 2025; 133:661-680. [PMID: 39819084 DOI: 10.1152/jn.00458.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/08/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS), which have been classically viewed as involved in CNS responses to damage and tissue repair. However, microglia are constantly sensing neuronal network activity and changes in the CNS milieu, establishing complex state-dependent microglia-neuron interactions that impact their functions. By doing so, microglia perform a wide range of physiological roles, including brain homeostasis maintenance, control of neural connectivity, network function modulation, as well as functional and morphological plasticity regulation in health and disease. Here, the author reviews recent evidence of the modulations induced by microglia, a highly heterogeneous cell type, on synaptic and intrinsic neuronal properties, and on neuronal network patterns during perinatal development and adulthood. The reviewed evidence clearly indicates that microglia are important, if not essential, for brain function and plasticity in both health and disease.
Collapse
Affiliation(s)
- Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| |
Collapse
|
50
|
Lukkahatai N, Ong IL, Benjasirisan C, Saligan LN. Brain-Derived Neurotrophic Factor (BDNF) as a Marker of Physical Exercise or Activity Effectiveness in Fatigue, Pain, Depression, and Sleep Disturbances: A Scoping Review. Biomedicines 2025; 13:332. [PMID: 40002745 PMCID: PMC11853410 DOI: 10.3390/biomedicines13020332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Brain-derived neurotrophic factor (BDNF) has been investigated as a potential mechanistic marker or therapeutic target to manage symptoms such as fatigue, pain, depression, and sleep disturbances. However, the variability in BDNF response to exercise or physical activity (exercise/PA) and its clinical relevance in symptom management remains unclear. This scoping review assesses existing studies exploring the relationships between exercise/PA, symptoms, and BDNF levels, specifically focusing on fatigue, pain, depression, and sleep disturbances in adults. Methods: Relevant studies indexed in PubMed and CINAHL were identified. Using systematic review software, two reviewers independently screened and evaluated full texts, based on the following criteria: human studies reporting BDNF levels in adults, using exercise/PA interventions, assessing symptoms (pain, fatigue, depression, and/or sleep disturbance) as outcomes, and published in English. Results: Of 950 records, 35 records met the inclusion criteria. While exercise/PA is broadly supported for managing symptoms, 74.3% (n = 26) of studies reported increased BDNF levels, and only 40% (n = 14) showed significant increases following exercise/PA. Only 14% (n = 5) of studies demonstrated a significant relationship between changes in BDNF and symptoms. No significant differences in BDNF levels and symptoms were observed between different types of exercise (e.g., aerobic vs. strength vs. flexibility/stretching) and PA. Conclusions: The current literature provides insufficient evidence to confirm BDNF as a marker for exercise/PA effectiveness on symptoms. Further clinical investigations are needed to validate its potential as a therapeutic target.
Collapse
Affiliation(s)
- Nada Lukkahatai
- School of Nursing, Johns Hopkins University, Baltimore, MD 21205, USA;
| | - Irvin L. Ong
- Research Development and Innovation Center, Our Lady of Fatima University, Valenzuela City 1440, Philippines;
- Department of Nursing and Health Sciences, Elmhurst University, Elmhurst, IL 60126, USA
| | | | - Leorey N. Saligan
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD 20892, USA;
| |
Collapse
|